http://researchspace.auckland.ac.nz ResearchSpace@Auckland Copyright Statement The digital copy of this thesis is protected by the Copyright Act 1994 (New Zealand). This thesis may be consulted by you, provided you comply with the provisions of the Act and the following conditions of use: • • • Any use you make of these documents or images must be for research or private study purposes only, and you may not make them available to any other person. Authors control the copyright of their thesis. You will recognise the author's right to be identified as the author of this thesis, and due acknowledgement will be made to the author where appropriate. You will obtain the author's permission before publishing any material from their thesis. To request permissions please use the Feedback form on our webpage. http://researchspace.auckland.ac.nz/feedback General copyright and disclaimer In addition to the above conditions, authors give their consent for the digital copy of their work to be used subject to the conditions specified on the Library Thesis Consent Form. AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Adrian P. Kells A thesis submitted in fulfillment of the requirements for the Degree of Doctor of Philosophy in Pharmacology at The University of Auckland, March, 2007. AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Abstract Progressive degeneration in the central nervous system (CNS) of Huntington’s disease (HD) patients is a relentless debilitating process, resulting from the inheritance of a single gene mutation. With limited knowledge of the underlying pathological molecular mechanisms, pharmaceutical intervention has todate not provided any effective clinical treatment strategies to attenuate or compensate the neuronal cell death. Attention has therefore turned to biotherapeutic molecules and novel treatment approaches to promote restoration and protection of selectively vulnerable populations of neurons in the HD brain. Rapid advances in vectorology and gene-based medicine over the past decade have opened the way for safe and efficient delivery of biotherapeutics to the CNS. With numerous factors known to regulate the development, plasticity and maintenance of the mammalian nervous system many proteins have emerged as potential therapeutic agents to alleviate HD progression. This investigative study utilised gene delivery vectors derived from the non-pathogenic adeno-associated virus (AAV) to direct highlevel expression of brain-derived neurotrophic factor (BDNF), glial cell-line derived neurotrophic factor (GDNF), Bcl-xL or X-linked inhibitor of apoptosis protein (XIAP) within the rodent striatum. Maintenance of the basal ganglia and functional behaviour deficits were assessed following excitotoxic insult of the striatum by quinolinic acid (QA), a neurotoxic model of HD pathology. Enhanced striatal expression of BDNF prior to QA-induced lesioning provided maintenance of the striosome-matrix organisation of the striatum, attenuating impairments of sensorimotor behaviour with a 36-38% increase in the maintenance of DARPP-32 / krox-24 expressing striatal neurons, reduced striatal atrophy and increased maintenance of striatonigral projections. Higher levels of BDNF however induced seizures and weight-loss highlighting the need to provide regulatable control over biotherapeutic protein expression. Continuous high-expression of BDNF or GDNF resulted in a downregulation of intracellular signal mediating proteins including DARPP-32, with AAV-GDNF not found to enhance the overall maintenance of striatal neurons. Neither of the anti-apoptotic factors provided significant protection of transduced striatal neurons but tended towards ameliorating QAinduced behavioural deficits, displaying behaviour – pathology correlations with the survival of parvalbumin-expressing neurons in the globus pallidus. The results of this thesis suggest BDNF as a promising putative biotherapeutic for HD, but emphasises the requirement to control expression following gene delivery, and for further elucidation of the physiological impact that enhanced expression of endogenous factors has on the host cells. Additionally the maintenance of neural networks beyond the caudate-putamen will be vital to ensuring efficient clinical outcomes for HD. II AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Acknowledgements I wish to specifically convey my gratitude to a number of people who have assisted either directly or indirectly over the past four years to ensuring the completion of this thesis. Specifically I would like to thank my supervisor Dr Bronwen Connor for having provided the opportunity to have conducted this research. Bronwen has always been optimistic towards all aspects of the research providing valuable rationality to study design and assessment, while always encouraging self belief to pursue my own initiatives. Thanks to Professor Richard Faull, my co-supervisor. A radiating enthusiasm for life and Richard’s passion to unravel the workings of the human brain was a constant inspiration. His interest in this study and vast knowledge of neuroanatomy always provided a fresh perspective to my evaluation. To Andrew, Rebecca, Kevin and Elena, your company as fellow PhD students throughout the highs and lows of our complementary investigations has been truly invaluable, and I sincerely wish you each the best in all your future endeavours. Particular thanks to Rebecca for patiently guiding me around the invisible barriers of molecular biology, and to Andrew for assistance with animal modelling and numerous discussions of the subsequent neurogenic phenomena – a welcome distraction. Dr Stephanie Hughes, for your counsel surrounding methodology and constructive feedback on written aspects of this thesis, I extent my gratitude. And to my remaining colleagues in the Neural Repair and Neurogenesis Laboratory and wider HRC Neuroscience collaboration that have provided advice, support and friendship, thank you. To all the staff of the Animal Resource Unit, your always willing assistance to ensure the greatest of care was provided for the multitude of rats that enabled these investigations to be conducted has been greatly appreciated. Finally to my Parents who have provided unwavering moral support throughout all of my university studies and have always encouraged me to pursue my own interests, without your loving support this thesis would not have been possible. And lastly to Petrea for your loyal friendship and patience in enduring with me through the final year of this thesis – this journey is now complete. Financial support of this thesis was provided by an Auckland Medical Research Foundation research grant, and scholarships awarded by both the NZ Foundation for Research Science and Technology, and The University of Auckland. III AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Journal Publications Research Articles Kells AP, Henry RA, Connor B. (2007) AAV-BDNF Mediated Attenuation of QA-Induced Neuropathology and Motor Function Impairment. Submitted to Gene Therapy. Kells AP, Henry RA, Connor B. (2007) AAV-Mediated Expression of Bcl-xL or XIAP Fails to Increase Neuronal Resistance against QA-induced Striatal Lesioning. Submitted to Experimental Neurology. Kells AP, Henry RA, Hughes SM, Connor B. (2006) Verification of Functional AAV-mediated Neurotrophic and Anti-apoptotic Factor Expression. Journal of Neuroscience Methods. 161(2): 291-300 Abstracts Kells AP, Henry RA, Hughes SM, Faull, RLM, Connor B. (2007) Attenuation of Functional Deficits in the QA Model of Huntington’s Disease following AAV Vector Delivery. 10th Annual Meeting of the American Society of Gene Therapy, Seattle, WA, USA. Molecular Therapy 15: S209 Kells AP, Henry RA, Hughes SM, Faull, RLM, Connor B. (2006) Protection against Huntington’s Disease Progression: AAV-mediated Delivery of Biotherapeutics. 9th Annual Meeting of the American Society of Gene Therapy, Baltimore, MD, USA. Molecular Therapy 13: S96 Kells AP, Henry RA, Hughes SM, Faull, RLM, Connor B. (2005) Investigating the Protective Effect of GDNF and Bcl-xL Gene Delivery in a Rat Model of Huntington’s Disease. 9th International Conference on Neural Transplantation and Repair, Taipei, Taiwan. IV AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Table of Contents Abstract II Acknowledgements III Journal Publications IV List of Figures XII List of Tables XV Abbreviations XVI Chapter 1 Review of Published Literature 1 1.1 Huntington’s Disease ..........................................................................................................1 1.2 Huntington’s Disease: Neuropathology .............................................................................2 1.3 Animal Models of Huntington’s Disease............................................................................5 1.3.1 Transgenic models of Huntington’s disease 5 1.3.2 Chemical neurotoxin models of Huntington’s disease 6 1.4 Therapeutic Intervention for Huntington’s Disease..........................................................8 1.5 In vivo Gene Delivery Vectors ..........................................................................................10 1.6 1.7 1.8 1.5.1 Herpes simplex and adeno viral vectors 10 1.5.2 Adeno-associated viral vectors 11 1.5.3 Lentiviral vectors 13 Neurotrophic Factors as Biotherapeutic Agents for HD.................................................13 1.6.1 Nerve growth factor 15 1.6.2 Brain derived neurotrophic factor 16 1.6.3 Glial cell-line derived neurotrophic factor 22 1.6.4 Ciliary neurotrophic factor 23 Apoptosis and Huntington’s Disease Neurodegeneration ...............................................24 1.7.1 Anti-apoptotic Bcl-2 and Bcl-xL proteins 29 1.7.2 Inhibitors of apoptosis 30 Summary...........................................................................................................................31 V AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study 1.9 Thesis Objectives ..............................................................................................................33 Chapter 2 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing 35 2.1 Overview ...........................................................................................................................35 2.2 AAV Vector Development and Production Procedures ..................................................35 2.3 2.4 2.2.1 Molecular biology protocols 2.2.1.1 PCR Cloning 2.2.1.2 Agarose Gel Extraction 2.2.1.3 p-GEM®-T Easy Vector 2.2.1.4 Heatshock Transformation 2.2.1.5 Colony PCR 2.2.1.6 Restriction Enzyme Digestions 2.2.1.7 Ligation into the AAV Expression Cassette 2.2.1.8 Plasmid Amplification and Purification 2.2.1.9 DNA Sequencing 36 36 37 38 38 38 38 39 39 40 2.2.2 AAV plasmid construction 40 2.2.3 Mammalian cell culture and analysis protocols 2.2.3.1 HEK 293 and HT-1080 Cell Culture 2.2.3.2 In Vitro Immunocytochemistry 2.2.3.3 Hoechst Nuclear Staining 44 44 45 45 2.2.4 AAV expression cassette transfection testing 46 2.2.5 AAV vector production 2.2.5.1 Packaging and Purification of AAV Vector Particles 2.2.5.2 Genomic Particle Titre 46 46 47 2.2.6 In vitro transduction testing 48 Functional AAV-Mediated Protein Expression Testing In Vitro ....................................49 2.3.1 Isolating and culturing primary cells 49 2.3.2 AAV-BDNF 50 2.3.3 AAV-GDNF 50 2.3.4 AAV-Bcl-xL 51 2.3.5 AAV-XIAP 52 Results ...............................................................................................................................52 2.4.1 AAV plasmid construction 52 2.4.2 AAV vector production 54 2.4.3 AAV vector transduction 54 2.4.4 Functional protein expression 2.4.4.1 Brain Derived Neurotrophic Factor 2.4.4.2 Glial cell-lne Derived Neurotrophic Factor 56 56 56 VI AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study 2.4.4.3 Bcl-xL 2.4.4.4 X-linked Inhibitor of Apoptosis Protein 2.5 59 61 Discussion..........................................................................................................................64 2.5.1 AAV vector construction 2.5.2 Functional protein expression 65 2.5.2.1 Differentiation Assays to Confirm Neurotrophic Factor Activity 65 2.5.2.2 Cell-Survival Assays to Confirm Anti-Apoptotic Protein Function 68 2.5.3 Summary Chapter 3 64 69 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion 71 3.1 Introduction ......................................................................................................................71 3.2 Optimisation and Characterisation Procedures ..............................................................71 3.3 3.4 3.2.1 Animals and surgeries 71 3.2.2 Behavioural assessment 72 3.2.3 Neuropathological analysis 72 QA Lesion Optimisation Analysis ....................................................................................73 3.3.1 Initial QA trial 73 3.3.2 Trial 2: Lower QA quantity 74 3.3.3 Trial 3: Behaviour testing – 30nmol QA 76 3.3.4 QA lesion model characterisation – 50nmol QA 76 3.3.4.1 Functional Behaviour Assessment of Unilateral QA Lesioned Rats 77 3.3.4.2 Neuropathological Assessment of QA Lesion 79 Discussion..........................................................................................................................82 3.4.1 QA-induced neuropathology 82 3.4.2 QA-induced behavioural impairments 83 Chapter 4 Neuroprotective Study Methods 85 4.1 Overview ...........................................................................................................................85 4.2 Neuroprotective Investigations ........................................................................................86 4.3 4.2.1 In vivo expression testing 86 4.2.2 Initial investigations: AAV-BDNF (high-titre), AAV-GDNF, AAV-Bcl-xL 86 4.2.3 Follow-up investigations: AAV-BDNF (diluted), AAV-Bcl-xL, AAV-XIAP 87 Animals .............................................................................................................................88 VII AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study 4.4 4.5 Stereotaxic Surgeries: AAV Vector Delivery and QA Injection .....................................89 4.4.1 Operating procedure 89 4.4.2 Determination of initial AAV vector injections parameters 90 Functional Behavioural Analysis .....................................................................................92 4.5.1 Spontaneous exploratory forelimb use 92 4.5.2 “Corridor” task 93 4.5.3 Drug-induced rotational analysis 94 4.6 Quantitative Transgene ELISA........................................................................................95 4.7 Immunohistochemical Analysis........................................................................................96 4.8 4.7.1 Brain tissue collection and processing 96 4.7.2 DAB-staining immunocytochemistry protocol 97 4.7.3 Fluorescent immunocytochemistry analysis 98 4.7.4 Microscopy and stereology 4.7.4.1 Striatal Neuron Stereology 4.7.4.2 Striatal Atrophy 4.7.4.3 Striatal TH-Staining Intensity 4.7.4.4 Maintenance of Striatonigral Projections 4.7.4.5 Pallidal Neuron Stereology 4.7.4.6 Confocal Microscopy 99 99 100 100 100 100 100 Statistical Analysis ..........................................................................................................101 Chapter 5 Neurotrophic Factor Delivery: AAV-BDNF and AAVGDNF 102 5.1 Overview .........................................................................................................................102 5.2 Procedures ......................................................................................................................102 5.3 Animal Welfare...............................................................................................................103 5.4 In Vivo AAV Vector Testing...........................................................................................104 5.5 BDNF and GDNF Expression Level ..............................................................................106 5.6 Functional Behaviour Assessment .................................................................................107 5.6.1 Spontaneous forelimb use 108 5.6.2 Drug-induced rotational behaviour 5.6.2.1 Apomorphine 5.6.2.2 Amphetamine 111 111 113 5.6.3 Sensorimotor “corridor” task 114 VIII AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study 5.7 5.8 Immunocytochemical Analysis.......................................................................................115 5.7.1 AAV-mediated transgene expression and QA lesioning of the striatum 5.7.1.1 Initial Study – AAV-BDNF and AAV-GDNF Delivery 5.7.1.2 Study 2 – Reduced Titre AAV-BDNF Delivery 5.7.1.3 Stereological Analysis – DARPP-32 5.7.1.4 Striatal Atrophy 116 117 119 120 122 5.7.2 Krox-24 expression 123 5.7.3 Analysis of transduced striatal cells 127 5.7.4 Dopaminergic innervations of the striatum 131 5.7.5 Cortical projection fibres in the striatum 133 5.7.6 Striatal projection nuclei 5.7.6.1 Substantia Nigra 5.7.6.2 Globus Pallidus 134 134 137 5.7.7 Pathological correlations with functional behaviour impairments 5.7.7.1 Spontaneous Ipsilateral Forelimb Use Bias 5.7.7.2 Contralateral Sensorimotor Neglect 5.7.7.3 Apomorphine-Induced Rotations 140 140 141 142 Discussion........................................................................................................................144 5.8.1 AAV-mediated BDNF or GDNF expression prior to QA 5.8.1.1 Undiluted AAV-BDNF 5.8.1.2 AAV-GDNF 144 144 146 5.8.2 Reduced-titre AAV-BDNF behavioural and pathological protection 5.8.2.1 Behavioural Protection 5.8.2.2 Neuropathological Protection 5.8.2.3 Pathological and Behavioural Correlations 149 149 151 153 5.8.3 Conclusion 155 Chapter 6 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAVXIAP 156 6.1 Overview .........................................................................................................................156 6.2 Procedures ......................................................................................................................156 6.3 In Vivo AAV Vector Testing...........................................................................................157 6.4 Bcl-xL and XIAP Expression Level ................................................................................160 6.5 Functional Behaviour Assessment .................................................................................161 6.5.1 Spontaneous forelimb use 161 6.5.2 Drug-induced rotational behaviour 6.5.2.1 Apomorphine 6.5.2.2 Amphetamine 164 165 166 IX AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study 6.5.3 6.6 6.7 Sensorimotor “corridor” task 167 Immunocytochemical Analysis.......................................................................................168 6.6.1 AAV-mediated transgene expression and QA lesioning of the striatum 6.6.1.1 Study 1 – AAV-Bcl-xL Delivery 6.6.1.2 Study 2 – AAV-Bcl-xL and AAV-XIAP Delivery 6.6.1.3 Stereological Analysis – DARPP-32 6.6.1.4 Striatal Atrophy 168 168 170 171 173 6.6.2 Krox-24 expression 174 6.6.3 Analysis of transduced striatal cells 176 6.6.4 Dopaminergic innervations of the striatum 178 6.6.5 Cortical projection fibres in the striatum 179 6.6.6 Striatal projection nuclei 6.6.6.1 Substantia Nigra 6.6.6.2 Globus Pallidus 181 181 184 6.6.7 Pathological correlations with functional behaviour impairments 6.6.7.1 Spontaneous Forelimb Use 6.6.7.2 Sensorimotor Neglect 6.6.7.3 Apomorphine-Induced Rotations 187 187 188 189 Discussion........................................................................................................................190 6.7.1 AAV-mediated Bcl-xL or XIAP expression prior to QA 6.7.1.1 Neuropathological Changes 6.7.1.2 Behavioural Protection 190 191 192 6.7.2 Comparison with previous studies 194 6.7.3 Conclusion 194 Chapter 7 General Discussion 196 7.1 AAV-mediated Gene Delivery........................................................................................196 7.2 Preventative Therapy for Huntington’s Disease ...........................................................198 7.3 Conclusion.......................................................................................................................204 Appendix A: Gene Sequences and DNA Plasmids A.1 206 cDNA Gene Sequences....................................................................................................206 A.1.1 Brain Derived Neurotrophic Factor 206 A.1.2 Glial cell-line Derived Neurotrophic Factor 207 A.1.3 Bcl-xL 208 A.1.4 X-linked Inhibitor of Apoptosis 208 X AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study A.2 DNA Plasmid Maps ........................................................................................................210 A.2.1 AAV Backbone Plasmid 210 A.2.2 AAV-Luciferase 211 A.2.3 AAV Helper Plasmids A.2.3.1 AAV rep and cap genes A.2.3.2 Adenoviral packaging genes 212 212 213 Appendix B: General Materials & Protocols 214 B.1 Molecular Biology...........................................................................................................214 B.2 Cell Culture.....................................................................................................................215 B.2.1 Tissue culture media 215 B.2.2 Cell counting 215 B.3 AAV Vector Genomic Titering ......................................................................................216 B.4 Immunocytochemistry Buffers.......................................................................................216 B.5 Antibodies .......................................................................................................................217 Literature References 218 XI AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study List of Figures Chapter 1 Review of Published Literature Figure 1-1 Potential mechanisms of mutant huntingtin induced cellular pathogenesis.....................4 Figure 1-2 Co-transfection rAAV packaging ................................................................................12 Figure 1-3 TrkB signalling pathways controlling dendritic protein synthesis ................................18 Figure 1-4 Mutant huntingtin disrupts transcription of BDNF and vesicle transportation from the cortex to the striatum. ............................................................................................20 Figure 1-5 Potential involvement of Ca2+ signalling in the induction of apoptosis in HD ..............26 Figure 1-6 Mitochondrial permeabilisation in intrinsic apoptosis..................................................27 Figure 1-7 Model of neurotrophic factor and anti-apoptotic factor supplied protection. ................32 Chapter 2 Adeno-associated Viral Vectors Figure 2-1 AAV expression plasmid ............................................................................................41 Figure 2-2 Transfection of AAV backbone plasmids ....................................................................53 Figure 2-3 AAV vector transduction in vitro ................................................................................55 Figure 2-4 Phenotypic differentiation of primary striatal cultures .................................................57 Figure 2-5 AAV-BDNF induced calbindin expression in embryonic striatal cultures....................57 Figure 2-6 Phenotypic differentiation of primary ventral mesencephalon cultures ........................58 Figure 2-7 AAV-GDNF induced TH expression in ventral mesencephalon cultures .....................58 Figure 2-8 AAV-Bcl-xL reduced staurosporine-induced apoptosis ................................................59 Figure 2-9 AAV-Bcl-xL transduced cortical cells following staurosporine-induced apoptosis .......60 Figure 2-10 AAV-XIAP reduced staurosporine-induced apoptosis .................................................61 Figure 2-11 Enhanced survival of AAV-XIAP transduced HT-1080 cells ......................................63 Chapter 3 Rodent Model of Huntington's Disease Figure 3-1 Initial 100 and 50 nmol QA lesion assessment ............................................................74 Figure 3-2 QA lesion testing: 50 and 30 nmol intrastriatal QA injections .....................................75 Figure 3-3 QA lesioning induced behavioural deficits ..................................................................78 Figure 3-4 Pathological characterisation of the 50nmol QA lesion................................................80 Figure 3-5 Striatal interneurons following QA lesioning...............................................................81 Chapter 4 Neuroprotective Study Methods Figure 4-1 Neuroprotective study timeline ...................................................................................85 Figure 4-2 Intrastriatal AAV vector delivery ................................................................................91 XII AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Figure 4-3 Spontaneous exploratory forelimb use.........................................................................92 Figure 4-4 Sensorimotor “corridor” task.......................................................................................94 Figure 4-5 Drug-induced rotational behaviour ..............................................................................95 Chapter 5 Neurotrophic Factor Delivery Figure 5-1 Spread of undiluted AAV-BDNF transduction and BDNF expression .......................104 Figure 5-2 Spread of AAV-GDNF transduction and GDNF expression ......................................105 Figure 5-3 Spread of the reduced-titre AAV-BDNF transduction and BDNF expression ............106 Figure 5-4 ELISA quantification of BDNF and GDNF expression in the striatum ......................107 Figure 5-5 Spontaneous ipsilateral forelimb use asymmetry score ..............................................109 Figure 5-6 Spontaneous exploratory rearing activity...................................................................110 Figure 5-7 Apomorphine-induced rotational behaviour ..............................................................112 Figure 5-8 Total apomorphine-induced rotations ........................................................................113 Figure 5-9 Amphetamine-induced rotational behaviour ..............................................................114 Figure 5-10 Preferential food selection in the sensorimotor “corridor” task ..................................115 Figure 5-11 High-titre AAV-BDNF transduction and striatal cell maintenance ............................116 Figure 5-12 AAV-GDNF transduction and striatal cell maintenance ............................................117 Figure 5-13 AAV-Luciferase transduction and striatal cell maintenance.......................................118 Figure 5-14 Diluted AAV-BDNF administration and striatal cell maintenance .............................119 Figure 5-15 Two-site AAV-Luciferase injection and striatal cell maintenance..............................120 Figure 5-16 Maintenance of DARPP-32 positive striatal projection neurons.................................121 Figure 5-17 Atrophy of the lesioned striatum ...............................................................................123 Figure 5-18 Double-immunofluorescent labelling of striatal neurons............................................124 Figure 5-19 Krox-24 expression in the striatum............................................................................125 Figure 5-20 Maintenance of krox-24 expressing striatal neurons ..................................................126 Figure 5-21 Immunofluorescent analysis of AAV-Luciferase transduced cells .............................128 Figure 5-22 Immunofluorescent analysis of AAV-BDNF transduced cells in the striatum ............129 Figure 5-23 Immunofluorescent analysis of AAV-GDNF transduced cells in the striatum............130 Figure 5-24 Tyrosine hydroxylase expression in the striatum .......................................................131 Figure 5-25 Maintenance of tyrosine hydroxylase striatal innervations.........................................132 Figure 5-26 SMI32 expression in the cortex and striatum.............................................................133 Figure 5-27 Visual assessment of the substantia nigra in control AAV-Luciferase treated rats......135 Figure 5-28 Visual assessment of the substantia nigra in reduced-titre AAV-BDNF treated rats...136 Figure 5-29 DARPP-32 innervations of the substantia nigra.........................................................137 Figure 5-30 Visual assessment of the globus pallidus following AAV-Luciferase treatment .........138 XIII AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Figure 5-31 Visual assessment of the globus pallidus following reduced-titre AAV-BDNF treatment...................................................................................................................139 Figure 5-32 Survival of parvalbumin-positive neurons in the globus pallidus ...............................140 Figure 5-33 Preferential forelimb use correlation with striatum and globus pallidus cell loss........141 Figure 5-34 Sensorimotor neglect correlation with striatal and pallidal neuronal cell loss.............142 Figure 5-35 Correlation of apomorphine-induced rotational behaviour with striatal and pallidal neuronal cell loss ......................................................................................................143 Chapter 6 Anti-apoptotic Factor Delivery Figure 6-1 Single delivery site for AAV-Bcl-xL transduction and subsequent Bcl-xL expression 157 Figure 6-2 AAV-Bcl-xL transduction and Bcl-xL expression following two-site striatal delivery 158 Figure 6-3 Spread of AAV-XIAP transduction and subsequent XIAP expression .......................159 Figure 6-4 ELISA quantification of Bcl-xL and XIAP expression in the striatum ........................161 Figure 6-5 Spontaneous ipsilateral forelimb use asymmetry scores.............................................162 Figure 6-6 Spontaneous exploratory rearing activity...................................................................163 Figure 6-7 Apomorphine-induced rotational behaviour ..............................................................164 Figure 6-8 Total apomorphine-induced rotations ........................................................................165 Figure 6-9 Amphetamine-induced rotational behaviour ..............................................................166 Figure 6-10 Preferential food selection in the sensorimotor “corridor” task ..................................167 Figure 6-11 Single injection site AAV-Bcl-xL transduction and striatal cell maintenance..............169 Figure 6-12 Dual injection site AAV-Bcl-xL transduction and striatal cell maintenance ................170 Figure 6-13 Dual injection site AAV-XIAP transduction and striatal cell maintenance.................171 Figure 6-14 Maintenance of DARPP-32 striatal projection neurons..............................................172 Figure 6-15 Atrophy of the lesioned striatum ...............................................................................173 Figure 6-16 Krox-24 expression in the striatum............................................................................175 Figure 6-17 Maintenance of krox-24 expressing striatal neurons ..................................................176 Figure 6-18 Immunofluorescent double-labelling of AAV-Bcl-xL transduced striatal neurons ......177 Figure 6-19 Immunofluorescent double-labelling of AAV-XIAP transduced striatal neurons .......178 Figure 6-20 Tyrosine hydroxylase expression in the striatum .......................................................179 Figure 6-21 SMI32 expression in the striatum ..............................................................................180 Figure 6-22 DARPP-32 innervations of the substantia nigra.........................................................181 Figure 6-23 Visual assessment of the substantia nigra in AAV-Bcl-xL treated rats........................182 Figure 6-24 Visual assessment of the substantia nigra in AAV-XIAP treated rats.........................183 Figure 6-25 Survival of parvalbumin-positive neurons in the globus pallidus ...............................184 Figure 6-26 Visual assessment of the globus pallidus following AAV-Bcl-xL treatment ...............185 Figure 6-27 Visual assessment of the globus pallidus following AAV-XIAP treatment ................186 XIV AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Figure 6-28 Preferential forelimb usage correlation with neuron loss in the striatum and globus pallidus .....................................................................................................................187 Figure 6-29 Sensorimotor neglect correlation with striatal and pallidal neuronal cell loss.............188 Figure 6-30 Correlation of apomorphine-induced rotational behaviour with striatal and pallidal neuronal cell loss ......................................................................................................189 List of Tables Chapter 1 Table 1-1 Chapter 2 Review of Published Literature Neuronal phenotypes within the striatum.......................................................................3 Adeno-associated Viral Vectors Table 2-1 Source of cDNA sequences and AAV Plasmids ..........................................................41 Table 2-2 PCR primer sequences ................................................................................................42 Table 2-3 Genes-of-interest.........................................................................................................43 Table 2-4 New England Biolabs (NEB) restriction enzymes and buffers .....................................43 Table 2-5 Primary antibodies for in vitro immunocytochemistry.................................................45 Table 2-6 Genomic titres of AAV vector stocks..........................................................................54 Chapter 3 Table 3-1 Chapter 4 Rodent Model of Huntington's Disease QA lesion optimisation trials.......................................................................................72 Neuroprotective Study Methods Table 4-1 Stereotaxic injection coordinates for AAV vector delivery ..........................................86 Table 4-2 First neuroprotective investigation timeline.................................................................87 Table 4-3 Second neuroprotective investigation timeline.............................................................87 Table 4-4 Allocation of animals ..................................................................................................88 Table 4-5 Dilutions used for ELISA quantification of in vivo transgenic protein expression. .......96 Table 4-6 Primary antibodies for DAB-staining immunocytochemistry.......................................98 Table 4-7 Antibodies for co-immunofluorescent labelling...........................................................99 XV AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study Abbreviations 3-NP AAV ABTS Ad Amp ANOVA A-P, M-L, D-V BCA Bcl-xL BDNF BSA CAG CBA cDNA ChAT CIP CMV CNS CNTF Ct DAB DARPP-32 DMEM DNA E14/15 EDTA E. coli FBS GABA GDNF GFRα1 GPe GPi HA HD Hdh 3-nitropropionic acid Adeno-associated virus 2,2-Azino-di-3-ethylbenzthiazoline sulfonate Adenovirus Ampicillin Analysis of variance Anterior-Posterior, Medial-Lateral, Dorsal-Ventral Bicinchinonic acid Bcl-2-like protein long Brain derived neurotrophic factor Bovine serum albumin Cytosine-adenine-guanine Chicken-β-actin Complementary DNA Choline acetyltransferase Calf intestinal phosphatase Cytomegalovirus Central nervous system Ciliary neurotrophic factor Cycle time 3-3 diaminobenzidine tetrahydrochloride Dopamine- and adenosine 3', 5'-monophosphate-regulated phosphoprotein of 32 kDa Dulbecco's Modified Eagle's Medium Deoxyribonucleic acid Embryonic day 14/15 Ethylenediaminetetraacetic acid Escherichia coli Fetal bovine serum Gamma-amino butyric acid Glial cell-line derived neurotrophic factor GDNF family receptor α-1 Globus pallidus external segment Globus pallidus internal segment Hemagglutinin Huntington’s disease Huntingtin gene XVI AAV-vector Mediated Gene Delivery for Huntington’s Disease: An Investigative Therapeutic Study HEK293 HIAP HSV HT-1080 IAP IMDM ITR LB Luc LV MAPK N171-82Q NADPHd NAIP NEB NeuN NGF NMDA NOS NR2B p75NTR PBS PCR Pen PNS poly-Q QA R6/2 rh RM ANOVA SNc SNr ssDNA Strep STS TE TH WPRE XIAP Human embryonic kidney 293 cells Human inhibitor of apoptosis Herpes simplex virus Human osteosarcoma cells Inhibitor of apoptosis protein Iscove’s Modified Dulbecco’s Media Inverted terminal repeats Luria-Bertani broth Luciferase Lentivirus Mitogen-activated protein kinase Transgenic mice with 171aa N-terminal fragment of Hdh with 82 CAG repeats Nicotinamide adenine diphosphate diaphorase Neuornal apoptosis inhibitor protein New England Biolabs Neuronal nuclei Nerve growth factor N-methly-D-aspartate Nitric oxide synthase NMDA receptor 2B subunit p75 neurotrophin receptor Phosphate buffered saline Polymerase chain reaction Penicillin Peripheral nervous system poly-glutamine tract Quinolinic acid Transgenic mice with exon 1 of Hdh containing ~150 CAG repeats Recombinant human Repeated measures analysis of variance Substantia nigra pars compacta Substantia nigra pars reticulata Single-stranded DNA Streptomycin Staurosporine Tris-EDTA buffer Tyrosine hydroxylase Woodchuck hepatitis post-transcriptional regulatory element X-linked inhibitor of apoptosis XVII Review of Published Literature Chapter 1 Review of Published Literature 1.1 Huntington’s Disease Huntington’s disease (HD) is an autosomal dominant genetic disorder that causes relentless neurodegeneration resulting in a progressive decline of physical motor function, cognitive abilities, and psychiatric disturbances first described by George Huntington (1872). Affecting up to 1 in 10,000 people depending on geographical location, physical HD symptom onset is generally between 30 and 50 years of age. While genetic testing is widely available to “at risk” relatives of HD patients, no effective treatment exists to prevent the onset, slow the progression, or alleviate the symptoms of HD. Despite the identification of the causative genetic defect – a trinucleotide cytosine-adenine-guanine (CAG) repeat expansion (>36 repeats) in exon-1 of the Hdh gene encoding a 350kDa protein termed huntingtin (Huntington's Disease Collaborative Research Group 1993), it remains elusive how this single gene defect results in the complex but selective pattern of HD neurodegeneration (Borrell-Pages et al. 2006). Huntingtin protein is widely expressed within the central nervous system (CNS) with the disease form containing an elongated polyglutamine (poly-Q) tract within the N-terminal domain (Trottier et al. 1995). With the lack of a clear understanding as to the molecular mechanisms connecting the selective cell death with the genetic defect, research towards therapeutic treatment has been focused on pharmacological intervention in general cell death processes, restoration of lost brain function by cellular transplantation, and genetic intervention to reduced the susceptibility of affected neurons (for review see (Handley et al. 2006)). Neurodegeneration generally occurs via three main mechanisms; metabolic compromise, excitotoxicity, or oxidative stress, acting independently or cooperatively (Alexi et al. 1997). Interfering with these processes or increasing the resistance of the neurons using neurotrophic factors has shown encouraging results in improving behavioural function and preventing neurodegeneration in HD models, thus providing hope that effective therapeutic treatment will soon become a reality for HD patients and their families. 1 Review of Published Literature 1.2 Huntington’s Disease: Neuropathology HD pathology is evident at a gross anatomical level by severe bilateral atrophy of the striatum and neocortex. Microscopically, neurodegeneration is most pronounced within the striatum where GABAergic projection neurons are selectively lost while interneurons resist degeneration (Reiner et al. 1988; Glass et al. 2000). The striatum is a highly organised structure receiving glutamatergic innervations from all cortical regions in a topographical arrangement (McGeorge and Faull 1989), and sending GABAergic efferent projections to the globus pallidus and substantia nigra, influencing basal ganglia output, thereby making the striatum a vital processing centre coordinating mood and movement (Selemon and Goldman-Rakic 1985; Alexander et al. 1986; Penney and Young 1986; Brown et al. 1997; Smith et al. 1998). Disruption of striatal function, responsible for the characteristic HD choreiform movements, is also likely to be the cause of the non-motor cognitive and emotional symptoms of HD (Penney and Young 1986; Reiner et al. 1988; Mitchell 1990). Vonsattel established a grading scale of end-point HD pathology based on post-mortem histopathological examination of the caudate nucleus and putamen (Vonsattel et al. 1985). This fivepoint grading scale provides a semi-quantitative assessment of the extent of neuronal cell loss in the caudate-putamen, and associated atrophy; with up to 30% neuronal loss but no apparent striatal atrophy in Grade 0, through to a 95% depletion of striatal neurons, severe atrophy and extensive gliosis in Grade 4. Immunohistochemical studies of the striatum have identified a number of distinct populations of neurons distinguishable by various neurochemical markers. The highly vulnerable GABAergic medium spiny projection neurons, comprising up to 95% of all striatal neurons, are divided into two groups by their neuro-modulatory transmitters’ enkephalin and substance P – corresponding with different projection targets (Table 1-1). The vast majority of these GABAergic projection neurons also contain the calcium binding protein calbindin, which is commonly used to visualise the extent of neurodegeneration within post-mortem HD human brains and in animal models of HD. The high expression of dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) by dopaminoceptive neurons can also be used as a general marker of the medium spiny projection neurons and their terminal projections (Ouimet et al. 1984; Ouimet et al. 1998). In addition to the projection neurons, the striatum also contains a varied population of interneurons of which the majority are also GABAergic and the remaining neurons being large aspiny cholinergic interneurons expressing the neurochemical marker choline acetyltransferase (ChAT). Aspiny GABAergic interneurons are divided into three subpopulations determined by their expression of the neurochemical markers calretinin, parvalbumin, or the co-expression of nicotinamide adenine 2 Review of Published Literature diphosphate diaphorase (NADPHd), nitric oxide synthase (NOS), neuropeptide Y and somatostatin (Table 1-1; (Alexi et al. 2000)). Neurons Transmitter Neurochemical markers Medium spiny projection neurons - Striatopallidal (GPe) GABA Enkephalin, calbindin, DARPP-32 - Striatoentopenduncular (GPi) and Striatonigral (SNr) GABA Substance P, dynorphin, calbindin, DARPP-32 - Striatonigral (SNc) GABA Substance P, dynorphin, DARPP-32 - Large aspiny ACh ChAT - Medium aspiny GABA NADPHd, NOS, neuropeptide Y, somatostatin - Medium aspiny GABA Parvalbumin - Small aspiny GABA Calretinin Aspiny interneurons Table 1-1 Neuronal phenotypes within the striatum Table modified from (Alexi et al. 2000) Progress in Neurobiology. Abbreviations: GPe, globus pallidus external segment; GPi, globus pallidus internal segment; SNc, substantia nigra pars compacta; GABA, γ-aminobutyric acid; ACh, acetylcholine; ChAT choline acetyltransferase; DARPP-32, dopamine- and cAMP-regulated phosphoprotein of 32 kDa; NADPHd, nicotinamide adenine diphosphate diaphorase; NOS nitric oxide synthase. HD involves a complex pattern of striatal degeneration in which there is not uniform degeneration throughout the striatum, but a “wave of degeneration” spreading laterally and ventrally through the caudate nucleus, putamen and nucleus accumbens in succession (Vonsattel et al. 1985). Enkephalin positive GABAergic medium spiny projection neurons projecting to the external globus pallidus are usually the first to degenerate correlating with the appearance of choreiform movements and early cognitive and psychiatric symptoms and indicating the degeneration / dysfunction of neurons within the caudate nucleus, which are largely involved in cognitive functions as well as motor control (Alexander et al. 1986; Reiner et al. 1988; Glass et al. 2000). Subsequent degeneration of substance P / dynorphin positive GABAergic medium spiny projection neurons correlates more with the onset of muscle dystonia (Reiner et al. 1988; Glass et al. 2000). Striatal interneurons are the least vulnerable with several subpopulations relatively spared in HD, specifically the somatostatin, neuropeptide Y, NADPHd positive and cholinergic interneurons (Reiner et al. 1988; Glass et al. 2000). Cortical neurons in layer III, V and VI of the cerebral cortex are also seen to degenerate in HD (Vonsattel et al. 1985; Hedreen et al. 1991; Macdonald and Halliday 2002). With layer III and V pyramidal neurons projecting to the striatum it is possible that the neurons are undergoing 3 Review of Published Literature secondary neurodegeneration following the loss of striatal connections, however with layer VI projecting to non-degenerative regions it would appear that independent degeneration is occurring within the cortex (Hedreen et al. 1991). In more severe grades of HD, neuronal cell loss occurs in many regions of the brain including the globus pallidus, subthalamic nucleus, cerebellum and thalamus (Vonsattel and DiFiglia 1998). This complex pattern of neurodegeneration does not reflect the expression pattern of huntingtin, nor the pattern of neuronal intranuclear inclusion or neuropil aggregate formation, suggesting that other Figure 1-1 Potential mechanisms of mutant huntingtin induced cellular pathogenesis Huntingtin is predominantly a cytoplasmic protein involved in numerous processes including vesicle transport of BDNF along microtubules through interaction with motor proteins which is inhibited by the expanded poly-Q tract causing abnormal protein folding. Mutant huntingtin also undergoes enhanced proteolytic cleavage with the N-terminal fragment containing the poly-Q repeat forming a β-sheet structure that can cause cellular disruption either as soluble monomers, oligomers or as insoluble aggregates. Toxicity in the cytoplasm may be induced via potentiation of NMDA glutamate receptors, mitochondrial effects, caspase activation or cellular disruption by perinuclear or neuritic aggregate formation. N-terminal fragments are also translocated to the nucleus where they form intranuclear inclusions can interfere with gene transcription of neuroprotective molecules including BDNF. (Reproduced from (Marx 2005), Science) 4 Review of Published Literature factors contribute to the selective vulnerability of the GABAergic medium spiny projection neurons (Saudou et al. 1998; Gutekunst et al. 1999). Although the molecular mechanism leading to HD neurodegeneration are not fully understood, the wild-type huntingtin protein has been shown to have clear roles in gene transcription, intracellular transport, and possesses anti-apoptotic properties; all of which are disrupted by the expanded poly-Q tract (Figure 1-1; (Borrell-Pages et al. 2006)). The disrupted regulation of transcription has been demonstrated by the reduced cytoplasmic sequestration of the transcription factor that inhibits BDNF transcription leading to a reduction in BDNF expression (Zuccato et al. 2001; Zuccato et al. 2003). Following N-terminal cleavage and translocation to the nucleus, a proposed requirement for HD neurodegeneration (Saudou et al. 1998), the poly-Q-huntingtin fragment can interfere with the regulation of transcription across a broad range of genes, inducing apoptosis (Luthi-Carter et al. 2000; Steffan et al. 2000; Nucifora et al. 2001). Intracellular microtubule-based vesicle transportation mediated by the binding of huntingtin with huntingtin-associated protein-1 and other components of the molecular motor machinery is disrupted by the expanded poly-Q tract causing a reduction in the vesicular transportation of BDNF (Engelender et al. 1997; Gauthier et al. 2004). Axonal transportation can also be physically blocked by the formation of N-terminal huntingtin aggregates within the axons (Li et al. 2000; Gunawardena et al. 2003). 1.3 Animal Models of Huntington’s Disease Animal models of neurodegenerative diseases are vital for advancing the development of therapeutic treatment strategies aimed at both preventing the degenerative progression and replacing the lost neurons. The development of genetic models of HD since the identification of the causative CAG repeat expansion in Hdh has greatly aided elucidation of pathological mechanisms and offer alternative cellular and animal models of HD to the acute neurotoxin induced mimicking of pathological neurodegeneration. Here I review current rodent models of HD with particular focus on their suitability for investigation of preventative therapeutic strategies to attenuate HD-induced neurodegeneration. The excitotoxic QA lesion model was ultimately selected as the most suitable model readily available for this thesis neuroprotective investigation. 1.3.1 Transgenic models of Huntington’s disease For HD, a single gene disorder, the possibility exists in theory that the exact neurodegenerative processes should be replicable in transgenic models. Numerous transgenic mice have been generated with either an N-terminal Hdh fragment containing an expanded CAG repeat in exon 1 (Mangiarini 5 Review of Published Literature et al. 1996; Schilling et al. 1999; Laforet et al. 2001), full-length Hdh (Reddy et al. 1998; Hodgson et al. 1999), or knock-in mice with additional CAG repeats inserted into the murine Hdh gene (Shelbourne et al. 1999; Wheeler et al. 2000; Lin et al. 2001). However to date these transgenic mice have not proven to completely replicate the specific neuronal cell death with only two lines showing significant striatal cell loss (Reddy et al. 1998; Laforet et al. 2001), although they do displayed some progressive phenotypic behaviours indicative of neurological impairment. Specifically the transgenic R6/2 mice progressively develop an irregular gait, resting tremor, stereotypic and abrupt irregularly timed movements, and epileptic seizures from nine weeks of age coinciding with a loss in body weight leading to death at 10-13 weeks (Mangiarini et al. 1996). Others show early hyperactivity followed by end stage hypoactivity (Reddy et al. 1998; Hodgson et al. 1999; Laforet et al. 2001). The restricted lifespan of these models may explain the absence of cell death despite neurological impairments and the presence of neuronal intranuclear inclusions, consisting of N-terminal fragments of the expanded huntingtin protein, similar to those found in the cortex and striatum of HD patients although these inclusions are more widespread in the transgenic mice (Morton et al. 2000). More recently a transgenic rat model of HD has been developed that better replicates the slow phenotypic progression of HD with emotional and cognitive impairments proceeding motor impairments (von Horsten et al. 2003). Specific loss of striatal neurons and intranuclear inclusions were evident from 12 months of age but not at six months, with no loss of cortical neurons never seen (Kantor et al. 2006). While transgenic models have become very valuable in elucidating the etiology of HD and the role of wild-type huntingtin, until recently they have been of restricted use for neuronal rescue / repair studies in which neuronal loss and function are the primary outcome measures. Overall it has become apparent that the more genetically accurate the model, the more subtle and variable the model becomes, and the greater it reflects the variability that is seen amongst HD patients (Hersch and Ferrante 2004). 1.3.2 Chemical neurotoxin models of Huntington’s disease As a substitute to genetic modelling, animal models that closely replicate the neuronal cell loss of HD can be generated through either excitotoxic lesioning of the striatum or metabolic impairment via mitochondrial disruption. Early hypotheses of HD etiology suggested the involvement of excitotoxins, with numerous studies using various excitotoxins to try and mimic the selective neurodegeneration (Beal et al. 1986; Schwarcz et al. 1988; Bruyn and Stoof 1990; DiFiglia 1990; Beal 1992a; Beal 1992b). Quinolinic acid (QA), an endogenous tryptophan metabolite present in the human brain at low concentrations, has proved to be the most successful excitotoxin at reproducing the selective degeneration of GABAergic medium spiny striatal projection neurons, while selectively 6 Review of Published Literature sparing somatostatin / neuropeptide Y striatal interneurons and fibres of passage (Schwarcz and Kohler 1983; Schwarcz et al. 1984; Beal et al. 1991; Brickell et al. 1999). Other excitotoxins, kanic acid, ibotenic acid, and N-methly-D-aspartate (NMDA), do not produce the selective sparing observed with QA (Beal et al. 1986). The resistance of neonatal animals and the high vulnerability of GABAergic medium spiny striatal projection neurons to QA gives support to the QA excitotoxin hypothesis of HD (Schwarcz et al. 1984). Neurons containing somatostatin or neuropeptide Y also express NADPHd, which could conceivably contribute to their survival in HD by allowing these neurons to take-up and metabolise QA (Schwarcz et al. 1984). This hypothesis that QA contributes to HD pathogenesis was recently supported by van Horsten and colleagues (2003) suggesting from indirect neurochemical data that QA synthesis may potentially be increased in their HD transgenic rats, which also maintain susceptibility to the excitotoxicity of QA (Winkler et al. 2006). Similarly, the chronic exposure to very low concentrations of QA has been shown to induce neuronal cell death (Chiarugi et al. 2001), and just recently it has been reported that wild-type huntingtin protects striatal neurons from QA-induced excitotoxicity (Leavitt et al. 2006). While the extent to which QA contributes to clinical HD is still to be conclusively determined, QA lesioned animals have been extensively used as models of HD pathology. Unfortunately while QA generates some of the behavioural and pathological changes observed in HD, the progressive nature of HD is not easily reproduced (Tobin and Signer 2000), with the majority of cell death occurring in the three days following intrastriatal injection (Portera-Cailliau et al. 1995; Hughes et al. 1996), and no further loss of phenotypic neuronal markers after seven days (Bazzett et al. 1994). Two models of chronic QA administration – microdialytic pump (Bazzett et al. 1993; Bazzett et al. 1994), and gradual release from polymeric implants (Haik et al. 2000) – have been developed and produce a progressive lesion more characteristic of HD, however the acute intrastriatal QA injection protocol is still widely utilized and readily produces consistent unilateral or bilateral striatal lesions. The progressive nature of HD is slightly better mimicked by 3-nitropropionic acid (3-NP), an irreversible inhibitor of succinate dehydrogenase that is commonly used as an alternative to excitotoxic animal models (Alexi et al. 1998). Inhibition of succinate dehydrogenase disrupts mitochondrial respiration and prevents ATP synthesis causing cell death by both apoptotic and necrotic mechanisms. The systemic administration of 3-NP via subcutaneously implanted minipumps causes the selective bilateral death of striatal GABAergic medium spiny projection neurons while relatively sparing the striatal interneurons and fibres of passage and thereby closely mimicking the pathology of HD (Borlongan et al. 1997). Systemic delivery allows for prolonged administration and therefore generates a prolonged insult, causing progressive neurodegeneration somewhat 7 Review of Published Literature reminiscent of the slow neuropathological progression of HD (Borlongan et al. 1997). This preferential loss of striatal projection neurons, over all other cell types following chronic systemic delivery of 3-NP, gives significant support to the hypothesis that energy impairment plays a major role in the etiology of HD. 3-NP does not however replicate the “wave of degeneration” or the sequential loss of subpopulations of striatal GABAergic projection neurons that occurs in HD (Sun et al. 2002). The striatal lesion is instead centred round the medial striatal artery and uniformly kills the projection neurons regardless of their target efferent nuclei (Nishino et al. 1995). In addition to the pathological neurodegeneration, the excitotoxic (QA) and energy impaired (3-NP) animal models also show varying degrees of behavioural deficits (Shear et al. 1998). A range of motor skill and cognitive function tests have been developed in addition to general open field observational tests to quantify behavioural impairments following unilateral or bilateral striatal lesions. Unilateral striatal lesions generally cause contralateral impairments of motor and sensory function which are commonly assessed by forelimb akinesia (Olsson et al. 1995), “staircase” testing – a measure of lateralised forelimb reaching and dexterity (Montoya et al. 1990; Montoya et al. 1991; Abrous et al. 1993), spontaneous exploratory forelimb use (Schallert et al. 2000), and sensory neglect (Salzberg-Brenhouse et al. 2003; Dowd et al. 2005). Amphetamine and apomorphine induced ipsilateral rotational behaviour is also characteristic of unilateral lesions resulting from an imbalance in striatal projection neurons or more specifically their dopamine receptors (Ungerstedt 1971; Schwarcz et al. 1979). Bilateral lesions show greater impairments in tests of cognitive ability (e.g. alternation T-maze and Morris water maze) than unilateral lesions in which the unlesioned hemisphere is though to be largely compensatory against the loss in cognitive ability (Pisa et al. 1981; Isacson et al. 1986; Popoli et al. 1994; Emerich et al. 1997a). 1.4 Therapeutic Intervention for Huntington’s Disease The development of efficient therapeutic treatment strategies for neurodegenerative diseases has been slow and relatively unsuccessful in generating long-term clinical benefits. The absence of longterm results for HD is thought to be related to the progressive nature of neurodegeneration, which is not addressed by any currently available pharmacological treatments that provide some symptomatic relief from the psychiatric disturbances, choreiform movements and cognitive deficits (Handley et al. 2006). As the molecular mechanisms by which mutant huntingtin appears to elicit neurodegeneration are pieced together, novel therapeutic approaches that directly interfere with the processing and 8 Review of Published Literature abnormal function of mutant huntingtin are beginning to be investigated (Qin et al. 2005). However with excitotoxicity appearing to play an important role in the selective pathology of HD, treatment strategies directed at intervening in general processes of cell death – including caspase activation, mitochondrial dysfunction and oxidative stress – may well hold significant clinical benefits for preventative therapy (Leegwater-Kim and Cha 2004; Handley et al. 2006). Independent of the specific therapeutic compound, efficient treatment against HD neurodegeneration will clearly require continuous supply of the therapeutic to the vulnerable neurons. This poses challenges associated with the need to cross the blood brain barrier, while avoiding deleterious side effects resultant from interactions with non-targeted cells, effectively preventing systemic delivery of many potentially beneficial compounds (Barinaga 1994; Tan and Aebischer 1996). However a number of pharmaceutical drugs – caspase inhibitors, mitochondrial enhancers and glutamate receptor blockers – have now been examined in clinical trials, having previously shown sufficient efficacy in preclinical research (Bonelli et al. 2004; Handley et al. 2006). With direct intraparenchymal infusion of therapeutic agents limited by the physical requirements necessary to achieve long-term administration, attention has been directed to the use of novel gene transfer techniques and cellular transplantation to produce biotherapeutics in situ. The use of encapsulated cells – ciliary neurotrophic factor (CNTF)-producing fibroblasts (Emerich et al. 1996; Emerich et al. 1997b; Mittoux et al. 2000) or choroid plexus cells (Borlongan et al. 2004; Emerich et al. 2006) – implanted into the striatum have provided protection against excitotoxic neuronal death and behavioural deficits in both rodent and primate studies. The in vitro manipulation of cells prior to implantation in semi-permeable capsules allows direct assessment of the secreted factors before implanting, with the encapsulation preventing host immune rejection and also allowing for graft recovery (Tan and Aebischer 1996; Emerich 2004b). Encapsulated cell implants do however suffer from limited diffusion and down-regulation of transgene protein expression (Emerich et al. 1996; Emerich and Winn 2004a). Alternatively direct transplantation of genetically modified cells (ex vivo gene therapy) can be performed with the potential for migration throughout the striatum, thereby reducing required diffusion distances (Martinez-Serrano and Bjorklund 1996). Transplantation of neuronal progenitor cells may also provide a method for replacing lost neurons in a potential joint protection-restoration treatment approach (Fallon et al. 2000; Hsich et al. 2002). However cell grafts cannot be recovered in the event of adverse reactions, although the incorporation of a suicide gene may allow for graft elimination (Garin et al. 2001). In vivo gene delivery is appealing in that it potentially avoids a number of problems associated with other methods of delivery, including the need for any permanent delivery device or the use of 9 Review of Published Literature immortalized cell-lines and their potential adverse effects (Hsich et al. 2002). However, in vivo gene delivery does rely on engineering the patients own host cells to drive production of the therapeutic proteins and is not without its own disadvantages, including potential adverse toxicity, immune responses, or oncogenic potential due to insertional mutagenesis (Monahan and Samulski 2000; Hsich et al. 2002). The ongoing development of in vivo vectors with improved safety, control and stability of transgene expression, and reduced pathogenicity / toxicity all increase the appeal of in vivo gene therapy for long-term delivery of therapeutic proteins directly to vulnerable neurons. With both ex vivo and in vivo gene therapy techniques showing promise for delivering biotherapeutics to the CNS, I elected to employ a direct in vivo gene delivery technique allowing direct manipulation of the vulnerable striatal neurons and thereby enabling the investigation of both secreted and non-secreted intracellular acting therapeutic molecules using the same gene therapy protocols. 1.5 In vivo Gene Delivery Vectors The main requirement of vectors used for in vivo gene delivery to the CNS is their ability to efficiently deliver transgenes to terminally differentiated neurons, generating stable protein expression while avoiding the stimulation of inflammation and immunogenic host responses (Hsich et al. 2002). Viral-derived vectors have become the vectors of choice for many neurological applications due to their natural inclination to transfer their genetic material to targeted cells (Bjorklund et al. 2000; Hsich et al. 2002). The most promising vectors for direct CNS gene delivery are derived from human viruses including herpes simplex virus (HSV), adenovirus (Ad), adenoassociated virus (AAV), and lentivirus (LV) (Thomas et al. 2003). Each of these vectors are briefly reviewed here with particular emphasise on the properties of AAV vectors which I utilised for in vivo transduction of the rodent striatal neurons in the neuroprotective investigations. 1.5.1 Herpes simplex and adeno viral vectors First generation HSV and Ad vectors with large transgene capacities (HSV ~50kb, Ad 7.5kb) are efficient for gene delivery to many brain cell types, however the expression of viral proteins stimulate adverse immune reactions (Hsich et al. 2002). Elimination of viral genes has led to the development of HSV amplicon vectors with essentially no toxicity, and “gutless” Ad vectors with greatly reduced toxicity and immunogenicity (Thomas et al. 2000; Hsich et al. 2002). However, transgene expression following Ad-mediated gene delivery is not long-lasting and the presence of 10 Review of Published Literature neutralising antibodies against the Ad structural proteins block transduction following readministration of the same serotype vector (Zoltick et al. 2001). Pre-exiting immunity to HSV is also a limiting factor for HSV vectors (Lauterbach et al. 2005), which also only generate transient expression after elimination of the viral genes (Samaniego et al. 1997). 1.5.2 Adeno-associated viral vectors AAV is a very small (25nm) non-pathogenic parvovirus dependent on a helper virus, usually adenovirus, to proliferate. The wild-type AAV genome contains two open-reading frames rep (genes required for replication) and cap (genes encoding the capsid proteins) flanked by 145 bp inverted terminal repeat sequences (ITR; (Srivastava et al. 1983)). However only the ITR sequences are required in cis for replication and encapsulation of single stranded DNA, up to ~4.7kb, into the icosahedral AAV virion particle of 60 capsid proteins (Monahan and Samulski 2000; Hsich et al. 2002). This allows for the complete removal of viral genes in recombinant AAV (rAAV) vectors eliminating the potential for an immune response against the expression of viral proteins (Jooss et al. 1998), although the production of neutralising antibodies against the capsid proteins is still a concern, particularly for repeat vector delivery (Halbert et al. 2006). Production of rAAV vectors is typically undertaken by DNA plasmid co-transfection of a packaging cell line, usually human embryonic kidney 293 cells (HEK293), with the AAV-rep and -cap genes plus the necessary Ad genes supplied on separate constructs in trans to the ITR-flanked AAV expression cassette (Figure 1-2; (Xiao et al. 1998)). With the characterisation of at least eight distinct serotypes of AAV showing different host cell tropisms, and recent identification of over 100 additional variants (Gao et al. 2004), the majority of attention for gene transfer has been given to serotype-2 as the first serotype fully characterised (AAV2; (Samulski et al. 1982; Srivastava et al. 1983); for current review see (Wu et al. 2006)). AAV2 alone presents strong affinity for the heparan sulphate proteoglycan receptor which appears to mediate its binding-to and transduction of host cells (Summerford and Samulski 1998), and also allows heparin affinity column purification to produce high-titre recombinant vectors (Zolotukhin et al. 1999). The apparent clinical safety of AAV2 as a gene delivery vector has now been well tested in clinical trials (Carter 2005). With different serotype capsid proteins conferring selective tissue and cellular tropism, attention has been directed to altering the tropism and transduction efficiency of AAV2 and other AAV serotypes by either genetic alteration of the capsid proteins (Muzyczka and Warrington 2005) or production of chimeric (also called mosaic) rAAV virions containing a mixture of different capsid proteins to combine the transduction properties of different serotypes (Hauck et al. 2003; Rabinowitz et al. 2004). In respect to the transduction of neurons in the CNS, serotype-1 and -5 rAAV vectors display higher transduction efficiencies than 11 Review of Published Literature AAV2 vectors (Davidson et al. 2000; Burger et al. 2004). Recent production of chimeric vectors with serotype-1 and -2 capsid proteins (AAV1/2), combined in a 1:1 ratio and retaining the ability to be purified by heparin column, show enhanced neuronal transduction over standard AAV2 vectors (Hauck et al. 2003; Richichi et al. 2004). In general the serotype-2 ITR sequences flanking the expression cassette have been retained, as the cis-acting element to direct replication and packaging of the vector genome, irrespective of the packaging capsids. While wild-type serotype-2 AAV can integrate into the human genome at a specific site on chromosome 19 (Kotin et al. 1992), the rAAV vectors have been observed to integrate at non-specific sites (Linden et al. 1996; Ponnazhagan et al. 1997; Yang et al. 1997) however, the predominant persistence of the AAV genome in vivo is extrachromosomal as circular monomeric or multimeric structures with enhanced episomal stability (Clark et al. 1997; Duan et al. 1998; Duan et al. 1999; Schnepp et al. 2005). With the high stability of AAV genome in host cells the persistence of transgene expression following rAAV transduction appears dependent on the regulatory elements controlling transgene expression. Transgene expression from the hybrid CMV- Figure 1-2 Co-transfection rAAV packaging Production of rAAV is typically undertaken by co-transfection of HEK293 cells with separate plasmids containing the recombinant AAV genome (pAAV) with the expression cassette flanked by ITRs (red), the AAV rep and cap genes (pHelper), and a plasmid containing the necessary Ad viral genes (pAd). The trans acting AAV rep and Ad genes direct replication of single stranded rAAV genome and encapsulation into AAV virions constructed following expression of the AAV capsid proteins (Capx). Vector serotype is determined by the cap genes in the pHelper plasmid (Modified from (Merten et al. 2005), Gene Therapy). 12 Review of Published Literature chicken-β-actin (CBA) promoter was still stable in the rat brain after 25 months (Klein et al. 2002), however the CMV promoter has shown susceptibility to silencing by hypermethylation in some cells (Prosch et al. 1996). A major limiting factor for rAAV vectors is the relatively small transgene capacity (~4.7 kb), restricting delivery of larger transgenes and the incorporation of regulatory elements that will be vital for controlling expression in some clinical applications (Monahan and Samulski 2000; Hsich et al. 2002). Delivery of rAAV vectors to the CNS through direct intracranial injection allows targeting of specific populations of cells, however for larger structures the transduction efficiency can be hindered by limited vector spread, requiring multiple injection sites (Bjorklund et al. 2000). Ultraslow injections and co-infusion of mannitol or heparin have both been shown to increase transduction efficiency and distribution of transduced neurons by 200-300% (Mastakov et al. 2001; Nguyen et al. 2001). More recently, systemic delivery of mannitol has shown to provide even greater enhancement of rAAV vector spread and transduction efficiency (Burger et al. 2005). 1.5.3 Lentiviral vectors LV vectors are retroviral vectors derived from the immunodeficiency viruses HIV (human) and FIV (feline) (Poeschla 2003; Wiznerowicz and Trono 2005). In contrast to other retroviral vectors, LV can stably integrate into non-dividing cells as well as dividing cells, therefore generating long-term transgene expression (Wong et al. 2006). Extensive use of LV vectors has been delayed due to safety issues surrounding the production and application of vectors derived from such a highly pathogenic virus. However these biosafety issues have been addressed with further elimination of viral genes and the development of a self-inactivating (SIN) version (Miyoshi et al. 1998; Zufferey et al. 1998). LV vectors have a transgene capacity of at least 8kb and have high affinity and transduction efficiency for fully differentiated neurons (Watson et al. 2002; Wong et al. 2004). Primate studies have reported the absence of toxicity or inflammatory responses (Kordower et al. 1999; Bjorklund et al. 2000; Kordower et al. 2000; Palfi et al. 2002). Thus, as safety concerns are minimized, LV becomes a very attractive vector for long-term transgene expression in the CNS. 1.6 Neurotrophic Factors as Biotherapeutic Agents for HD Neurotrophic factors are proteins produced and secreted by neurons and glia that regulate the development, survival and function of neurons within the developing central and peripheral nervous system (Huang and Reichardt 2001). In addition, neurotrophic factors also contribute to activity13 Review of Published Literature dependant neuronal plasticity, biochemical function and survival of neurons within the mature adult nervous systems (Sofroniew et al. 2001). Expressed heterogeneously within the CNS, neurotrophic factors function through specific receptors differentially expressed by various neuronal and nonneuronal populations of cells (Mufson et al. 1999). Target-derived support appears vital in directing the development of neuronal circuits with growing axons expressing receptors that compete for limited neurotrophic factor expression in the target nuclei, thereby promoting neurite growth, synapse formation and regulating neuron survival to limit the number of neurons (Purves 1988; Burke 2006). While target-derived support is essential in the developing CNS, the degree to which mature, terminally differentiated neurons depend on target-derived trophic support is unclear with autocrine / paracrine activities potentially more important, although the ability to retrogradely transport neurotrophic factors is still maintained (Mufson et al. 1999; Murer et al. 2001). Widespread CNS neurotrophic factor expression, in addition to regions of abundant expression, and co-localization with neurotrophic factor receptors support proposed autocrine / paracrine interaction in promoting neuronal function and survival (Connor and Dragunow 1998). Increases in neurotrophic factor expression within adult animals following a neuronal degenerative event or neuronal injury is indicative of the support role that neurotrophic factors play in the adult CNS where they can act as anti-excitotoxins and antioxidants or can up-regulate calcium binding proteins and anti-apoptotic signals (Alexi et al. 2000; Alberch et al. 2004). Neurotrophic factor deprivation has also been shown to cause up-regulation of pro-apoptotic gene transcription, indicating the neurotrophic factor dependence of some mature neurons to survive and maintain neuronal function (Johnson et al. 1989; Oppenheim 1991). The expression of neurotrophic factors in the striatum and their modified expression in response to striatal lesioning (Marco et al. 2002), and in neurological diseases including HD (Ferrer et al. 2000), has led to neurotrophic factors being viewed as potential therapeutic agents (Alexi et al. 2000; Alberch et al. 2004). Administration of neurotrophic factors to the striatum in animal models of HD have been investigated via three broad techniques: direct intracerebellar infusion, transplantation of neurotrophic factor producing cells, or in vivo gene delivery. Four neurotrophic factors – the neurotrophins nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), glial cellline derived neurotrophic factor (GDNF), and CNTF – have received significant attention in relation to their potential therapeutic value as neuroprotective agents for HD (Alberch et al. 2004) and are reviewed here with particular focus on BDNF and GDNF which I selected to investigate further with in vivo AAV-mediated gene delivery prior to QA lesioning. Additionally many other neurotrophic factors have been demonstrated to have neuroprotective actions on striatal neurons including neurotrophin 4/5 (Alexi et al. 1997), neurturin (Perez-Navarro et al. 2000), transforming growth 14 Review of Published Literature factor-α (Alexi et al. 1997), basic fibroblast growth factor (Frim et al. 1993b; Kirschner et al. 1995) and activin-A (Hughes et al. 1999). The method of neurotrophic factor delivery to the striatal neurons has proven to be highly influential on the ultimate capacity to impart potent neuroprotection, with the prospect of efficient clinical treatment becoming reality, proved safe and efficient delivery procedures are developed. 1.6.1 Nerve growth factor NGF, the first identified neurotrophic factor (Levi-Montalcini and Hamburger 1953), is widely expressed throughout the CNS with high expression in regions of the CNS innervated by cholinergic neurons of the basal forebrain, particularly the hippocampus, olfactory bulb and neocortex (LeviMontalcini 1987; Mufson et al. 1994; Das et al. 2001). Expression of NGF in the developing striatum is seen to correlate with the differentiation of cholinergic interneurons which express the high-affinity NGF receptor TrkA and low-affinity p75 receptor (Mobley et al. 1989; Ringstedt et al. 1993; Steininger et al. 1993; Barbacid 1994). Co-localization of NGF and TrkA in the CNS (Barde 1989) and high NGF expression throughout adulthood, suggests NGF regulates maintenance and survival of adult neurons through autocrine / paracrine activity (Kokaia et al. 1993; Miranda et al. 1993). Enhanced striatal NGF levels following injection of glutamate receptor agonists further support the suggestion that NGF provides endogenous neuroprotection of the cholinergic neurons (Perez-Navarro et al. 1994; Strauss et al. 1994; Canals et al. 1998). NGF has been investigated in several animal models of HD (Alberch et al. 2004) with significant neuronal protection observed when released from NGF-producing cells transplanted into the striatum or corpus callosum prior to ipsilateral QA (Schumacher et al. 1991; Frim et al. 1993a; MartinezSerrano and Bjorklund 1996; Kordower et al. 1997), 3-NP (Frim et al. 1993c) or NMDA-agonist (Frim et al. 1993b) induced lesioning. These studies have demonstrated that locally produced NGF results in a significant reduction in striatal lesion volumes by 60-80% compared with untreated animals (Schumacher et al. 1991; Frim et al. 1993a; Frim et al. 1993b; Frim et al. 1993c; MartinezSerrano and Bjorklund 1996; Kordower et al. 1997). Martinez-Serrano and Bjorklund (1996) also observed protection of striatal neurons and their efferent projections to the substantia nigra and globus pallidus. Complete sparing of ChAT-immunoreactive cholinergic interneurons and sparing of NADPH histochemically stained neurons was reported following transplantation of NGF-producing cells (Schumacher et al. 1991; Frim et al. 1993a; Martinez-Serrano and Bjorklund 1996; Kordower et al. 1997). In contrast, direct intrastriatal NGF protein injections (Davies and Beardsall 1992) or prolonged infusions (Davies and Beardsall 1992; Venero et al. 1994; Anderson et al. 1996) failed to 15 Review of Published Literature protect the vulnerable striatal GABAergic projection neurons, but selectively protected cholinergic interneurons (Altar et al. 1992a; Perez-Navarro et al. 1994). The lack of NGF receptor expression by the GABAergic medium spiny projection neurons suggests that the neuroprotection of striatal GABAergic neurons, reportedly provided by NGF-producing cells, is not directly due to the actions of NGF, but possibly related to other factors secreted by the grafted cells, such as basic fibroblast growth factor that has been demonstrated to provide significant neuroprotection (Frim et al. 1993b; Kirschner et al. 1995), or the antioxidant properties of the grafted cells (Galpern et al. 1996). However both Frim et al. (1993b) and Martinez-Serrano and Bjorklund (1996) reported considerably less protection of the medium spiny projection neurons in their parallel investigations of BDNF ex vivo gene therapy than was provided by NGF-secreting cellular grafts, indicating a differential effect of the NGF or BDNF transgenic protein secretion. 1.6.2 Brain derived neurotrophic factor Closely related to NGF, BDNF is widely expressed throughout the CNS with expression increasing during fetal and early postnatal development, and is maintained throughout adult life (Friedman et al. 1991; Connor and Dragunow 1998). Increasing expression as CNS regions mature suggests BDNF plays a greater role in the maintenance and plasticity of the adult CNS than in development (Altar et al. 1997). Abundant BDNF expression in the hippocampus is thought to be important for maintaining a high degree of neuronal plasticity vital for the hippocampus’s role in learning and memory (Lindsay et al. 1994). Expression and secretion of BDNF is modified by numerous events including bioelectrical activity, neurotransmitters, hormones and cellular insults (Dragunow et al. 1993; Marty et al. 1997; Hughes et al. 1999; Murer et al. 2001). Activity-dependant release of BDNF from neurons (Allendoerfer et al. 1994) is proposed to be induced by depolarisation through a calcium-dependent mechanism – glutamate receptor agonists induce BDNF, while GABAA receptor agonists inhibit BDNF expression (Goodman et al. 1996; Marty et al. 1997). Cellular insults – hypoxia-ischemia and hypoglycaemic coma – have also been observed to increase both BDNF and TrkB receptor expression suggesting that endogenous BDNF plays a significant role in the general protection of the CNS (Merlio et al. 1993). More specifically related to HD, an intrastriatal injection of QA was found to induce an up-regulation of BDNF in cortical layers II, III, V and VI due to damage in the striatal target area (Canals et al. 2001), but no BDNF increase in the striatum (Canals et al. 1998). The TrkB receptor was also up-regulated by cortical neurons following intrastriatal QA injection (Checa et al. 2001), suggesting an endogenous mechanism to support cortical neurons following striatal damage. 16 Review of Published Literature High affinity TrkB receptors for BDNF are found throughout the CNS on both neurons and glia. Receptors are mostly localized to the axons, synaptic terminals and dendritic spines supporting the proposal that synapses are the main site at which BDNF functions in the CNS, with TrkB mediating synaptic plasticity and retrograde transport of BDNF (Drake et al. 1999; Watson et al. 1999; Aoki et al. 2000). It has been demonstrated that glial cells have the ability to internalise BDNF via truncated TrkB receptors lacking catalytic kinase activity, thus regulating BDNF availability and restricting diffusion (Yan et al. 1994; Biffo et al. 1995; Rubio 1997; Altar 1999). Although lacking catalytic kinase activity a truncated TrkB receptor isoform is the predominantly expressed isoform in the adult brain and is hypothesised to regulate BDNF signalling by impairing catalytic TrkB receptors and may also act as a cellular adhesion molecule regulating synapse plasticity and axonal outgrowth (Armanini et al. 1995; Murer et al. 2001). TrkB receptor expression appears to be regulated by mechanisms similar to those that also regulate BDNF expression, including bioelectrical activity and cellular insults – hypoxia-ischemia, hypoglycaemic coma and axotomy (Merlio et al. 1993). The dendritic targeting of BDNF and TrkB induced by bioelectrical activity suggests that autocrine activity is important in maintaining and regulating dendritic tree function (Tongiorgi et al. 1997). Intracellular TrkB receptors are also translocated to the plasma membrane in response to depolarisation, thus rapidly increasing responsiveness to BDNF potentially important for maintaining synapses (Meyer-Franke et al. 1998). Prolonged BDNF exposure however causes a down-regulation of catalytic TrkB receptors (Frank et al. 1996). In addition to the high affinity TrkB receptor, BDNF also binds to p75 neurotrophin receptor (p75NTR) which has traditionally been viewed as a coreceptor for the Trk neurotrophic factor receptors facilitating Trk dimerisation to allow high-affinity neurotrophin binding and modulation of Trk signalling (Chao 1994; Chao and Hempstead 1995). More recently however, p75NTR, a member of the tumor necrosis factor receptor family, has been demonstrated to elicit biological functions following neurotrophic factor binding including the induction of apoptosis in the absence of Trk receptors (Dobrowsky et al. 1994; Bamji et al. 1998; Boyd and Gordon 2002; Troy et al. 2002). As with all neurotrophins, BDNF is synthesised as a pro-neurotrophin which undergoes proteolytic cleavage by furin or proconvertases to produce the mature form of BDNF protein. However a number of recent studies have demonstrated that proneurotrophins are also physiologically active (Nykjaer et al. 2004), are endogenously secreted by neurons (Mowla et al. 2001; Chen et al. 2004; Teng et al. 2005), and can be cleaved extracellularly by plasmin and metalloproteinases (Lee et al. 2001). ProBDNF itself however does not stimulate TrkB phosphorylation or promote neurite outgrowth, but rather can induce apoptosis through activation of p75NTR and the coreceptor sortilin 17 Review of Published Literature (Teng et al. 2005). The induction of apoptosis appears to be dependent on the presence of the highaffinity sortilin receptor, with extracellular proteolytic cleavage to mature BDNF occurring in the absence of sortilin (Nykjaer et al. 2004). These entirely opposing actions of pro- and mature forms of BDNF indicate the delicate role that this neurotrophic factor plays in facilitating the survival and plasticity of BDNF dependent neurons in the mature CNS, with an ability to respond rapidly to a wide range of stimuli. In order to modulate neuronal plasticity BDNF acts in a localised synapse-specific manner that is thought to be regulated by the activity level of individual synapses potentially influencing the dendritic targeting of BDNF mRNA, the secretion of BDNF at active synapses, TrkB receptor expression and receptor-neurotrophin internalisation (for reviews see (Lu 2003; Kuipers and Bramham 2006)). Mature BDNF acting through the TrkB receptor appears to be a vital protein in the establishment of late-phase long-term potentiation (Pang et al. 2004) through the activation of the microtubule-associated protein (MAP) kinase cascade enhancing the transcription of several immediate early genes which are responsible for controlling specific neuronal function (Friedman and Greene 1999; Bolton et al. 2000). The synthesis of proteins within dendritic regions is activitydependent with BDNF-TrkB signalling central to regulating the translation of mRNA through phosphorylation of the eukaryotic initiation factor 4E (eIF4E) and eukaryotic elongation factor 2 (eEF2; Figure 1-3; (Bramham and Messaoudi 2005; Soule et al. 2006)). Figure 1-3 TrkB signalling pathways controlling dendritic protein synthesis BDNF binding to postsynaptic TrkB receptors activates a number of major signalling pathways which ultimately provide control over dendritic translation. BDNF-TrkB activation of PI3K and ERK cascades causes phosphorylation of eIF4E and enhances the initiation of cap-dependent mRNA translation. Bi-directional control over eEF2 phosphorylation state by TrkB signalling provides control over mRNA translation with phosphorylated eEF2 inhibiting peptide chain elongation. (Reproduced from (Soule et al. 2006)) 18 Review of Published Literature BDNF has also been demonstrated to play an important role in the promotion of neurogenesis in the adult brain, enhancing neuron generation and the migration of neuroblasts from the sub ventricular zone to the striatum and olfactory bulb (Zigova et al. 1998; Benraiss et al. 2001; Chmielnicki et al. 2004; Chen et al. 2007; Henry et al. 2007). BDNF-stimulated promotion of neurogenesis appears to be mediated via p75NTR signalling, with neurogenic precursor cells in the adult sub ventricular zone expressing the p75NTR receptor (Young et al. 2007). Survival and maturation of neuroblasts by BDNF has been shown to occur through TrkB signalling (Benraiss et al. 2001), however neurogenic precursor cells do not express Trk receptors suggesting that BDNF interacts with p75NTR to direct differentiation of precursor cells into neuroblasts by promoting withdrawal from the cell cycle and terminal differentiation (Cattaneo and McKay 1990; Ito et al. 2003; Chittka et al. 2004; Young et al. 2007). Although known primarily as a cell death inducing receptor, p75NTR expression by precursor cells in the sub ventricular zone does not appear to affect survival of precursor cells in the presence or absence of BDNF (Young et al. 2007), but has been shown to stimulate cell death in neuroblasts which are prevented from migrating (Gascon et al. 2007). Within both the developing and adult striatum, medium spiny neurons rely on BDNF to maintain their phenotype, function and survival (Mizuno et al. 1994; Ivkovic and Ehrlich 1999; Baquet et al. 2004) through activation of TrkB mediated signalling (Hetman et al. 1999; Vaillant et al. 1999; Gavalda et al. 2004). In addition, BDNF also supports neuronal survival by up-regulating anti-apoptotic Bcl-2 expression via TrkB mediated Akt activation (Schabitz et al. 2000; Almeida et al. 2005; Perez-Navarro et al. 2005). High levels of BDNF are present in the striatum with the striatal projection neurons expressing TrkB receptors (Costantini et al. 1999) and displaying a dependence on BDNF for survival (Saudou et al. 1998; Baquet et al. 2004). However with low levels of BDNF mRNA in the striatum, the majority of striatal BDNF is thought to be produced in the cortical neurons and anterogradely transported to the striatum ((Altar et al. 1997); Figure 1-4). Normal aged rats display a reduction by half in striatal BDNF levels (Katoh-Semba et al. 1998) suggesting a normal decline with aging that may increase the vulnerability of the striatal neurons. BDNF protein expression in HD has been investigated by Ferrer et al. (2000) using human post-mortem samples from grade-3 HD patients and age-matched non-neurological controls. BDNF protein expression was found to be reduced 53-82% in the caudate nucleus and putamen of the HD patients, but preserved in the parietal cortex, temporal cortex, and hippocampus. Most striatal BDNF is contained within the corticostriatal projections following anterograde transport, however the density of synaptic terminals in the HD striatum were unchanged and BDNF-positive fibres were still observed in HD patients (Ferrer et al. 2000). While this suggested a selective defect of striatal neurons and not a loss of cortical BDNF input, more recent investigation has shown mutant huntingtin both disrupts the transcription of BDNF (Zuccato et al. 19 Review of Published Literature 2001; Zuccato et al. 2003) and reduces the axonal trafficking of BDNF vesicles along microtubules ((Gauthier et al. 2004); Figure 1-4). Therefore maintaining or increasing BDNF protein expression directly within the caudate nucleus and putamen of HD patients may potentially be of therapeutic benefit in preventing or slowing the progression of neurodegeneration in HD. A B C D Figure 1-4 Mutant huntingtin disrupts transcription of BDNF and vesicle transportation from the cortex to the striatum. A large proportion of striatal BDNF is produced by the cortical neurons and anterogradely transported via corticostriatal projections and released at synapses in the striatum. Wild-type huntingtin is crucial for the transcription and transportation of BDNF, both of which are impaired by the expanded poly-Q tract. BDNF expression is regulated by a neuron-restrictive silencer element (NRSE) in the promoter region that facilitates the blocking of transcription through association with a repressor complex. The normal sequestration of Repressor element 1-silencing transcription factor (REST) by huntingtin in the cytoplasm prevents repressor complex formation allowing BDNF transcription (A). Mutant huntingtin is less capable of preventing REST from entering the nucleus and silencing the expression of NRSE regulated genes (B). Huntingtin is also involved in controlling microtubule transportation of BDNF vesicles through association with motor protein complexes (C). Mutant huntingtin is thought to cause a conformational change through enhanced binding to huntingtin-associated protein 1 (HAP1) attenuating movement along microtubules (D). coREST, REST co-repressor; HDAC, histone deacetylase; SIN3A, SIN3 homologue A, a transcription regulator. (Modified from (Cattaneo et al. 2005), Nature Reviews: Neuroscience). 20 Review of Published Literature In vitro studies of striatal neurons have shown BDNF to be a potent neurotrophic factor for GABAergic striatal neurons, promoting differentiation, increasing soma size, and increasing neurite branching (Mizuno et al. 1994; Widmer and Hefti 1994; Ventimiglia et al. 1995). BDNF has also been demonstrated in vitro to protect GABAergic striatal medium spiny projection neurons against excitotoxic stress and to inhibit apoptosis in striatal cells engineered to express huntingtin with an expanded CAG repeat sequence (Nakao et al. 1995; Saudou et al. 1998). Despite the promising neuroprotective actions of BDNF observed in vitro, direct infusion of BDNF protein into the rat striatum failed to provide any neuronal protection against QA lesioning (Anderson et al. 1996). Transplantation of BDNF-producing fibroblasts to the corpus callosum failed to supply protection against excitotoxic insult of the striatum (Frim et al. 1993b), however similar transplantation into the striatum prior to QA injection resulted in a reduced lesion size and increased survival of the striatal projection neurons (Perez-Navarro et al. 2000a). Martinez and Bjorklund (1996) using BDNFproducing neural stem cells observed a partial protection of DARPP-32 expressing striatal projection neurons. The use of direct in vivo gene transfer to direct the expression of BDNF in the striatum has also been previously investigated using an Ad-BDNF vector injected intrastriatally prior to QA lesioning (Bemelmans et al. 1999). They reported a 55% reduction in QA lesion size with an increase in the survival of DARPP-32 striatal projection neurons from 46% in control Ad-βGal treated animals to 64% survival in the Ad-BDNF animals. A slight shrinkage of the striatum was attributed to the toxicity of the first-generation adenoviral vector, which possibly restricted transgene expression, thus potentially limiting neuroprotection (Bemelmans et al. 1999). An earlier investigation conducted by my colleagues and I (Kells et al. 2004) showed AAV-BDNF mediated partial protection of striatal projection neurons and NOS expressing striatal interneurons against QA-induced excitotoxicity. The spectrum of neuroprotection observed across the different BDNF delivery procedures highlights the importance of the delivery method in determining the ability for the neurotrophic factors to provide efficient neuronal protection. Together the previous studies suggest that continuous local production of BDNF is vital for this neurotrophic factor to provide neuronal protection, possibly explaining the lack of efficacy for direct BDNF injection (Anderson et al. 1996) and BDNFproducing fibroblast transplanted into the corpus callosum (Frim et al. 1993b). The necessity for high level, local production of BDNF is possibly explained by its limited penetration / diffusion of BDNF through the brain parenchyma or rapid clearance, most likely due to the high density of BDNF receptors in the striatum and glial cell uptake via truncated trkB receptors (Mufson et al. 1999). 21 Review of Published Literature 1.6.3 Glial cell-line derived neurotrophic factor A potent neurotrophic factor for dopaminergic neurons (Lin et al. 1993), GDNF is globally expressed throughout the CNS, with the highest expression occurring in regions receiving dense dopaminergic innervations including the developing striatum, ventral pallidum, olfactory tubercle, and piriform cortex (Bohn 1999). The developing striatum exhibits the highest level of GDNF with mRNA expression peaking at birth before rapidly declining and becoming undetectable by in situ hybridisation after 4 weeks (Stromberg et al. 1993; Choi-Lundberg and Bohn 1995). Sensitive RTPCR assays have however reported that a low level of GDNF mRNA expression is maintained in the adult rat and human striatum (Schaar et al. 1993; Springer et al. 1994; Choi-Lundberg and Bohn 1995), with neuronal expression localised to large cholinergic interneurons (Pochon et al. 1997; Trupp et al. 1997). This expression pattern illustrates the target-derived role of GDNF in directing development of the nigrostriatal dopaminergic connections, with the dopaminergic neurons in the substantia nigra pars compacta (SNc) expressing high GDNF expression in the adult brain providing long-term autocrine / paracrine maintenance (Pochon et al. 1997; Marco et al. 2002). GDNF signalling is mediated by the ret receptor but requires the initial binding to the GDNF specific coreceptor GDNF family receptor α-1 (GFRα1) (Jing et al. 1996; Treanor et al. 1996). Both ret and GFRα1 show similar striatal localisation to GDNF, with low expression in the striatal projection neurons (Marco et al. 2002). However, following excitotoxic lesioning, GDNF and GFRα1 expression is largely localised to astrocytes with the suggestion that reactive astrocytes may present the GFRα1/GDNF complex to the striatal neurons thereby enhancing ret-mediated signalling to promote neuron survival (Trupp et al. 1997; Yu et al. 1998; Marco et al. 2002). The therapeutic effects of GDNF have been extensively investigated in animal models of Parkinson’s disease and amyotrophic lateral sclerosis. GDNF delivery can both prevent the degeneration and promote the regeneration of dopaminergic neurons, and enhance survival of motorneurons (Bohn 1999; Bjorklund et al. 2000; Bohn et al. 2000a; Bohn et al. 2000b). Enhanced survival of motor neurons by GDNF has been shown to be dependent on anti-apoptotic caspase inhibitors ((Perrelet et al. 2002), refer Section 1.7.2). GDNF also shows promise for HD with both direct intraventricular GDNF protein infusions (Araujo and Hilt 1997) and striatal transplantation of GDNF-secreting fibroblasts (Perez-Navarro et al. 1996; Perez-Navarro et al. 1999) partially rescuing striatal medium spiny projection neurons from QA induced cell death. Only striatonigral projection neurons were maintained by the GDNF-producing fibroblasts, with the striatopallidal projection neurons and striatal parvalbumin-positive interneurons not surviving QA lesioning (Perez-Navarro et al. 1996; Perez-Navarro et al. 1999). Araujo and colleagues (1997) reported that in addition to partial 22 Review of Published Literature protection of medium spiny projection neurons, an intraventricular infusion of GDNF protein prior to QA lesioning significantly reduced amphetamine-induced rotational asymmetry and attenuated the numerous neurochemical deficits caused by the QA lesions. They also found that GDNF infusion provides protection against 3-NP induced neuronal cell loss in the striatum and associated locomotion deficits (Araujo and Hilt 1998). More recently, GDNF-derived support of striatal neurons was demonstrated following in vivo AAV-GDNF gene delivery to the striatum prior to 3-NP administration (McBride et al. 2003) and QA lesioning (Kells et al. 2004). McBride and colleagues (2003) reported a 70% increase in surviving NeuN immunopositive striatal neurons accompanied by significant preservation of motor performance following overexpression of GDNF in the striatum. This has been followed up with an investigation in a transgenic mouse model of HD (N171-82Q) where the presymptomatic mice received AAV-GDNF to enhance GDNF expression in the striatum, and showed significant protection of the striatal neurons and attenuated behavioural deficits (McBride et al. 2006). In contrast, LV-GDNF delivery to the R6/2 mouse model of HD failed to protect against striatal cell loss or functional behaviour impairments (Popovic et al. 2005). The conflicting results of these investigations were proposed to have arisen due to the greater severity of the R6/2 model or possibly the need for presymptomatic delivery of the GDNF vectors (McBride et al. 2006). 1.6.4 Ciliary neurotrophic factor Although beyond the focus of this thesis, CNTF has shown considerable promise for treating HD. While not vital for CNS development (Takahashi et al. 1994; DeChiara et al. 1995), CNTF expressed by glia enhances survival and differentiation of neurons in the CNS and PNS (Conover et al. 1993; Helgren et al. 1994; Sleeman et al. 2000). CNTF deficiency is associated with motor neuron degeneration (Masu et al. 1993) and is up-regulated in astrocytes following neuronal injury (Friedman et al. 1992; Ip et al. 1993; Rudge et al. 1995). Therefore CNTF is proposed to play a critical role in maintaining and possibly restoring neuronal function following CNS injury (Rudge et al. 1995). Abundant expression of CNTF receptors in the striatum (Ip et al. 1993; Rudge et al. 1995) suggest striatal neurons may rely on CNTF for survival (Choi 1988; Rudge et al. 1994). In contrast to most other neurotrophic factors, significant protection of GABAergic striatal neurons has been achieved by direct intrastriatal protein infusion of CNTF starting 3-4 days prior to QA injection, with 69 ± 17% maintenance up from 29 ± 11% survival in untreated animals (Anderson et al. 1996). Implantation of CNTF-secreting cells into the lateral ventricle in rat (Emerich et al. 1996; Emerich et al. 1997a) and primate (Emerich et al. 1997b) models of HD have been extensively 23 Review of Published Literature investigated with the continuous supply of CNTF providing significant protection to striatal neurons (Emerich 1999). Behavioural studies undertaken in both unilaterally and bilaterally QA-lesioned rats with CNTF-secreting implants displayed improvement in both locomotor and cognitive functions to the extent that bilaterally lesioned rats with CNTF secreting implants were non-distinguishable from sham-lesioned animals (Emerich et al. 1996; Emerich et al. 1997a). Emerich and colleagues showed additional protection of layer V motor cortex neurons and maintenance of GABAergic innervations of the globus pallidus and substantia nigra pars reticulata using a primate model of HD (Emerich et al. 1997b). Mittoux et al. (2000) reported similar results using encapsulated CNTF-secreting cells implanted bilaterally into the striatum of a primate 3-NP model of HD, with secretion of CNTF beginning at symptom onset providing restoration of cognitive and motor functions associated with persistent neuronal loss or dysfunction (Mittoux et al. 2000). Direct in vivo gene delivery to direct enhanced CNTF expression throughout the striatal parenchyma has also been investigated. LV-CNTF vector delivery prior to QA lesioning significantly prevented neurodegeneration of DARPP-32 striatal projection neurons, cholinergic neurons and NADPHd containing interneurons, significantly attenuating apomorphine-induced rotational behaviour (de Almeida et al. 2001). Assessment of Ad-CNTF gene delivery to the striatum demonstrated retrograde vector transport and anterograde CNTF transport resulting in widespread distribution of CNTF protecting the integrity of the cortical-striatal-pallidal connections from 3-NP induced cell death (Mittoux et al. 2002). 1.7 Apoptosis and Huntington’s Disease Neurodegeneration With the molecular mechanisms underlying HD neurodegeneration not yet fully elucidated, the extent to which apoptotic mechanisms contribute to the selective neuronal cell loss remains to be conclusively determined. While the expression of mutant huntingtin – both in cell culture and in transgenic mice – induces apoptotic cell death (Reddy et al. 1998; Hodgson et al. 1999; Wang et al. 1999; Li et al. 2000; Rigamonti et al. 2000; Jana et al. 2001), the relative lack of clear apoptotic features in HD brains has caused controversy over whether the neurons die by apoptotic or necrotic cell death (Hickey and Chesselet 2003; Sawa et al. 2003). Apoptosis is typically characterised by nuclear and cytoplasmic condensation, DNA fragmentation, membrane blebbing and finally the phagocytosis of membrane-bound apoptotic bodies (Kerr et al. 1972). This programmed cell death commonly occurs in isolated cells requiring the initiation of a cell death pathway to induce the expression and activation of pro-apoptotic proteins including 24 Review of Published Literature numerous proteases (e.g. caspases) and endonucleases (Martin and Green 1995; Sastry and Rao 2000). DNA fragmentation has been commonly used to study apoptosis by in situ TUNEL labelling of DNA fragments (Gavrieli et al. 1992), however random DNA cleavage is often observed in neurotic tissue making apoptotic and necrotic cell death indistinguishable by DNA fragment labelling alone (Charriaut-Marlangue and Ben-Ari 1995). Therefore while TUNEL labelling has been observed in the post-mortem striatum of HD patients (Dragunow et al. 1995; Portera-Cailliau et al. 1995; Thomas et al. 1995; Butterworth et al. 1998), the lack of typical apoptotic DNA laddering (Cohen et al. 1994) suggests these cells may not be undergoing apoptotic cell death (Dragunow et al. 1995; Portera-Cailliau et al. 1995). More recently the implication of apoptotic mechanisms contributing to HD has been indicated by an increase in the expression of pro-apoptotic proteins in post-mortem HD brains, including caspase-9, Bax, cytochrome c and poly(ADP-ribose) polymerase (Kiechle et al. 2002; Vis et al. 2005). Vis and colleagues (2005) suggested that the difficulties which have been encountered in positively identifying neurons exhibiting typical apoptotic morphology are due to the slow progressive nature of HD, meaning that only a few cells are likely to be undergoing cell death at any one time. Apoptosis of striatal neurons has been found to occur in the excitotoxic QA lesion model of HD (Dure et al. 1995; Portera-Cailliau et al. 1995; Hughes et al. 1996; Qin et al. 1996; Bordelon et al. 1999; Nakai et al. 1999), and also in full-length transgenic animal models which display neuronal cell loss (Reddy et al. 1998; Hodgson et al. 1999). Following QA injection into the rat striatum both apoptotic and necrotic cell death is observed to occur, with the apoptotic cells observed mainly in the dorsomedial striatum (Bordelon et al. 1999). Portera-Cailliau and colleagues (1995) found typical apoptotic DNA laddering in the hours following QA lesioning but not at later time points suggesting that both apoptotic and necrotic mechanisms may contribute towards the QA induced cell death process. Investigations with genetic models of HD, including the YAC transgenic mice (full-length huntingtin with 46, 72 or 128 poly-Q repeats; (Hodgson et al. 1999)), have demonstrated greater vulnerability to excitotoxic death mediated by an enhancement of NMDA receptor activity resulting in increased intracellular calcium levels (Levine et al. 1999; Cepeda et al. 2001; Laforet et al. 2001; Zeron et al. 2002). More specifically the poly-Q huntingtin protein induces potentiation of the NR2B receptor subunit which is predominantly expressed by the selectively vulnerable striatal projection neurons (Landwehrmeyer et al. 1995; Testa et al. 1995; Chen et al. 1999; Sun et al. 2001; Zeron et al. 2001; Zeron et al. 2002). Increased calcium levels have been well established to induce apoptosis via the intrinsic, mitochondrial regulated apoptotic pathway (Figure 1-5; (Hajnoczky et al. 2003; Orrenius et al. 2003)). 25 Review of Published Literature Molecular mechanisms responsible for triggering apoptosis are dependant on the specific apoptotic stimuli with different cell death pathways being activated by different insults, although most ultimately lead to the activation of an “executioner” caspase (Adams 2003). The two most well understood pathways are the extrinsic death-receptor pathway and the intrinsic apoptotic pathway. The extrinsic pathway requires activation of death receptors, such as tissue necrosis factor or Fas, which in turn activate “initiator” caspases (caspase-8 and -10), subsequently leading to the activation of the executioner caspases-3 and / or -7 (Ashkenazi and Dixit 1998). Intrinsic apoptosis is initiated Figure 1-5 Potential involvement of Ca2+ signalling in the induction of apoptosis in HD Excessive cytosolic Ca2+ can lead to mitochondrial Ca2+ overload and the release of apoptogenic factors, including cytochrome c, inducing intrinsic apoptosis. Striatal neurons abundantly express NDMA receptors (NMDAR) and mGluR5 which when stimulated by corticostriatal glutamate (Glu) release cause an increase in cytosolic Ca2+ – NMDAR directly through allowing Ca2+ entry, and mGluR5 via inositol 1,4,5-trisphosphate (InsP3) signalled release of Ca2+ from intracellular storage organelles. In HD, expanded huntingtin (Httexp) disrupts Ca2+ signalling through potentiation of the 2B subunit of NMDAR, sensitisation of InsP3 receptors (InsP3R1), and destabilisation of mitochondrial handling. Therefore normal glutamate stimulation of striatal neurons can result in supranormal Ca2+ responses contributing to the vulnerability of medium spiny neurons (MSN) to HD induced degeneration. Although not directly targeted at the disrupted Ca2+ signalling, anti-apoptotic factors may potentially attenuate the induction of apoptosis of striatal neurons through maintaining mitochondrial membrane integrity – Bcl-xL – or inhibiting caspase activation – XIAP. Mito, mitochondria; MCU, mitochondrial Ca2+ uniporter. (Modified from (Tang et al. 2005), PNAS). 26 Review of Published Literature internally by cellular stress causing increased free calcium uptake into the mitochondria and opening of the mitochondrial permeability transition pores allowing the release of pro-apoptotic factors, including cytochrome c, from the mitochondria (Figure 1-5 and 1-5; (Hajnoczky et al. 2003; Orrenius et al. 2003)). Pro-apoptotic Bcl-2 family proteins such as Bax and Bak also appear to be critical for inducing apoptosis through recruitment to the outer mitochondrial membrane inducing the release of apoptotic factors, although there is still considerable question over how this occurs (Figure 1-6; (Gross et al. 1999; Martinou and Green 2001; Newmeyer and Ferguson-Miller 2003)). Cytochrome c is a co-factor for apoptotic protease activating factor-1 which can bind procaspase-9 in the presence of cytochrome c allowing the formation of large apoptosomes and the activation of bound caspase-9 leading to the subsequent activation of caspases-3 and -7 (Zimmermann et al. 2001). Additional death-promoting factors released from the mitochondria include Smac/DIABLO Figure 1-6 Mitochondrial permeabilisation in intrinsic apoptosis In healthy cells the integrity of the mitochondrial outer membrane is maintained by anti-apoptotic Bcl-2-like proteins (Bcl-2) with caspases in the cytosol present as inactive pro-proteins. Apoptotic signalling causes release of sequested BH3-only proteins to interact with Bcl-2, allowing the recruitment of pro-apoptotic Bcl-2 family proteins (Bax, Bak) to the mitochondrial membrane facilitating release of apoptogenic proteins. Cytochrome c induces binding of Apaf-1 and procaspase-9 to form apoptosomes activating procaspase-9 (c9) and subsequently caspase-3 (c3). Cytosolic IAP are inhibited and degraded by the release of Smac/DIABLO (Diablo) and HtrA2/OMI (Omi) proteins, while further apoptogenic proteins (apoptosis inducing factor (AIF), endonuclease G (endoG)) can enter the nucleus inducing DNA degradation. (Reproduced from (Adams 2003), Genes and Development) 27 Review of Published Literature and HtrA2/OMI proteins that bind and degrade inhibitor of apoptosis proteins (IAP), preventing caspase sequestration and inhibition (Figure 1-6; (Srinivasula et al. 2003; Saelens et al. 2004)). In addition to the potentiation of NMDA receptor signalling, the poly-Q huntingtin protein has been shown to directly impair mitochondrial function (Figure 1-5; (Sawa et al. 1999; Panov et al. 2002; Choo et al. 2004)) and initiate cytotoxic mechanisms following N-terminal caspase cleavage (Wellington et al. 2002) including transcriptional dysregulation, protesome inhibition and the formation of aggregates and neuronal intranuclear inclusions (Figure 1-1; (Sawa et al. 2003)). Rigamonti and colleagues have shown that wild-type huntingtin has pro-survival properties that protect neurons against apoptotic cell death at the level of procaspase-9 processing (Rigamonti et al. 2000; Rigamonti et al. 2001), suggesting that the expanded poly-Q tract not only has toxic “gain-offunction” consequences, but normally occurring anti-apoptotic functions are also impaired. A recent investigation found enhanced expression of full-length wild-type huntingtin provided neuroprotection against QA-induced excitotoxicity with reduced activation of caspase-3 (Leavitt et al. 2006). Activation of caspase-3 has been shown to be induced by mutant N-terminal huntingtin fragments suggesting a destructive feedback loop maybe initiated resulting in extensive activation of caspase-3 (Wellington et al. 2002; Gafni et al. 2004). Induced activation of caspase-3 in HD suggests that whether or not the striatal projection neurons die strictly by apoptosis, apoptotic mechanisms are likely to underlie the neurodegeneration by stressing normal cellular functions and increasing the neurons sensitivity to other potentially neurotoxic insults. Therefore an enhancement of endogenous anti-apoptotic factors or administration of apoptotic inhibitors could conceivably be of therapeutic benefit by preventing the caspase-mediated cleavage of huntingtin, thereby reducing the production of toxic N-terminal fragments and maintaining pro-survival function, or via general augmentation of the cells ability to withstand cellular stress. With the pathogenesis of HD appearing to involve disruptions to numerous cellular processes involved in the induction of apoptotic related neurodegeneration, a number of targets have been identified for potential therapeutic intervention including NMDA receptor-mediated excitotoxicity, mitochondrial dysfunction, transcriptional dysregulation, and apoptotic executing caspases. A number of pharmaceuticals have shown efficacy in attenuating HD neuronal death in preclinical trials, with some clinical trials currently being conducted (Leegwater-Kim and Cha 2004; Handley et al. 2006). However a less studied approach is the use of gene delivery to enhance the expression of naturally occurring anti-apoptotic factors that may counteract pro-apoptotic mechanisms while additionally increasing the capacity of the neurons to handle neurotoxic insult (Mochizuki et al. 2002). 28 Review of Published Literature 1.7.1 Anti-apoptotic Bcl-2 and Bcl-xL proteins Bcl-2 family proteins are involved in regulating the release of mitochondrial pro-apoptotic factors, including cytochrome c, which is a key event in the initiation of the intrinsic apoptotic pathway (Figure 1-6; (Cory and Adams 2002)). There are three groups of Bcl-2-like proteins: anti-apoptotic proteins (e.g. Bcl-2, Bcl-xL), pro-apoptotic proteins (e.g. Bax, Bak) and BH3-only proteins (e.g. Bid, Bad). Anti-apoptotic Bcl-2 and Bcl-xL are integral membrane proteins located on the mitochondrial, endoplasmic reticulum and nuclear membranes that are thought to preserve the permeability of the membranes against the potential pore-forming pro-apoptotic proteins, although the actual molecular mechanisms remain controversial (Martinou and Green 2001; Kuwana et al. 2002; Newmeyer and Ferguson-Miller 2003). The pro-apoptotic proteins appear to be recruited to the membranes following a cellular apoptotic stimulus to facilitate the release of pro-apoptotic factors (Figure 1-6). However in order for apoptosis to occur, the inhibitory effects of the anti-apoptotic proteins need to be overcome; this is thought to involve interactions with BH3-only proteins (Huang and Strasser 2000; Gross 2001). Over-expression of Bcl-2 or Bcl-xL can prevent apoptosis occurring following a variety of cytotoxic stimuli including neurotrophic deprivation, ionizing radiation and oxidative stress. There does not appear to be a significant difference in the protective effects of Bcl-2 and BclxL, however endogenous expression of Bcl-2 is seen to peak during embryonic development while Bcl-xL expression continues to increase postnatally, reaching peak expression in the adult CNS (Gonzalez-Garcia et al. 1994; Gonzalez-Garcia et al. 1995). The therapeutic potential for using Bcl-2 or Bcl-xL in neurodegenerative diseases has been shown by a few studies using viral vectors to cause overexpression in the CNS. HSV vector delivery of Bcl-2 to the substantia nigra prior to 6-hydroxydopamine lesioning doubled the survival rate of dopaminergic neurons (Yamada et al. 1999). Similarly AAV-mediated overexpression of Bcl-2 in the CA1 hippocampal neurons attenuated cell death and reduced DNA fragmentation following ischemia even when AAV delivery was delayed until one hour after the ischemic insult (Shimazaki et al. 2000). Motor neuron survival in a transgenic mouse model of amyotrophic lateral sclerosis was also increased by AAV mediated delivery of Bcl-2 which consequently delayed the onset of muscular deficits (Azzouz et al. 2000). The LV vector mediated overexpression of Bcl-xL in septal cholinergic neurons was highly efficient in preventing axotomised cell death of transduced neurons (Blomer et al. 1998), while AAV-Bcl-xL retrogradely delivered to entorhinal projection neurons also protected against axotomy-induced apoptosis (Kaspar et al. 2002). A limitation of anti-apoptotic factor mediated protection is that only the individual transduced cells that are over-expressing the anti-apoptotic protein can be expected to show increased resistance to an apoptotic stimulus in 29 Review of Published Literature contrast to a secreted neuroprotective molecule that can have paracrine activity and therefore provide assistance to neighbouring cells. The simultaneous delivery of a neurotrophic factor and an antiapoptotic factor as investigated by Natsume and colleagues (2001) using HSV vector delivery of both Bcl-2 and GDNF to the substantia nigra ensured that secreted GDNF may at least provide some protective support to those neurons not expressing the anti-apoptotic factor. Co-injection of HSV vectors encoding Bcl-2 and GDNF resulted in the additive survival rate of dopaminergic neurons following 6-hydroxydopamine induced degeneration (Natsume et al. 2001). More recently neuroprotection against glutamate excitotoxicity has been reported following AAVBcl-xL transduction of neuroblastoma cells and primary motor neurons as a model of amyotrophic lateral sclerosis (Garrity-Moses et al. 2005). Similarly, LV-Bcl-2 delivery to the CA1 region of the hippocampus attenuated neuronal cell death following an excitotoxic NMDA injection (Wong et al. 2005). While these studies have indicated Bcl-2 / Bcl-xL is capable of imparting neuroprotection against excitotoxic insult, an earlier investigation of QA-induced cell death in the striatum of transgenic mice over-expressing Bcl-2 reported no attenuation of the loss of DARPP-32 expressing striatal projection neurons (Maciel et al. 2003). 1.7.2 Inhibitors of apoptosis With caspases playing a vital role in the initiation and physical execution of apoptosis, they present a reasonably specific target for inhibiting apoptosis independent of the apoptotic pathway that is initiated. The function of inhibiting caspases is carried out in healthy cells by a family of proteins known as inhibitors of apoptosis (IAP) (Robertson et al. 2000). Six members of this family have been identified in humans: Neuronal apoptosis inhibitor protein (NAIP), X-chromosome-linked IAP (XIAP), Human IAP-1 (HIAP-1), HIAP-2, Survivin and Bruce (Deveraux and Reed 1999). The neuroprotective properties of these IAPs have been investigated with the most potent caspase inhibitor proving to be XIAP (Simons et al. 1999) which has two sites capable of inhibiting caspase activity – one site that is specific for caspase-3 and -7, and the other a specific inhibitor of caspase-9 (Deveraux et al. 1999). By inhibiting the initiator caspase-9, XIAP is capable of inhibiting both the activation and the activity of the executioner caspases-3 and -7. Robertson and colleagues have reported that NAIP is up-regulated in cholinergic striatal interneurons following ischemic insult, suggesting that the neuronal resistance to ischemia and excitotoxicity may be related to the inhibition of caspases (Xu et al. 1997). Ad-vector mediated delivery of NAIP prior to 6-OHDA lesioning provided protection to nigrostriatal dopaminergic neurons, attenuating the amphetamine-induced motor deficits (Crocker et al. 2001). Over-expression of NAIP (Xu et al. 30 Review of Published Literature 1997) or XIAP (Xu et al. 1999) in hippocampal CA1 neurons prevented ischemic-induced apoptosis by inhibiting caspase-3 activation. The Ad-XIAP delivery to CA1 neurons increased survival 5-6 fold and attenuated the loss of spatial memory as measured in the Morris water maze (Xu et al. 1999), while delivery to retinal ganglion cells after axotomy of the optic nerve prevented any secondary apoptotic cell death (Kugler et al. 2000). More recently a reduction in DNA methylationinduced apoptosis in photoreceptors has been shown following AAV-XIAP transduction (Petrin et al. 2003a; Petrin et al. 2003b). Perrelet and colleagues have similarly shown that the adenoviral mediated overexpression of NAIP, HIAP-1 or HIAP-2 are each capable of attenuating the degeneration of motor neurons following sciatic nerve axotomy (Perrelet et al. 2000). In addition they have demonstrated by selective antisense inhibition of IAPs the absolute requirement of XIAP and NAIP to mediate the neuroprotective effects of GDNF, but not BDNF or CNTF (Perrelet et al. 2002). Only GDNF treatment upregulated XIAP and NAIP expression showing the stimulation of anti-apoptotic pathways in GDNF mediated neuroprotection (Perrelet et al. 2002). Ad-mediated over-expression of XIAP and GDNF have also been shown to act synergistically to protect nigrostriatal dopaminergic neurons against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced cell death and maintain striatal dopamine levels (Eberhardt et al. 2000). While XIAP alone was sufficient to rescue the nigrostriatal neurons from cell death, the dopaminergic striatal terminals degenerated in the absence of GDNF (Eberhardt et al. 2000). Recent evidence that XIAP and HIAP-1 undergo degradation in the HD caudate, following aberrant release of pro-apoptotic mitochondrial proteins Smac/DIABLO and HtrA2/OMI, further suggests upregulation of IAP may be therapeutically beneficial for HD (Goffredo et al. 2005). 1.8 Summary The growing understanding of pathogenic processes initiated in HD by an expanded poly-Q tract in huntingtin, has provided an opportunity to develop therapeutic intervention tailored towards either the potentiation of an impaired cellular mechanism, or conversely the attenuation of acquired cytotoxic functions. With a large pre-symptomatic window-of-opportunity available through genetic testing for HD in “at-risk” individuals, preventative therapeutic intervention instigated prior to the initiation of neuronal dysfunction leading to selective neurodegeneration is a serious proposition for HD patients. Excitotoxicity appears to play a significant role in the ultimate death of the vulnerable striatal neurons with mutant huntingtin causing a reduction in vital neurotrophic support through interruption of BDNF transcription and transportation, and interference with the molecular 31 Review of Published Literature machinery of programmed cell death resulting in impaired ability to handle the naturally high corticostriatal glutamate stimulation. Figure 1-7 Model of neurotrophic factor and anti-apoptotic factor supplied protection. AAV-mediated gene delivery to targeted striatal neurons and possibly adjacent glial cells results in stable biotherapeutic protein expression within the striatum. Non-secreted anti-apoptotic factors – Bcl-xL and XIAP – remain contained within the transduced cells acting in the cytosol and at the mitochondrial membrane to inhibit apoptotic mechanisms. Only directly transduced neurons are exposed to enhanced anti-apoptotic factor expression. Neurotrophic factors – BDNF and GDNF – are secreted following production within transduced cells providing autocrine and paracrine actions on the target striatal neurons through cell surface receptors and therefore providing trophic support to a greater number of neurons than actually transduced. The transduction of cells in the projection nuclei may additionally provide target-derived trophic support through retrograde transportation of the neurotrophic factors. (Modified from (Blomer et al. 1998), PNAS) From current research it appears that while neurotrophic factors and anti-apoptotic factors hold real potential as therapeutic agents, the need for a continuous supply across the entire striatum makes the mechanism of delivery vitally important for the success of these factors as effective therapeutic agents. Cellular transplantation and direct in vivo gene delivery techniques appear to be the most promising delivery methods for neurotrophic factors, as physiologically relevant concentrations can be continuously produced locally within the striatum directly supplying support to surrounding striatal neurons, while limiting any adverse effects resultant of activity in non-targeted regions. Nonsecreted biotherapeutics, such as anti-apoptotic factors, require very efficient delivery techniques, generally requiring in vivo gene delivery vectors, as only cells directly expressing / receiving the 32 Review of Published Literature therapeutic agent will benefit from any effects. Therefore further research and development of effective gene delivery techniques are vital for pursuing the true potential of targeted biotherapeutics for attenuating the insidious progression of HD. 1.9 Thesis Objectives This thesis set out to investigate if continuous neurotrophic factor or anti-apoptotic factor expression achieved via in vivo gene delivery can attenuate neuronal cell loss and behavioural deficits in the excitotoxic QA lesion animal model of HD. Although neurotrophic factors have been previously demonstrated to partially prevent the death of striatal projection neurons, difficulties in achieving efficient delivery of neurotrophic factors to the striatum in vivo may limit the true potential that neurotrophic factors hold for counteracting the neurodegenerative process of HD. Ongoing improvement in non-toxic, non-immunogenic viral based vectors has allowed for the efficient delivery of therapeutic proteins to the CNS following in vivo gene transfer. In vivo gene delivery is suited to the continuous production of therapeutic proteins within a specific population of cells such as the neurons of the striatum, and this local production of neurotrophic factors at physiologically effective concentrations across the entire striatum may prove to be therapeutically beneficial at preventing or slowing the progression of HD. Construction of recombinant AAV vectors and the verification of functional transgenic protein expression is detailed in Chapter 2 (Kells et al. 2006). With a large number of neuroprotective compounds currently under investigation following the ongoing elucidation of molecular mechanisms underlying HD, it has been recently suggested that preclinical studies of pharmaceutical and biotherapeutic agents for HD should be proven efficacious in transgenic models of HD, by two independent research groups, prior to moving forward into nonhuman primate and clinical trials (Bates and Hockly 2003). However the increasing evidence that excitotoxic mechanisms contribute to HD neurodegeneration ensure that the QA model is still a relevant research tool to investigate neuroprotective agents. Chapter 3 covers the optimisation and characterisation of a unilateral QA lesion rat model of HD. Previous in vivo gene delivery mediated expression of neurotrophic factors in the striatum has demonstrated the protection of striatal neurons by BDNF, GDNF and CNTF (Bemelmans et al. 1999; de Almeida et al. 2001; Mittoux et al. 2002; McBride et al. 2003; Kells et al. 2004). While neurotrophic factors show promise in these studies, there is a need for further investigation of more efficient gene delivery vectors, assessment of functional behavioural protection and determination of 33 Review of Published Literature neuronal fibre protection in order to fully evaluate the most promising approaches to advance through to primate studies and clinical trials. Preventative therapeutic delivery of BDNF or GDNF prior to QA striatal lesioning is investigated in Chapter 5. Targeted delivery of anti-apoptotic factors to the striatum in an attempt to prevent striatal cell death has not previously been investigated as a potential treatment for HD, although in vivo gene delivery of Bcl-2-like proteins (Bcl-2 and Bcl-xL) and IAPs (XIAP and NAIP) have provided protection to other neurons following apoptotic stimulating insult. With apoptotic mechanisms being induced in HD, anti-apoptotic factor expression may hold therapeutic potential to enhance the survival of striatal neurons. In Chapter 6 I investigate if enhanced Bcl-xL or XIAP expression can attenuate QAinduced excitotoxic neuronal cell death. Ultimately this thesis research aimed to investigate whether the use of chimeric AAV1/2 vectors for the in vivo gene delivery of the neurotrophic factors BDNF or GDNF, or the anti-apoptotic factors Bcl-xL or XIAP could efficiently generate continuous production within the rat striatum at a level that is sufficient to maintain the functional integrity of the striatal neurons in the face of an acute excitotoxic insult. This was achieved by assessment of neuropathological changes and functional behavioural impairments. 34 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Chapter 2 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing *† 2.1 Overview AAV derived recombinant vectors have become the vector of choice for many investigators studying potential clinical applications of gene delivery primarily due to their efficient stable transduction of post-mitotic cells and non-pathogenic viral origin (Peel and Klein 2000). To mediate gene transfer to striatal neurons I produced high-titre recombinant AAV vectors containing cDNA sequences for each of the potential biotherapeutic proteins. It is clearly essential for gene delivery applications that the protein products expressed following gene transfer into the target cells are correctly translated and processed into functionally active molecules capable of influencing cellular activity. Therefore it was important that I developed procedures to assess the functional impact of each transgenic protein following rAAV vector transduction. Given the well characterised functional activity of the neurotrophic factors and anti-apoptotic proteins under investigation, I devised efficient in vitro assays to demonstrate the expected functional impact of each protein. 2.2 AAV Vector Development and Production Procedures Production of recombinant AAV vectors was undertaken in vitro by co-transfection into HEK293 cells of a plasmid containing my AAV vector expression cassette along with helper plasmids encoding AAV rep and cap proteins and adenoviral genes essential for AAV production. The use of separate helper plasmids supplying the necessary AAV replication and packaging proteins in trans ensures the production of replication deficient vectors with only the expression cassette DNA flanked by AAV specific inverted terminal repeat (ITR) sequences being packaged into the AAV particles. * The functional assessment of AAV vectors has been published in Journal of Neuroscience Methods Kells, A.P., Henry, R.A., Hughes, S.M. and Connor, B. (2006). Verification of functional AAV-mediated neurotrophic and anti-apoptotic factor expression. Journal of Neuroscience Methods: doi:10.1016/j.jneumeth.2006.11.006. Note: Production of the AAV-BDNF plasmid construct was kindly undertaken by Rebecca Henry, Department of Pharmacology, The University of Auckland. Functional testing of the AAV-BDNF vector was performed in conjunction with Rebecca Henry. 35 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Replication deficient viral particles were subsequently harvested from the HEK293 cells, purified by heparin affinity column and centrifuge concentrated to provide high titre stocks for in vivo gene delivery. Development of rAAV vectors for the neuroprotective gene delivery investigations involved cloning the therapeutic genes-of-interest into a previously constructed AAV expression cassette (Klugmann et al. 2005), packaging and purifying the AAV vector particles, testing for efficient AAV transduction and assaying for functional transgenic protein expression in vitro. 2.2.1 Molecular biology protocols 2.2.1.1 PCR Cloning To facilitate ligation of the genes-of-interest into AAV expression cassettes I PCR amplified the cDNA sequences from source plasmids (Table 2-1) using customised primer pairs (Table 2-2) incorporating unique restriction sites to flank the cDNA sequences and complement the restriction sites at the insertion site of the AAV backbone plasmid. Direct replacement of the cDNA stop codons with either an EcoRI or BglII restriction site sequence allowed in-frame ligation with the downstream HA-tag sequence. The PCR program to clone each gene-of-interest was tested using Red Hot® DNA polymerase (ABgene) to optimise the primer annealing temperature and cycle number required to produce a single clean DNA product. Each reaction was setup with 2.5µL Red Hot® PCR Reaction buffer (ABgene), 2.0µL MgCl2 (25mM; ABgene), 1.0µL dNTPs (2.5mM; Invitrogen), 0.2µL forward primer (10µM; Invitrogen), 0.2µL reverse primer (10µM; Invitrogen), 1.0µL miniprep DNA plasmid template, 0.25µL Red Hot® DNA Polymerase (ABgene) and made up to 25.0µL with sterile dH2O. Reactions were run in a GeneAmp® PCR System 9700 (Applied Biosystems) thermal cycler programmed for: Initial denaturing Denaturing Annealing Elongation Final Elongation Hold 95°C 5 min 1 cycle 30 sec 30 sec 60 sec 35 cycles 72°C 10 min 1 cycle 4°C ∞_- 95°C 56 / 60 / 65°C 72°C PCR products were visualised on a 1% agarose gel against a 1kb Plus DNA ladder (Invitrogen). 36 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Following optimisation of the PCR program with Red Hot® DNA polymerase a larger scale PCR was performed using an Expand High Fidelity PCR System (Roche). Each PCR was setup with 5.0µL Expand High Fidelity Buffer containing 15mM MgCl2 (Roche), 4.0µL dNTPs (2.5mM; Invitrogen), 1.5µL forward primer (10µM; Invitrogen), 1.5µL reverse primer (10µM; Invitrogen), 1.0µL miniprep DNA plasmid template, 1.0µL Expand High Fidelity Enzyme mix (Roche) and made up to 50.0µL with sterile dH2O. PCR program: Initial denaturing 94°C 2 min Denaturing Annealing Elongation 94°C 62°C 72°C 15 sec 60 sec 90 sec Final Elongation Hold 72°C 7 min 4°C ∞±- 1 cycle 30–35 cycles 1 cycle PCR products were visually checked by running a sample on a 1% agarose gel and then separated out by gel extraction or with a Wizard® SV Gel and PCR Clean-Up System kit (Promega). 2.2.1.2 Agarose Gel Extraction The extraction of DNA fragments from agarose gel was performed to isolate a specific length DNA product following PCR or restriction digests. Reaction mixtures were run on a 1% low melting point agarose gel in TAE buffer. A single band containing the DNA product-of-interest was excised, transferred to a Costar Spin-X column (Corning) and frozen at -80°C for 5mins. The DNA was eluted through the column by centrifugation at 12,000rpm for 10mins (Heraeus Biofuge Stratos). Spin-X column was discarded and 600µL phenol:chloroform:isoamyl alcohol mixture (SigmaAldrich) added to separate out DNA by further centrifugation at 12,000rpm for 10mins. The top layer containing my DNA product was transferred to a new centrifuge tube and made up to 150µL with NaCl solution to give a final concentration of 200nM NaCl. DNA was precipitated and pelleted by adding 375µL 95% ethanol, centrifuging at 12,000rpm for 2min and carefully discarding supernatant. The DNA was washed with 70% ethanol and dried at room temperature before resuspending in 20µL sterile dH2O and storing at -20°C. The Wizard® SV Gel and PCR Clean-Up System (Promega) was also used as an alterative to the Costar Spin-X columns for agarose gel extraction or for direct PCR product purification. Briefly the gel band or PCR mixture was incubated with a Membrane Binding Solution and loaded onto a SV Minicolumn by centrifugation. The DNA bound to the SV Minicolumn was washed with an ethanol based Membrane Wash Solution and then eluted with 50µL sterile dH2O. 37 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing 2.2.1.3 p-GEM®-T Easy Vector PCR products encoding single cDNA gene sequences flanked by unique restriction sites were ligated into pGEM®-T Easy TA cloning vectors (Promega) as per the suppliers’ instructions. Ligations were setup with 5.0µL Rapid Ligation Buffer, 1.0µL pGEM®-T Easy vector, 3.0µL PCR product, 1U T4 DNA ligase and incubated overnight at 4°C. Overnight ligation mixture was used to heatshock transform Escherichia coli (E. coli) competent cells (Section 2.2.1.4). Insertional disruption of the LacZ gene spanning the pGEM®-T Easy vector ligation site provides ready identification of colonies containing an insert by their white appearance when grown on LB agar (USB) plates containing ampicillin (amp; 50 µg/mL; Sigma-Aldrich) and X-gal (20 mg/mL; Sigma-Aldrich). A single transformed colony containing the correct insert was selected by colony PCR screening and used to inoculate overnight cultures for plasmid mini prep and for long-term storage as a 30% glycerol stock. 2.2.1.4 Heatshock Transformation Transformation of E. coli cells was performed by mixing the plasmids with competent cells and providing a heatshock stimulus to induce plasmid uptake. A vial of DH5α E. coli Max Efficiency Competent cells (500µL; Invitrogen) was thawed on ice from -80°C and gently mixed with 0.1µL plasmid mini-prep. After 30mins incubation on ice the cells were heatshocked at 42°C for 60secs and placed back on ice for 1-2mins before transferring to a centrifuge tube with 450µL LB and incubating at 37°C for 1hr. The cells were then plated overnight at 37°C on LB agar containing ampicillin, with or without X-gal, to select for transformed E. coli colonies. 2.2.1.5 Colony PCR Colony PCRs were used to screen E. coli colonies following heatshock transformation for the presence of a plasmid containing a cDNA sequence of the specific gene-of-interest. Individual colonies were picked off an LB agar / amp plate with a sterile tip, streaked gently across a fresh LB agar / amp plate and then suspended in 20µL LB. Using 2.5µL of the resuspended culture for the DNA plasmid template I performed PCRs to amplify the gene-of-interest using the Red Hot® DNA polymerase protocol as described in Section 2.2.1.1. 2.2.1.6 Restriction Enzyme Digestions Complementary restriction digests were performed in preparation for transferring the genes-ofinterest from the pGEM®-T Easy vectors into the AAV expression cassette. Digests were setup in 1.5mL eppendorf tubes using 2.0µL DNA plasmid prep, 2.0µL NEB Buffer (Table 2-4), 2.0µL BSA (NEB), 20U of each restriction endonuclease (Table 2-4), made up to 20µL with sterile dH2O and incubated at 37°C for 2-3hrs. Following restriction digestion the desired DNA fragment was gel 38 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing extracted on a low melting point 1% agarose gel using the Wizard® SV Gel and PCR Clean-Up System. 2.2.1.7 Ligation into the AAV Expression Cassette Ligation of the gene-of-interest into the AAV expression cassette was facilitated by complimentary ssDNA “sticky” ends on the AAV vector and DNA inserts generated by the restriction digests. To restrict self ligation of the AAV backbone vector it was treated with calf intestinal phosphatase (CIP) to remove the 5’ phosphate groups. Following restriction digest of the AAV vector the restriction enzymes were inactivated by incubation at 65°C for 20mins, 1.0µL of CIP was added to the digestion reaction mix and incubated at 37°C for 30min. CIP was inactivated by incubation at 75°C for 30mins. Ligation reactions were setup with 1.0µL CIP treated AAV expression cassette digest, 5.0µL insertion DNA, 1.0µL T4 ligase buffer (Invitrogen), 2.0µL dH2O, 1.0µL T4 ligase enzyme (Invitrogen) and incubated overnight at 4°C. 2.2.1.8 Plasmid Amplification and Purification DNA plasmid mini-prep Small-scale plasmid purifications were undertaken with a High Pure Plasmid Isolation kit (Roche). E.coli cells containing the plasmid-of-interest were pelleted from a 3-4mL overnight LB / amp culture by centrifugation at 9,000rpm for 30secs. Cells were resuspended in 250µL Suspension Buffer containing RNase and lysed by the addition of 250µL Lysis Buffer. Chromosomal DNA and cellular debris was precipitated by adding chilled Binding Buffer and separated by centrifugation at 13,000rpm for 10min. Supernatant containing the DNA plasmid was transferred to a High Pure filter tube and centrifuged at 13,000rpm for 1min. The bound plasmids were washed twice by centrifugation with Wash Buffers before eluting the plasmid DNA with 100µL Elution Buffer (10mM Tris-HCl). Samples of the mini-preps were run on 1% agarose gel to check plasmid yield. DNA plasmid preps were stored at -20°C. DNA plasmid maxi-prep Large-scale plasmid purifications were performed using a QIAGEN® Plasmid Maxi Kit. An E.coli culture containing the plasmid to be amplified was used to inoculate 500mL LB / amp and incubated overnight at 37°C with vigorous shaking. The overnight culture was transferred to 250mL centrifuge bottles and the E.coli cells harvested by centrifugation at 4,300rpm for 15mins (Sorvael RT7Plus). The supernatant was discarded and the E. coli cells resuspended in buffer solution containing RNase (Buffer P1: 50mM Tris-Cl, 10mM EDTA, 100µg/mL RNase A, pH 8.0). Cells were lysed by 39 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing addition of lysis buffer (Buffer P2: 200mM NaOH, 1% SDS) and the cellular debris precipitated by neutralization buffer (Buffer P3: 3.0M potassium acetate). Following centrifugation the plasmid containing supernatant was transferred to an equilibrated QIAGEN-tip. The QIAGEN-tip was washed (Buffer QC: 1.0M NaCl, 50mM MOPS, 15% isopropanol, pH 7.0) and the plasmid eluted (Buffer QF: 1.25M NaCl, 50mM Tris-Cl, 15% isopropanol, pH 8.5). DNA was precipitated with isopropanol and pelleted by centrifugation at 11,000rpm for 30mins. The pelleted DNA was washed with 70% ethanol and air-dried before reconstituting in Tris-EDTA (TE) buffer and stored at -20°C. The plasmid concentration was measured by DNA spectrophotometry (Ultrospec™ 2100, Biochrom). 2.2.1.9 DNA Sequencing All DNA sequencing was performed on a 3100 capillary sequencer (Applied Biosystems) by the Genomics Unit, Centre for Genomics and Proteomics, School of Biological Sciences, The University of Auckland. 2.2.2 AAV plasmid construction DNA plasmids containing an empty AAV expression cassette, AAV rep and cap genes and the Ad helper genes required for AAV packaging were made available by Professor Matthew J. During and Dr Deborah Young, Department of Molecular Medicine and Pathology, FMHS, The University of Auckland (Table 2-1, Appendix A.2). The AAV backbone plasmid (Figure 2-1) contained an AAV expression cassette regulated by a strong constitutively active chicken-β-actin (CBA) / cytomegalovirus (CMV) hybrid enhancer-promoter to drive continuous stable expression in all transduced cells (Xu et al. 2001), a woodchuck post-transcriptional regulatory element (WPRE) to further enhance the transgene expression level (Loeb et al. 1999; Paterna et al. 2000) and a bovine growth hormone poly-A (bGH poly-A) tail sequence. Inverted terminal repeat (ITR) sequences from AAV serotype-2 flanked the expression cassette to direct ssDNA packaging into the vector particles. The multiple cloning site for transgene insertion was directly adjacent to a nine amino acid HA sequence that allowed in-frame fusion of an HA epitope-tag to the 3’-end of the transgene by direct replacement of the stop codon with either an EcoRI or BglII restriction enzyme recognition site. Incorporation of the short HA-tag at the C-terminal allowed for unequivocal immunocytochemical staining of the transgenic protein expression, distinct from endogenous neurotrophic factor or antiapoptotic factor expression. A fully constructed AAV-Luciferase plasmid containing a Firefly Luciferase cDNA sequence with the same regulatory elements, but without an HA-tag, was also supplied by MJ During and D Young to be used as a control for AAV transduction and transgenic protein expression (Appendix A.2.2). 40 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Plasmid Construct Supplier BDNF pAM/CRE-BDNF-WPRE-bGHpA During GDNF AAV/NSE-GDNF-WPRE-bGHpA During Bcl-xL pcDNA3-HA-Bcl-xL Science Reagents XIAP pCI-neo-Flag-XIAP Science Reagents Empty AAV backbone pAM/CAG-C-termHA-WPRE-bGHpA During AAV-Luciferase pAM/CBA-Luciferase-WPRE-bGHpA During AAV-2 rep and AAV-2 cap pRV1 During AAV-2 rep and AAV-1 cap pH21 During Adenovirus E2A, E4, VA pF∆6 During Transgene cDNA Sequences* AAV Expression Cassettes† AAV Packaging Plasmids† Table 2-1 Source of cDNA sequences and AAV Plasmids cDNA sequences of the genes-of-interest were sourced from either expression cassette plasmids previously constructed by the MJ During and D Young laboratory group, Department of Molecular Medicine and Pathology, The University of Auckland; or commercially sourced from Science Reagents. The empty AAV expression cassette, control AAV-Luciferase and AAV helper packaging plasmids were all kindly supplied by MJ During and D Young. * Full sequences in Appendix A.1. † Plasmid maps in Appendix A.2. Figure 2-1 AAV expression plasmid Diagrammatic representation of the AAV expression cassette plasmid showing the location of the inserted transgene between the CBA/CMV promoter and WPRE sequence. Left and right ITR sequences flanking the expression cassette direct packaging of ssDNA into the rAAV particles. 41 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing To investigate the neuroprotective potential of the four proteins-of-interest, BDNF, GDNF, Bcl-xL and XIAP, cDNA sequences for each were transferred into separate AAV expression cassettes. Each of the cDNA sequences were PCR cloned out of their source plasmids (Table 2-1; Appendix A.1) using customised primers (Table 2-2) to add unique restriction sites to each end of the cDNA sequence in preparation for ligation into the AAV expression cassette. All of the forward primers were constructed with a XhoI restriction site (CTCGAG) upstream of the ATG methionine start codon. Reverse primers were designed to allow fusion of the transgene protein with the C-terminal HA-tag by replacing the stop codons with a BglII restriction site (AGATCT) for the BDNF, GDNF and Bcl-xL cDNA sequences. Reverse primer for the XIAP sequence incorporated a EcoRI restriction sequence (GAATTC) due to the presence of a BglII recognition site within the XIAP cDNA sequence. Primer Sequence 5’ → 3’ TM BDNF – Forward CAACTCGAGCACCAGGTGAGAAGAGTGATGAC 82 BDNF – Reverse CTAGATCTTCTTCCCCTTTTAATGGTCAATG 75 GDNF – Forward GAACTCGAGCGGACGGGACTCTAAGATGAAG 82 GDNF – Reverse CAAGATCTGATACATCCACACCGTTTAGCG 78 Bcl-xL – Forward GCCCTCGAGTTATAAAAATGTCTCAGAGC 77 Bcl-xL – Reverse GAAGATCTGCGGTTGAAGCGTTCCTGGCC 82 XIAP – Forward GCGCTCGAGAATGACTTTTAACAGTTTTGAAGG 78 XIAP – Reverse CGAATTCGCGAGACATAAAAATTTTTTGCTTG 75 Table 2-2 PCR primer sequences Customised primer pairs were designed to PCR amplify the cDNA sequences from plasmid templates. The forward primers incorporated a unique XhoI restriction site (underlined) upstream of the cDNA coding region (bold). Reverse primers complimentary to the 3’ end of the cDNA gene sequence contained a BglII or EcoRI restriction site (underlined) in place of the stop codon. Optimisation of PCR programs to clone the cDNA sequences from source plasmid was performed using Red Hot DNA polymerase with annealing temperatures of 56, 60 and 65°C (Section 2.2.1.1). The cleanest PCR fragments were produced with the 65°C annealing temperature, however product yield was reduced compared with 60°C annealing. Reducing the number of cycles for XIAP from 35 to 30 and diluting the template DNA mini-prep 1:200 in dH2O resulted in a cleaner PCR product. After PCR program optimisation, High Fidelity DNA Polymerase was used to ensure accurate cloning of the cDNA sequences. High Fidelity PCRs were run with an annealing temperature of 62°C cycling 35 times for the BDNF, GDNF and Bcl-xL sequences and 30 times for XIAP. 42 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Following PCR cloning of each cDNA sequence I ligated the PCR products into pGEM cloning vectors for long-term storage and easy handling (Section 2.2.1.3). The pGEM plasmids were heatshock transformed into competent E. coli cells (Section 2.2.1.4) and individual colonies screened by colony PCR with the same primers (Table 2-2) to confirm insertion of the correct gene-of-interest (Section 2.2.1.5). A single colony generating the correct length PCR product for each gene-ofinterest was used to inoculate an LB culture for DNA plasmid mini-prep and storage as a glycerol stock (Section 2.2.1.8). DNA sequencing from plasmid mini-preps to confirm error-free cloning of each cDNA gene sequence was performed using T7 and Sp6 primers to sequence bidirectionally across the pGEM®-T Easy vector insertion site (Section 2.2.1.9). The cDNA sequences were each fully BLAST aligned with matching mRNA database sequences to confirm sequence homology (Table 2-3; Appendix A.1). The Bcl-xL sequence had a C-terminal truncation of the last 21 amino acids containing a transmembrane domain for mitochondrial targeting which has previously been shown to retain anti-apoptotic properties (Fang et al. 1994; Muchmore et al. 1996). BDNF, GDNF and XIAP cDNA sequences had full homology with database mRNA sequences (Table 2-3). Gene-of-Interest Description Accession Number BDNF Homo sapiens BDNF mRNA NM 170735.4 GI: 60218885 GDNF Rattus norvegicus GDNF mRNA NM 019139.1 GI: _9506720 Bcl-xL* Homo sapiens Bcl-2-like 1 mRNA NM 138578.1 GI: 20336334 XIAP Homo sapiens BIRC4 mRNA NM 001167.2 GI: 32528298 Table 2-3 Genes-of-interest * The Bcl-xL sequence encoded only the first 212 amino acids with a truncation of the final 21 codons encoding a transmembrane domain. Transfer of the cDNA sequences from pGEM cloning vectors into the AAV expression cassette was performed by excision of the cDNA sequences by restriction digestion (Section 2.2.1.6) followed by ligation into the AAV backbone plasmid (Section 2.2.1.7). Endonuclease digestion of the pGEM plasmids and the AAV backbone plasmid with the same pairs of restriction enzymes (Table 2-4) generated ssDNA overhangs on the excised cDNA sequences that complemented the ssDNA overhangs on the linearised AAV backbone plasmid. pGEM vector insert Restriction Enzymes Buffer Upstream Downstream BDNF / GDNF / Bcl-xL XhoI BglII NEB #3 XIAP XhoI EcoRI NEB EcoRI Table 2-4 New England Biolabs (NEB) restriction enzymes and buffers 43 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Digested cDNA sequence fragments were isolated by agarose gel extraction and the AAV backbone DNA CIP treated to prevent self-ligation. Complimentary pairing between the ssDNA overhangs directed orientation dependant ligation of cDNA gene sequences into the AAV expression cassette inframe with the C-terminal HA-tag (Section 2.2.1.7). The ligated AAV expression cassette plasmids were heatshock transformed into competent E. coli cells and plated onto LB agar / amp (Section 2.2.1.4). Individual colonies were screened for the presence of my cDNA gene sequences by colony PCR (Section 2.2.1.5). An E. coli colony that generated the correct length PCR product was selected to inoculate an LB culture for DNA plasmid mini-prep and storage as a glycerol stock (Section 2.2.1.8). To check that the cDNA sequences were correctly inserted into the AAV expression cassette in-frame with the HA-tag sequence, I used a DNA primer complementary to the downstream WPRE sequence (5’→3’ CAAATTTTGTAATCCAGAGGTTC) to reverse sequence across the HAtag and into the 3’ end of my inserted cDNA gene sequences (Section 2.2.1.9). After confirming the insertion of each cDNA gene sequence into the AAV expression cassette I performed DNA plasmid maxi-preps to generate large quantities of each plasmid for rAAV vector packaging (Section 2.2.1.8). In addition to the AAV backbone plasmids, maxi-preps were performed for each of the helper plasmids (pRV1, pH21 and pF∆6) required for rAAV packaging. The quantity of DNA in each maxi-prep was determined by spectrophotometry (Ultrospec™ 2100, Biochrom) and I performed restriction digests to check the purity of each plasmid by the length of digested DNA fragments on a 1% agarose gel (Appendix A.2.3). 2.2.3 Mammalian cell culture and analysis protocols 2.2.3.1 HEK 293 and HT-1080 Cell Culture Human embryonic kidney 293 cells (HEK 293) or human osteosarcoma cells (HT-1080) were used for rAAV vector packaging and checking transgene protein expression following plasmid transfection and AAV vector transduction. HT-1080 cells are more receptive to AAV transduction and were utilised for checking transgene protein expression and functional anti-apoptotic activity of AAVmediated XIAP expression. BDNF, GDNF and Bcl-xL expression was verified using HEK293 cells. A vial containing ~1 × 106 HEK293 or HT-1080 cells was removed from liquid nitrogen storage dewar, quickly thawed and transferred to cold Dulbecco’s Modified Eagle Media (DMEM; GIBCO®) containing 10% fetal bovine serum (FBS). To remove the storage media I pelleted the cells by centrifugation at 1000rpm for 5min (Heraeus Multifuge 3S-R), discarded the supernatant and resuspended the cells in 20mL DMEM / 10% FBS. The resuspended cells were transferred to a single T75 cell culture flask (Nunc) and incubated at 37°C, 5% CO2 changing media with fresh 44 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing DMEM as required. When the cells reached ~70% confluence I passaged them by rinsing with sterile PBS and incubating with 1mL 0.05% trypsin / 0.53mM EDTA for 3-5mins until the cells lifted from the plate with gentle tapping. Once the cells had detached from the culture flask I added 10mL DMEM / 10% FBS media to inhibit the trypsin and stop the dissociation reaction before centrifuging at 1000rpm for 5mins. The cells were resuspended in fresh DMEM / 10% FBS and split into 5 new T75 flasks or counted using a standard hemocytometer (Appendix B.2.2) and seeded onto cell culture plates at the required density. 2.2.3.2 In Vitro Immunocytochemistry Immunocytochemical staining was performed to visualise transgenic proteins and the expression of phenotypic markers following in vitro rAAV transduction. In vitro cell cultures were fixed by incubation with 4% paraformaldehyde for 10 mins and stored at 4°C in PBS. Endogenous peroxidase activity was blocked by adding a solution of 1% hydrogen peroxide / 50% methanol to the cells for 10 mins and then gently washing four times with PBS-Triton. The primary antibodies were diluted in PBS-Triton with 1% normal goat serum and incubated with the cells overnight at 4°C (Table 2-5). Secondary biotinlyated antibodies raised against the primary antibody host species were diluted 1:500 in PBS-Triton with 1% normal goat serum and incubated with the cells for 2-3hrs at room temperature with gentle rocking following four washes of the cells with PBS-Triton. Goat serum was omitted when staining with goat anti-Luciferase primary antibody. Extravidin peroxidase was diluted 1:500 in PBS and incubated with the cells for 2-3hrs at room temperature before I gently washed the cells four times with PBS and added DAB or DAB-nickel solution for 10mins to visualise cells expressing the target protein. Marker Antibody Dilution Supplier HA-epitope HA.11 1:1000 Covance Luciferase Luciferase 1:10000 Chemicon GABAergic Neurons Calbindin 1:5000 Swant Dopaminergic Neurons Tyrosine Hydroxylase 1:500 Chemicon Table 2-5 Primary antibodies for in vitro immunocytochemistry 2.2.3.3 Hoechst Nuclear Staining To visually assess apoptotic cell death in vitro, the cells were fixed with 4% paraformaldehyde and then incubated in 10 µg/mL Hoechst 33258 for 10 mins. Hoechst nuclear staining was visualised on an inverted Nikon Eclipse TE2000-U microscope with a UV filter. 45 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing 2.2.4 AAV expression cassette transfection testing Transfection of HEK293 or HT-1080 cells with my AAV expression plasmids was undertaken to confirm protein expression from cDNA gene sequences inserted into the AAV expression cassettes prior to vector packaging. HEK293 / HT-1080 cells cultured in high glucose DMEM (GIBCO®) with 10% FBS (GIBCO®) were plated into 12-well poly-D-lysine (Sigma-Aldrich) coated cell culture plates (Nunc) at a seeding density of 2 × 105 cells/well. After overnight incubation the cells were ~70% confluent and I replaced the media with 1mL IMDM (GIBCO®), 5% FBS for 2hrs prior to calcium precipitate transfection with my AAV backbone plasmids. To form a DNA CaPO4 precipitate I added 5µg of my AAV plasmid prep to 30µL 2.5M CaCl2, made up to 250µL with dH2O, filtered with a 0.2µm syringe filter (Pall) and transferred to 250µL 2× HeBS buffer (50mM HEPES, 280mM NaCl, 1.5mM NaHPO4) while vortexing. After resting for 1min I added the finely precipitated DNA solution dropwise to the HT-1080 cells media, swirled to mix and incubated for 4-8hrs before changing the media back to DMEM, 10% FBS. Following transfection the cells were incubated with DMEM media for three days before washing with sterile PBS and fixation with 4% paraformaldehyde. Immunocytochemistry was performed with an HA antibody to visualise the expression of the HA-tagged transgenic protein (Section 2.2.3.2). 2.2.5 AAV vector production 2.2.5.1 Packaging and Purification of AAV Vector Particles Production of chimeric rAAV vector particles was undertaken as previously described (During et al. 2003; Hauck et al. 2003) by co-transfection of HEK293 cells with the AAV expression cassette plasmid and AAV packaging plasmids containing the AAV Rep and Cap genes and Adenoviral E2A, E4 and VA genes required to assemble the AAV viral capsule and direct packaging (Matsushita et al. 1998). HEK 293 cells grown in high glucose DMEM, 10% FBS were seeded into five 15cm cell culture plates (Nunc) at 2.0-2.2 × 107 cells per plate one day prior to transfection. With the HEK 293 cells ~70% confluent I replaced the media with 25mL IMDM, 5% FBS 2hrs prior to CaPO4 transfection. A CaCl2–DNA solution was prepared using 12.5µg AAV backbone plasmid, 25.0µg pF∆6, 6.25µg pRV1, 6.25µg pH21, 330µL 2.5M CaCl2 (final concentration of 0.3M), made up to 13.75mL with dH2O and filter sterilised through a 0.2µm syringe filter (Pall). To precipitate the DNA for transfection I added 2.5mL 2× HeBS buffer to an equal volume of the CaCl2-DNA solution while 46 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing vortexing. The transfection solution was then left for 50secs to form a fine CaPO4 precipitate before adding drop-wise to one 15cm plate of HEK 293 cells and swirling gently to mix. This was repeated for each of the 5 plates. After overnight incubation (8-15hrs) I replaced the media with 25mL DMEM, 10% FBS. 48-60hrs after transfection I harvested the HEK 293 cells by gently washing with PBS, adding another 25mL PBS to each plate and using a cell scraper to lift the cells. The harvested cells were pooled together by centrifugation at 200g for 5min, resuspended in 50mL of a 150mM NaCl 20mM Tris pH 8.0 solution, split into two 25mL lots and frozen overnight at -20°C. Isolation of the rAAV vector particles from the harvested HEK 293 cells was performed by adding sodium deoxycholate (final conc. 0.5%) plus Benzonase® endonuclease (50U/mL) and incubating at 37°C for 30mins. Initial cellular debris was separated by centrifugation at 3000g for 15mins, 4°C. The supernatant was transferred to a new centrifuge tube and incubated at 56°C for 15mins. A freeze thaw cycle using a dry ice/ethanol bath and a 56°C water bath followed by centrifugation (3000g; 15mins; 4°C) to pellet cell debris was run three times before freezing supernatant overnight at -20°C. HiTrap Heparin HP columns (Amersham Biosciences) were used to purify the rAAV vector particles by re-centrifuging the harvested rAAV after thawing and passing the supernatant through a HiTrap Heparin HP column pre-equilibrated with 10mL of a 150mM NaCl, 20mM Tris pH 8.0 solution using a Harvard infusion pump set at 1mL/min. The rAAV loaded column was washed with 20mL 100mM NaCl / 20mM Tris pH 8.0 solution before eluting the purified rAAV vector from the column using 1mL solutions with increasing concentrations of NaCl - 200, 300, 400, 450, 450 and 500mM NaCl buffered with 20mM Tris pH 8.0. All of the eluted factions were pooled and the rAAV particles concentrated using 100K MWCO MicrosepTM Centrifugal Devices (Pall). The concentrators were centrifuged at 3000g, 4°C until there was ~300µL remaining. The concentrated rAAV vector was then loaded into a Slide-A-Lyzer dialysis cassette (Pierce) and suspended overnight at 4°C in a sterile container of PBS containing 1mM MgCl2. Following overnight dialysis I filtered the rAAV vector through a 0.2µm syringe filter (Pall) and divided into 10 or 15µL aliquots for storage at -80°C. 2.2.5.2 Genomic Particle Titre Concentrations of my rAAV vector stocks were determined by real time quantitative-PCR to measure the genomic particle titre for each vector preparation. The rAAV vector DNA was extracted in triplicate reactions from the rAAV vector particles using DNAseI (Invitrogen) and Proteinase K (Sigma-Aldrich). 2.0µL of the AAV vector was diluted in 10µL DNAseI reaction buffer (Invitrogen) plus 86.5µL dH2O and incubated with 0.5µL DNAseI at 37°C for 30min before inactivating the DNase at 70°C for 10mins. 0.5µL Proteinase K was then added and incubated at 50°C for 1hr before inactivation at 95°C for 20mins. The extracted DNA was further diluted 1:50 in dH2O for a total 47 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing vector dilution of 1:2500 and stored at -20°C. Standards with known genome copy number were prepared from a maxi-prep of the AAV-Luciferase backbone plasmid containing the WPRE sequence (Appendix B.3). The standard AAV backbone plasmid was diluted to 1 × 107 plasmid copies per µL and then ten-fold serial dilutions were done down to 1 × 104 copies/µL giving four plasmid DNA standards of known copy number. Quantitative-PCR was performed using primers to amplify the WPRE sequence from the expression cassette within each of my rAAV vectors DNA. Each quantitative-PCR sample was run in triplicate on a 96 well plate using 12.5µL ABsoluteTM QPCR SYBR Green mix (ABgene), 0.5µL forward WPRE primer (10µM; 5’→3’ GGCTCGGCTGT TGGGCACTGAC), 0.5µL reverse WPRE primer (10µM; 5’→3’ GGGCCGAAGGGACGTAGCA GAA), 6.5µL dH2O and 5.0µL of extracted vector DNA, standard WPRE plasmid dilution or dH2O as a no template control. The plate was sealed with an ABI PrismTM Optical Adhesive Cover (Applied Biosystems) and inserted into an ABI Prism® 7700 Sequence Detection System (v1.9; Applied Biosystems) programmed to run the following thermal cycle: Polymerase activation 95°C 10 min Denaturing 95°C 15 sec Annealing and Extension 60°C 60 sec Final Extension 72°C 10 min 1 cycle 40 cycles 1 cycle Mean cycle time (Ct) values from triplicate reactions were used to calculate the number of genomic copies in each of the AAV vector stocks (Appendix B.3). A standard curve was generated from the Ct values of the known concentration standards plotted against the Log value of the absolute number of plasmid copies in the reaction mix. Using the standard curve Ct values for the rAAV samples were converted to Log input quantities. To calculate actual genomic titres these values were inverse Logged, multiplied by the extracted DNA dilution factor of 2500 and multiplied by 200 to convert from the 5µL used in the reactions to copies per mL. To account for the single stranded AAV genome in contrast to the double stranded plasmid DNA the concentrations calculated from the plasmid standards were multiplied by 2 to give the actual genomic particle titre of my rAAV vectors. 2.2.6 In vitro transduction testing Each of the newly constructed rAAV vector stocks were tested on HEK 293 or HT-1080 cells to ensure they contained infectious particles. HEK 293 or HT-1080 cells were grown on poly-D-lysine coated plates (24-well, Nunc) to ~70% confluence in 1mL DMEM media with 10% FBS. The media 48 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing was replaced with fresh DMEM and 2.0µL of the rAAV stock was added to each well. Cells were incubated for one week refreshing the media four days after rAAV transduction. The final media from AAV-BDNF, AAV-GDNF and AAV-Luciferase transduced cultures was collected and stored at -80°C for ELISA quantification of secreted neurotrophic factors. Cells were then gently washed with sterile PBS and fixed with 4% paraformaldehyde. Immunocytochemistry was performed with either HA (Covance) or Luciferase (Chemicon) antibodies to visualise transgene protein expression within successfully transduced cells (Section 2.2.3.2). Control immunostaining was performed with the same antibodies on non-transduced cultures. Quantitative ELISAs were run on the collected media from the AAV-BDNF and AAV-GDNF transduced HEK293 cells to measure secreted BDNF and GDNF protein using Promega Emax ImmunoAssay kits (BDNF, G7610; GDNF, G7620). The conditioned culture media was diluted 1:2 and 1:4 in DMEM media and run in triplicate wells on the ELISA plate against the recommended eight-point standard series for each kit. Conditioned media collected from non-transduced HEK293 cells was also assessed on the same ELISA plates to provide controls for any endogenous neurotrophic factor expression. Optical absorbance was read at 450nm on a Spectra max plate reader (Molecular Devices) and the quantity of neurotrophic factor expression calculated from the standard curves. 2.3 Functional AAV-Mediated Protein Expression Testing In Vitro Functional assays were performed with each rAAV vector construct to ensure that the encoded protein-of-interest was expressed in a functionally active form following AAV transduction. Differentiation assays with primary neuronal cultures were used to visualise neurotrophic factor assisted phenotypic differentiation. Anti-apoptotic protein function was assessed with a cell death assay kit to measure the level of apoptosis following the induction of apoptosis in rAAV transduced in vitro cultures. 2.3.1 Isolating and culturing primary cells Primary neuronal cells were cultured from embryonic day 14 (E14) or day 15 (E15) Wistar rat embryos. Dopaminergic precursor neurons were isolated from the E14 ventral mesencephalon, GABAergic neuronal precursors from the E15 striatum and mixed cortical cultures from E15 cortex. Tissue from each brain region was isolated under a dissecting microscope as described by Barker and 49 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Johnson (1995). Three independent parallel cultures were established for each region from separate timed pregnant Wistar dams, harvesting tissue from 8-12 embryos per dam. The tissue from each embryo was finely diced in chilled Leibovitz L15 media (GIBCO®) with added glucose and pen/strep, and pooled together. Treating each dam separately, the dissected tissue was centrifuged at 300g for 2min and the L15 media replaced with 2mL fresh chilled L15 media. Three fire-polished Pasteur pipettes of decreasing opening diameter were used sequentially to gently triturate the diced tissue into single cells. After letting the debris settle the cloudy supernatant containing single cells was removed and centrifuged at 1000g for 5min to pellet cells. The cells were then resuspended in Neurobasal media (GIBCO®) with added glucose, L-glutamine, pen/strep and 2% B27, and plated as required onto 24- or 96-well plates (Nunc) coated with 0.1mg/mL poly-D-lysine and pre-incubated with 10% FBS in PBS. 2.3.2 AAV-BDNF The expression of functional BDNF was assessed by the presence of calbindin expressing neurons following AAV-BDNF transduction of primary striatal cultures. Primary striatal neurons isolated from E15 Wistar rat embryos from three independent Wistar dams were seeded onto 24-well plates at 2 × 105 cells per well (Section 2.3.1). AAV-BDNF or AAV-Luciferase vectors were added to the Neurobasal culture media 24hours after seeding at a concentration of 1,000 genomic particles per cell (2 × 108 AAV genomic particles per well). Triplicate culture wells seeded from each independent dam received either AAV-BDNF, AAV-Luciferase or were left untreated as negative controls (n = 9 per treatment). The cultures were maintained for 21 days at 37°C, 5% CO2 changing 50% of the Neurobasal media every three days before gently washing with PBS and fixation with 4% paraformaldehyde. The differentiation of the primary striatal neurons was assessed by immunocytochemistry with anticalbindin (1:5000, Swant) to visualise calbindin protein expression (Section 2.2.3.2). Cells expressing calbindin were quantified under a 40× objective (Nikon Eclipse TS100) in five randomly placed fields of view for each culture well. 2.3.3 AAV-GDNF To verify functional GDNF protein expression I transduced embryonic ventral mesencephalic cultures with AAV-GDNF to induce maturation towards dopaminergic neurons. Primary E14 ventral mesencephalon cells from three independent isolations were seeded onto 24-well culture plates at 2 × 105 cells per well in Neurobasal media and incubated overnight (Section 2.3.1). Twenty-four hours 50 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing after seeding the cells, AAV-GDNF or AAV-Luciferase vectors were added to the Neurobasal culture media, at a concentration of 1,000 genomic copies per cell (2 × 108 genomic copies per well), in triplicate wells for each culture. An additional three wells from each dam received 10ng/mL rhGDNF protein (R&D Systems) as a positive control and three wells were left untreated as a negative control. All cultures (n = 9 per treatment) were maintained at 37°C, 5% CO2 changing 50% of the media every three days with fresh Neurobasal and supplementing the positive control wells with 10ng/mL rhGDNF protein. Three weeks after plating the cultures were gently washed with sterile PBS and fixed with 4% paraformaldehyde. Dopaminergic neurons were visualised by immunocytochemistry with anti-tyrosine hydroxylase (anti-TH; 1:1000, Chemicon; Section 2.2.3.2) and quantified under a 40× objective (Nikon Eclipse TS100) in five randomly placed fields of view for each culture well. 2.3.4 AAV-Bcl-xL To test for functional Bcl-xL expression, primary cortical cultures were transduced with AAV-Bcl-xL and used to perform an apoptosis assay. Primary cortical cells from three independent cultures were plated onto 96-well plates at a density of 4 × 104 cells per well with 200µL Neurobasal media and incubated overnight at 37°C, 5% CO2 (Section 2.3.1). Twenty-four hours after plating the cells were transduced with either AAV-Bcl-xL (six wells per culture), or AAV-Luciferase (three wells per culture). The rAAV vectors were added to the Neurobasal culture media at a concentration of 1,000 genomic copies per cell and maintained for three weeks replacing half of the Neurobasal media every two-or-three days with fresh media. Apoptosis was induced three weeks after transduction in half of the AAV-Bcl-xL transduced wells and all of the AAV-Luciferase transduced wells by incubating the cultures with Neurobasal media containing 0.5µM staurosporine (Sigma-Aldrich) for 48hrs (n = 9 per treatment). The remaining three AAV-Bcl-xL transduced wells from each culture received PBS as non-apoptosis induced controls. A Cell Death Detection ELISAPLUS kit (Roche) was used to quantify the level of apoptosis occurring within each culture well by detecting apoptosis generated nucleosomes after 48hrs of staurosporine exposure. Briefly the culture media was carefully discarded and the cells lysed using the supplied lysis buffer and centrifuged to remove cellular debris and unfragmented DNA. Supernatant from each culture well was incubated in a streptavidin coated ELISA plate with immunoreagent solution containing both biotin labelled anti-histone and peroxidase conjugated anti-DNA antibodies. The quantity of bound nucleosomes was photometrically determined using ABTS (2,2-azino-di-3ethylbenzthiazoline sulfonate) as a substrate for the bound peroxidase. Absorbance was read at 51 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing 405nm on a Spectra max plate reader (Molecular Devices) with the non-apoptosis induced control wells used to normalise data across the three independent cultures. For visual analysis with immunocytochemical staining I cultured and treated a second set of primary cortical cells in parallel to the cultures used for the apoptosis quantification. Immunocytochemistry was undertaken with anti-HA (1:1000, Covance) and anti-Luciferase (1:10000, Chemicon) to visualise the transduced cells (Section 2.2.3.2). Hoechst 33258 nuclear staining allowed identification of cells undergoing nuclear condensation and fragmentation (Section 2.2.3.3). 2.3.5 AAV-XIAP To assess functional XIAP expression I performed an apoptosis assay on HT-1080 cell cultures transduced with either AAV-XIAP or AAV-Luciferase. HT-1080 cells were plated onto 96-well plates at a low seeding density of 1 × 104 cells per well in DMEM, 10% FBS and incubated overnight at 37°C, 5% CO2. AAV-XIAP or AAV-Luciferase were added to the culture media at a concentration of 10,000 genomic copies per cell, transducing 18 wells with each vector. Cells were grown for three days following AAV-XIAP transduction to ensure transgenic protein expression before incubating half of the wells from each group with DMEM containing 0.2µM staurosporine for 12hrs (n = 9 per treatment). Non-staurosporine induced wells served as controls. A Cell Death Detection ELISAPLUS kit was used to quantify the level of apoptotic cell death in each well by measuring apoptosis generated nucleosomes (as described for AAV-Bcl-xL in Section 2.3.4). Immunocytochemistry on parallel cultures was undertaken with either anti-HA or anti-Luciferase to visualise AAV-XIAP and AAV-Luciferase transduced cells (Section 2.2.3.2). Hoechst 33258 nuclear staining was performed to visually identify cells displaying apoptotic features of nuclear condensation and fragmentation (Section 2.2.3.3). 2.4 Results The production of recombinant AAV vectors was divided into two distinct elements: AAV plasmid construction, and AAV vector packaging. This was then followed by functional testing of the newly constructed AAV vector particles. 2.4.1 AAV plasmid construction Construction of the AAV expression cassettes for the neuroprotective investigations was greatly aided by the availability of an empty AAV expression cassette containing all of the desired regulatory 52 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing elements, requiring only the insertion of a gene-of-interest. Following the insertion of cDNA sequences encoding BDNF, GDNF, Bcl-xL or XIAP into separate AAV expression cassettes I transfected HEK293 and/or HT-1080 cells with each newly constructed AAV plasmid. This was performed to check protein expression driven from the AAV expression cassette prior to AAV vector packaging. Immunostaining of the HEK293 or HT-1080 cells with an HA antibody confirmed the expression of transgenic proteins following transfection for each of my four AAV plasmids (Figure 2-2A-D). The fusion of a small HA-epitope to the transgenic proteins allowed easy identification of transgenic protein expression in transduced cells without potential staining of endogenously A B C D E Figure 2-2 Transfection of AAV backbone plasmids Expression of HA-tagged proteins following AAV backbone plasmid transfection of HT-1080 cells visualised by immunocytochemistry with an HA antibody – (A) BDNF, (B) GDNF, (C) Bcl-xL, (D) XIAP and (E) non-transfected control culture. Scale bar = 25µ m. 53 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing expressed protein that is likely to occur with antibodies against the neurotrophic factors and antiapoptotic factors under investigation. All of the plasmids directed good expression of HA-tagged proteins with expression of BDNF and GDNF concentrated at the perimeter of the cells (Figure 2-2A,B) compared with the more evenly distributed anti-apoptotic factors (Figure 2-2C,D). No HA staining was detectable in any of the non-transfected culture wells (Figure 2-2E). 2.4.2 AAV vector production After successful vector packaging, the concentration of each AAV vector stock was quantified by real time PCR to determine its genomic titre. The AAV packaging and purification protocol produced reasonably consistent AAV titres of ~1012 AAV genomes per mL (Table 2-6) although the AAV-BclxL was lower at 5.1 x 1011 genomes per mL and AAV-XIAP significantly higher than the other vectors at 1.9 x 1013 genomes per mL. To allow reasonable comparisons between the different vectors I diluted the AAV-XIAP 10-fold with sterile PBS prior to use. AAV vector Genome Copy Number AAV-BDNF 2.0 × 1012 AAV-GDNF 2.2 × 1012 AAV-Bcl-xL 5.1 × 1011 AAV-XIAP* 1.9 × 1013 AAV-Luciferase 4.6 × 1012 Table 2-6 Genomic titres of AAV vector stocks High-titre recombinant AAV vector stocks purified by heparin affinity chromatography ranged from 1011 – 1013 genomic particles per mL. * The AAV-XIAP vector was diluted 10-fold to provided comparable vector titres of ~1012. 2.4.3 AAV vector transduction In vitro transduction of HEK293 or HT-1080 cells with each of the recombinant AAV vectors was confirmed by immunocytochemical staining with either HA or Luciferase antibodies (Figure 2-3). Immunopositive cells were present in all culture wells that received one of the AAV vectors. Specificity of the antibodies to the transgenic proteins was confirmed by the lack of staining in the non-transduced cultures (Figure 2-3B,G). Transgenic protein expression following AAV-BDNF or AAV-GDNF transduction once again appeared localised to the cell surface in contrast to the whole cell accumulation of the anti-apoptotic factors and Luciferase. The HT-1080 cell line appeared more receptive to rAAV transduction than the HEK293 cells, used initially to test the AAV vectors, as evidenced by greater numbers of HA- and Luciferase-positive cells in the HT-1080 cultures. 54 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Secretion of the neurotrophic factors into the culture media was quantified by ELISA to be approximately 350 pg/mL of BDNF and 600 pg/mL of GDNF. A B C D E F G Figure 2-3 AAV vector transduction in vitro Successful AAV vector transduction of HEK293 or HT-1080 cells and the expression of transgenic proteins visualised by immunocytochemistry with (A,B) Luciferase or (C-G) HA antibodies: (A) AAV-Luciferase; (C) AAV-BDNF; (D) AAV-GDNF; (E) AAV-Bcl-xL; (F) AAV-XIAP; (B,G) nontransduced cultures. Scale bar = 25µm. 55 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing 2.4.4 Functional protein expression 2.4.4.1 Brain Derived Neurotrophic Factor AAV-BDNF mediated expression of functional BDNF was assessed following AAV-BDNF transduction of primary embryonic day-15 striatal cultures. BDNF enhances GABAergic neuronal differentiation and induces calbindin expression in the striatal neurons (Mizuno et al. 1994; Ventimiglia et al. 1995). Calbindin-positive cells were observed in all of the striatal cultures (Figure 2-4). To standardise the quantification of calbindin-positive neurons across the three independent cultures I calculated calbindin-positive cell counts as a ratio of the untreated control wells for each culture (Figure 2-5). Addition of AAV-BDNF resulted in a 96 ± 2% increase in the number of calbindin-positive cells over untreated wells (P < 0.001). AAV-Luciferase transduction did not significantly alter the number of calbindin-expressing cells compared with untreated cultures. Additionally, calbindin-positive neurons in the AAV-BDNF transduced cultures were observed to have a greatly enhanced dendritic morphology compared with the restricted number of projections in the control cultures (Figure 2-4). 2.4.4.2 Glial cell-line Derived Neurotrophic Factor Transduction of undifferentiated ventral mesencephalon neuronal cultures isolated from embryonic day-14 rat embryos with AAV-GDNF was undertaken to verify the expression of functional GDNF (Figure 2-6). GDNF induces differentiation of neurons towards a dopaminergic fate in the developing mid-brain (Stromberg et al. 1993; Choi-Lundberg and Bohn 1995). Quantification of the TH-positive neurons was standardised to untreated control cultures to account for variations between the three independent source cultures (Figure 2-7). AAV-GDNF transduction induced a 62 ± 2% increase in TH-positive neurons compared with the number of TH-positive neurons in untreated cultures (P < 0.05). Supplementation of the culture media with rhGDNF as a positive control increased the number of TH-positive neurons by 46 ± 2%, however this failed to reach statistical significance (P > 0.05). The AAV-Luciferase vector control cultures were not significantly different from untreated cultures. TH-positive neurons within AAV-GDNF and rhGDNF treated cultures developed more extensive dendritic networks than the TH expressing cells in the AAV-Luciferase and untreated negative control cultures (Figure 2-6). 56 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing A B C D Figure 2-4 Phenotypic differentiation of primary striatal cultures Representative images of calbindin-expressing neurons within embryonic striatal cultures threeweeks post AAV-BDNF or AAV-Luciferase transduction. (A,B) AAV-BDNF transduced cultures contained calbindin-positive neurons that developed an extensive dendritic morphology. (C,D) AAVLuciferase and non-transduced control cultures had fewer calbindin-positive neurons which displayed restricted dendrite growth. Scale bar = 100µm for (A,C) and 50µ m for (B,D). Figure 2-5 AAV-BDNF induced calbindin expression in embryonic striatal cultures Graph showing the increased number of calbindin-expressing neurons following AAV-BDNF transduction relative to the AAV-Luciferase transduced and un-treated cultures. Cell counts from untreated control wells were used to normalise data between the three independent cultures. One-way ANOVA P < 0.001 with Bonferroni’s post-hoc tests * P < 0.01, ** P < 0.001. 57 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing A B C D Figure 2-6 Phenotypic differentiation of primary ventral mesencephalon cultures Representative images of TH-expressing neurons within embryonic ventral mesencephalon cultures three-weeks post (A) AAV-GDNF transduction, (B) supplementation with rhGDNF, (C) AAVLuciferase transduction or (D) non-transduced cultures. AAV-GDNF and rhGDNF treated cultures contained numerous TH-positive neurons that developed into groups of extensively networked neurons. A smaller number of the ventral mesencephalon neurons in control cultures also differentiated into TH-expressing neurons. Scale bar = 100µm. Figure 2-7 AAV-GDNF induced TH expression in ventral mesencephalon cultures Graph showing the increased number of TH-expressing neurons following AAV-GDNF transduction, and the addition of rhGDNF protein, relative to AAV-Luciferase transduced and un-treated cultures. Cell counts from untreated controls wells were used to normalise data between the three independent cultures. One-way ANOVA P < 0.05 with Bonferroni’s post-hoc tests * P < 0.05. 58 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing 2.4.4.3 Bcl-xL Expression of functionally active Bcl-xL protein from the AAV-Bcl-xL vector was checked by measuring staurosporine induced apoptotic cell death in primary cortical cultures transduced with AAV-Bcl-xL (Figure 2-9). Apoptotic cell death across each of my three independent cortical cultures was assessed relative to the quantity of apoptotic nucleosomes detected in the AAV-Luciferase vector control wells following staurosporine exposure (Figure 2-8). The transgenic expression of Bcl-xL by cortical cells in vitro, following AAV-Bcl-xL transduction, reduced apoptotic nucleosomes formation by 19 ± 2% compared with my control AAV-Luciferase transduced cultures. Apoptosis in the control AAV-Bcl-xL transduced cultures not exposed to staurosporine was measured at a level equivalent to 45 ± 2% of apoptosis cell death occurring in the AAV-Luciferase transduced controls exposed to staurosporine. Immunocytochemical staining with HA or Luciferase antibodies showed that only a small population of the cortical cells within each culture were successfully transduced (Figure 2-9A,C,E). Cells expressing transgenic Bcl-xL protein in the absence of staurosporine appeared healthy with extensive dendritic morphology (Figure 2-9A). Following the induction of apoptosis, AAV-Bcl-xL transduced Figure 2-8 AAV-Bcl-xL reduced staurosporine-induced apoptosis Graph showing AAV-Bcl-xL mediated reduction in the level of apoptotic cell death occurring in primary cortical cultures following staurosporine exposure relative to the AAV-Luciferase transduced wells used to normalise data across the three independent cultures. A substantial level of noninduced apoptotic cell death was recorded in non-staurosporine exposed AAV-Bcl-xL treated cultures. Mann-Whitney analysis * P < 0.01, ** P < 0.001. 59 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing A B C D E F Bcl-xL No STS Bcl-xL + STS Luc + STS Figure 2-9 AAV-Bcl-xL transduced cortical cells following staurosporine-induced apoptosis Representative images of primary cortical cultures transduced with (A-D) AAV-Bcl-xL or (E,F) AAV-Luciferase showing transgene expression and Hoechst nuclear staining. (A) Cortical cells transduced by AAV-Bcl-xL express HA-tagged Bcl-xL protein as visualised by HA-positive neurons not exposed to staurosporine. (C) After 48hrs of staurosporine exposure HA-positive cells were observed to resist cell death. (E) Very few Luciferase-positive cells resisted staurosporine-induced apoptosis. (B,D,F) Hoechst nuclear staining displays both large rounded healthy cells (arrowheads) and cells under going apoptosis induced nuclear fragmentation (arrows). Scale bar = 50µm (DAB and Hoechst images are not matched). Abbreviations: Luc, Luciferase; STS, Staurosporine. 60 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing cells were still present within the cortical cultures however they displayed condensed cell bodies with restricted dendritic projections (Figure 2-9C). Very few cells expressing Luciferase were seen to survive staurosporine induced cell death (Figure 2-9E). Hoechst staining showed widespread nuclear condensing and fragmentation within all staurosporine induced cultures (Figure 2-9D,F), in contrast to the predominantly large, rounded cells in the AAV-Bcl-xL transduced, non-staurosporine induced wells (Figure 2-9B). 2.4.4.4 X-linked Inhibitor of Apoptosis Protein AAV-XIAP mediated expression of functional XIAP was verified by transducing HT-1080 cells with AAV-XIAP and assessing the level of apoptosis following staurosporine-induced apoptotic cell death (Figure 2-11). HT-1080 cells are readily transduced by AAV vectors providing a clean substrate for assessing the effect of non-cell dependent protein expression. Transduction with AAV-XIAP resulted in a 52 ± 2% reduction in apoptosis compared with the apoptosis induced by staurosporine in AAVLuciferase cultures (Mann-Whitney P < 0.001; Figure 2-10). Non-staurosporine induced cultures transduced with either AAV-XIAP or AAV-Luciferase only had marginally detectable levels of apoptotic induced nucleosomes with no significant difference between the two groups. Figure 2-10 AAV-XIAP reduced staurosporine-induced apoptosis Graph showing AAV-XIAP mediated reduction in the level of apoptotic cell death occurring in HT-1080 cultures following staurosporine exposure relative to the AAV-Luciferase transduced wells used to normalise data across the three independent cultures. Negligible apoptotic cell death was detected in non-staurosporine exposed cultures. Mann-Whitney analysis * P < 0.001. 61 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Visualisation of the transduced cells with immunocytochemical staining for HA or Luciferase showed a population of the HT-1080 cells were transduced and expressed transgenic XIAP or Luciferase (Figure 2-11). Staurosporine induced extensive cell death in all cultures however a number of HA stained cells survived in the AAV-XIAP transduced wells and exhibited a neuronal-like morphology (Figure 2-11C). A few AAV-Luciferase transduced cells were observed after staurosporine exposure however they were located within patches of cellular debris non-representative of the HT-1080 cells (Figure 2-11G). Co-staining of the transduced cells with Hoechst 33258 further showed the survival of non-fragmented HT-1080 cells in the AAV-XIAP transduced cultures including both the surviving HA-positive transduced cells (Hoechst staining is compromised by the DAB staining) and some additional non-transduced cells (Figure 2-11D). Cells displaying nuclear condensation and fragmentation were observed with Hoechst staining in the AAV-Luciferase control transduced wells and to a lesser extent within the AAV-XIAP transduced wells following staurosporine exposure, with the total number of HT-1080 cells remaining clearly reduced in comparison to the non-staurosporine treated cultures. Figure on following page Figure 2-11 Enhanced survival of AAV-XIAP transduced HT-1080 cells Representative images of HT-1080 cells transduced with (A-D) AAV-XIAP or (E-H) AAVLuciferase showing transgene expression and Hoechst nuclear staining of the same cells (arrows; DAB staining reduces intensity of Hoechst staining). (A,E) HT-1080 cells are readily transduced by AAV vectors with individual XIAP expressing cells (C) surviving staurosporine exposure but with shrunken cytoplasm. (G) A few AAV-Luciferase transduced cells survived staurosporine treatment but were observed within clumps of cellular debris. Scale bar = 50µ m. Abbreviations: Luc, Luciferase; STS, Staurosporine. 62 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing A B C D E F G H XIAP No STS XIAP + STS Luc No STS Luc + STS Figure 2-11 Enhanced survival of AAV-XIAP transduced HT-1080 cells 63 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing 2.5 Discussion 2.5.1 AAV vector construction AAV vectors were constructed to deliver DNA expression cassettes, encoding the potential biotherapeutic proteins BDNF, GDNF, Bcl-xL or XIAP, to targeted neuronal populations in the CNS. To direct transgenic protein expression I used an expression cassette containing the constitutive CBA / CMV promoter-enhancer hybrid shown to generate high level transgene expression (Klein et al. 2002) without down-regulation, ensuring continuous long-term protein production. A downstream WPRE sequence and poly-A tail help to further enhance transgene expression by stabilising the mRNA allowing greater protein translation (Loeb et al. 1999; Paterna et al. 2000; Xu et al. 2001). Each protein-of-interest was cloned into the expression cassette, in-frame with a nine amino acid Cterminal HA-tag epitope to allow clear immunocytochemical visualisation of transgenic protein expression following cellular transduction. The expression cassettes were flanked by ITR sequences derived from serotype-2 AAV which have been extensively utilised in both preclinical research and clinical trails (Grieger and Samulski 2005). The ITR sequences are the only viral-derived DNA packaged into the rAAV vector particles which can hold up to ~4.7 kb of ITR flanked DNA. The expression cassettes I constructed ranged in length from ~2.9 for AAV-GDNF and AAV-Bcl-xL, ~3.0kb for AAV-BDNF, ~3.8kb for AAV-XIAP and ~4.1kb for AAV-Luciferase. Prior to rAAV packaging I confirmed protein expression from each AAV expression cassette by transforming the AAV-plasmids into in vitro cell cultures. HA-positive cells were present in all transformed cultures with the neurotrophic factors localising primarily to the cell surface and the anti-apoptotic factors appearing to disperse throughout the cytoplasm of the cells (Section 2.4.1). The expression cassettes were packaged into chimeric rAAV particles consisting of serotype-1 and -2 capsid proteins in a 1:1 ratio. The capsid proteins influence cellular transduction with this combination of serotype-1/2 giving enhanced transduction efficiency of neuronal cells over either capsid -1 or -2 alone (Hauck et al. 2003; Richichi et al. 2004). Utilising the heparin affinity of the serotype-2 capsid proteins (Zolotukhin et al. 1999), AAV vectors were purified by heparin affinity column and concentrated to ~1012 genomic particles per mL. While I tested that the vector stocks contained infectious particles by transduction of cell-lines in vitro (Section 2.4.3), I did not undertake infectious particle titres. I would assume that following heparin purification all remaining genomic copies were packaged within AAV particles. However as AAV production requires capsid assembly followed by the rate-limiting encapsulation of the ssDNA genome a large proportion of empty virions 64 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing are typically present within the final vector stocks and their theoretical impact cannot be discounted (Timpe et al. 2005). Empty capsids may compromise the gene delivery by unnecessarily increasing exposure of AAV particles which may influence transduction efficiency or enhance antibody generation, a potential concern for repeated AAV administration. 2.5.2 Functional protein expression The production of functionally active biotherapeutic molecules following gene transfer is clearly essential for any gene therapy procedure. It was therefore important that I performed functional assays with each recombinant AAV vector to determine that the proteins-of-interest were correctly produced in a biologically active form following cellular transduction. Taking advantage of the well characterised properties of neurotrophic factors in assisting neuronal differentiation and antiapoptotic factors in promoting cell survival, I designed and performed efficient phenotypic differentiation and cell-survival assays to confirm expected functional impact of the transgenic proteins following in vitro AAV vector transduction. 2.5.2.1 Differentiation Assays to Confirm Neurotrophic Factor Activity Neurotrophic factors are important for directing both phenotypic differentiation within the developing CNS, and maintaining cell plasticity and survival within the adult brain (Krieglstein 2004; Levy et al. 2005; Van Ooyen 2005). Neurotrophic factors are secreted proteins that signal via cell surface receptors and have both autocrine and paracrine activity. Therefore the biological effect of transgenic neurotrophic factor expression following gene delivery is expected to extend beyond the cells actually transduced by the viral vector. I developed functional assays for BDNF and GDNF using primary embryonic neuronal cultures isolated from gestation day 14 or 15 (E14 or E15) Wistar rat pups to visualise the effect of these neurotrophic factors on phenotypic differentiation following AAVmediated gene transfer. Previous studies have shown that BDNF enhances the differentiation of GABAergic neurons (Mizuno et al. 1994; Ventimiglia et al. 1995), while GDNF plays a key role in the differentiation and maturation of dopaminergic neurons (Lin et al. 1993; Widmer et al. 2000). Primary embryonic cultures were chosen to verify transgenic neurotrophic factor functionality as immature embryonic neurons at E14/15 are naturally primed by their micro-environment and express appropriate cell surface receptors for neurotrophic factors which assist in directing their phenotypic differentiation and neurite outgrowth (Ernfors et al. 1992). Functional assessment of virally mediated neurotrophic factor expression has previously been undertaken by collecting conditioned media containing the transgenic protein from transduced cell lines, applying this media to non-transduced cultures or explants and assessing morphological changes (Martinez-Serrano et al. 1995; Watabe et 65 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing al. 2000; Sakamoto et al. 2003; Hu et al. 2005). The use of conditioned media is important for assessing any novel biological functions of secreted molecules as it avoids the potentially confounding involvement of physical AAV vector transduction or altered protein expression. However the functional assays I performed demonstrated that AAV-mediated expression of functional BDNF and GDNF can effectively be assessed in a readily reproducible protocol by directly transducing undifferentiated primary neuronal cultures. Brain Derived Neurotrophic Factor The biological activity of BDNF was assessed following AAV-BDNF transduction of primary embryonic striatal cultures isolated from E15 Wistar rat pups (Section 2.4.4.1). Embryonic striatal neurons were chosen as they predominantly differentiate into GABAergic medium spiny projection neurons in vivo, and express TrkB the high-affinity cell-surface receptor for BDNF (Ernfors et al. 1992; Mizuno et al. 1994). BDNF is vital for both striatal development and maintenance throughout adult life with mature striatal neurons both producing BDNF as well as receiving a significant supply via anterograde transport of BDNF from the cortex via corticostriatal axonal projections (Altar et al. 1997; Kokaia et al. 1998). Primary striatal cultures were transduced with AAV-BDNF and maintained in vitro for three weeks to ensure a stable level of transgenic BDNF expression and provide sufficient exposure of BDNF to the striatal cultures to enhance GABAergic differentiation and maturation. Functional activity of BDNF was assessed by the induction of calbindin expression. Calbindin is a calcium binding protein expressed by a population of striatal GABAergic neurons (Gerfen et al. 1985; DiFiglia et al. 1989; Mizuno et al. 1994) shown to be induced in striatal neurons by BDNF (Ventimiglia et al. 1995; Ivkovic and Ehrlich 1999). Three-weeks post AAV-BDNF transduction there were up to twice the number of calbindin-positive neurons in the AAV-BDNF transduced cultures compared with both AAV-Luciferase vector control and untreated cultures. This significant increase in the number of calbindin-positive neurons observed in AAV-BDNF transduced cultures indicates that the HA tagged BDNF transgenic protein produced was functionally active in influencing the GABAergic differentiation of E15 embryonic striatal neurons. In addition to the increased differentiation, it was observed that calbindin-positive neurons in the AAV-BDNF transduced cultures had enhanced dendritic morphology compared to calbindin-positive neurons within non-transduced cultures, suggesting that the transgenic protein expression has enhanced maturation of these striatal neurons. This enhanced neurite outgrowth and maturation is consistent with increased BDNF expression providing additional supporting evidence that the transgenic BDNF is being correctly produced in a biologically active form. 66 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing Glial cell-line Derived Neurotrophic Factor Functional GDNF expression was verified following in vitro AAV-GDNF transduction of E14 ventral mesencephalon cultures. GDNF is a potent dopaminergic neurotrophic factor that assists with the differentiation and maturation of dopamine neurons. Neurogenesis and migration of dopaminergic precursor neurons into the ventral mesencephalon region occurs between gestation days E10 to E16 in rats, peaking around E14 (Altman and Bayer 1981; Prakash and Wurst 2006). The normal endogenous expression of GDNF, which peaks at birth in the CNS, assists in directing the dopaminergic precursor neurons to develop a dopaminergic phenotype (Stromberg et al. 1993; ChoiLundberg and Bohn 1995). GDNF receptors expressed on the terminals of dopaminergic precursor neurons allow retrograde transport back to the substantia nigra to direct the axon projections towards high-level GDNF expression within the developing striatum (Lin et al. 1993; Tomac et al. 1995). Three weeks after AAV transduction of E14 ventral mesencephalic cultures I performed immunocytochemical staining for TH, the rate-limiting enzyme in the production of dopamine, a phenotypic marker for dopaminergic neurons. With the E14 ventral mesencephalon cells already primed for a dopaminergic neuronal fate I observed a large number of TH-positive cells within all cultures. AAV-GDNF transduced wells however exhibited a significant (more than 50%) increase in TH-positive cells over AAV-Luciferase and untreated cultures with more extensive dendritic networking between the TH-positive neurons. Supplementation with rhGDNF failed to generate a statistically significant increase in TH-positive neurons despite showing an enhancement of THdifferentiation. This contrast with AAV-GDNF treatment was possibly due to the concentration of GDNF present in the cultures. AAV-GDNF transduced cells were likely to have been secreting a constant supply of GDNF that may well have led to significantly greater GDNF bioavailability than the 10 ng/mL rhGDNF added once daily to the E14 cells. AAV-Luciferase treatment did not alter the number of TH-expressing cells compared with untreated wells. The impact of AAV-GDNF in influencing differentiation into TH-positive neurons, exceeding the influence of rhGDNF supplementation, is consistent with the known activity of endogenous GDNF in driving undifferentiated ventral mesencephalon neurons towards a dopaminergic fate in vivo, and is strongly indicative that the transgenic GDNF-HA is functional active. The additional observation of enhanced dendritic networking by the TH-positive neurons, possibly due to axonal targeting of GDNF expressing neurons and retrograde transportation of the GNDF, is further supportive of an increased bioavailability of functional GDNF assisting the maturation of dopaminergic neurons. 67 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing 2.5.2.2 Cell-Survival Assays to Confirm Anti-Apoptotic Protein Function Anti-apoptotic proteins play a vital role in ensuring cell survival by counteracting or inhibiting the function of pro-apoptotic caspases and mitochondrial proteins. Enhancing the expression level of anti-apoptotic factors is likely to shift the apoptosis balance towards cell survival and therefore increase the cells resistance to specific apoptotic induction cues (Simons et al. 1999; Xu et al. 1999; Yamada et al. 1999; Shimazaki et al. 2000; Perrelet et al. 2002; Miagkov et al. 2004; Malik et al. 2005). Apoptosis can be quantified by detecting changes in apoptotic parameters including the activation of apoptotic caspases, mitochondrial release of pro-apoptotic proteins or DNA fragmentation (Loo and Rillema 1998; Goldstein et al. 2005). To verify functional anti-apoptotic activity of the Bcl-xL and XIAP protein expression following AAV-mediated transduction I quantified the generation of cytoplasmic histone-associated-DNA-fragments (nucleosomes) following the induction of apoptosis. Apoptosis was induced following AAV-Bcl-xL and AAV-XIAP transduction by incubating the cultures with staurosporine, a potent kinase inhibitor that induces apoptotic cell death through both caspase-dependent and caspase-independent mechanisms (Zhang et al. 2004). Staurosporine provides a broad approach to apoptosis allowing the assessment of various anti-apoptotic factors in different cell lines independent of a specific pathological process. Bcl-xL is a pro-survival member of the Bcl-2 family of apoptosis regulating proteins involved in controlling both caspase-9 activation via the mitochondrial release of cytochrome c and other caspase-independent apoptogenic proteins (Kim 2005). XIAP is a potent anti-apoptotic factor that functions as an inhibitor of both initiator caspase-9 and the executioner caspases -3 and -7 through its dual caspase inhibiting sites (Deveraux and Reed 1999). Bcl-xL Functionally active Bcl-xL expression was confirmed by comparing staurosporine-induced apoptotic cell death following AAV-Bcl-xL or AAV-Luciferase transduction of rat E15 cortical cultures. Bcl-xL has been previously shown to prevent staurosporine-induced apoptotic cell death (Kaspar et al. 2002). Apoptotic generated nucleosomes were detected in all cultures, including the non- staurosporine exposed wells, possibly due to the high cell density or the age of the primary cultures with many cells observed to be undergoing nuclear fragmentation. However the staurosporineinduced apoptotic cell death in AAV-Bcl-xL transduced cultures was significantly lower than the AAV-Luciferase transduced control cultures. A population of HA-positive AAV-Bcl-xL transduced cells were observed to survive staurosporine induced apoptosis in contrast to the loss of most AAVLuciferase transduced cells. This enhanced survival of Bcl-xL expressing cells reflects the 19% reduction in detected nucleosomes following AAV-Bcl-xL transduction. Assuming that non- staurosporine induced apoptosis occurred consistently in all cultures independent of treatment, the 68 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing actual reduction in staurosporine induced apoptosis following AAV-Bcl-xL transduction was ~35%. As Bcl-xL is not a secreted protein, only cells transduced by the AAV-Bcl-xL vector will be exposed to higher Bcl-xL levels and therefore have enhanced resistance against staurosporine-induced apoptosis. Given that I only transduced a proportion of the cultured cells the significant reduction in staurosporine induced apoptotic cell death of ~19% confirmed the expression of functionally active HA-tagged Bcl-xL protein. X-linked Inhibitor of Apoptosis Protein The functional activity of XIAP was assessed by inducing apoptotic death in HT-1080 cells cultures following AAV-XIAP and AAV-Luciferase transduction. Due to the highly permissive nature of the HT-1080 cell line towards AAV-mediated transduction, this cell line provides a reliable basis from which to assess functional expression from AAV vectors, and can be used as an alternative cell source when primary cells are not available or intrinsically necessary for assessing transgene function. The addition of staurosporine induced rapid cell loss and morphological changes in the HT1080 cultures as visualised by light microscopy and Hoechst 33258 nuclear staining. However a population of AAV-XIAP transduced cells survived the staurosporine insult and the level of nucleosomes detected was halved by AAV-XIAP transduction. Only a minimal level of apoptosis was detectable within the non-staurosporine induced cultures. Given that I only transduced a restricted proportion of the cultured cells, the large reduction in staurosporine-induced apoptosis provided by AAV-XIAP transduction verified that the transgenic XIAP expressed is functionally active. 2.5.3 Summary AAV vectors encoding each of my potential biotherapeutic molecules BDNF, GDNF, Bcl-xL or XIAP were successfully constructed and shown to direct the expression of functionally active proteins following cellular transduction. The use of efficient functional assays for known protein function provided reassurance that the therapeutic molecules were correctly translated and processed prior to proceeding with investigative in vivo gene transfer studies. The functional assays I conducted to verify neurotrophic factor functionality offer an efficient reproducible method for direct assessment of phenotypic changes within AAV vector transduced cultures in which there is continuous expression and secretion of the neurotrophic factors. The use of conditioned media from transduced cell lines which is commonly used to assess the biological activity of secreted factors restricts functional assessment to the secreted proteins acting on unmodified cultures and results in a variable level of protein exposure as the neurotrophic factors are 69 Adeno-Associated Viral Vectors: Plasmid Cloning, Vector Packaging and In Vitro Functional Testing used and degraded. In contrast, direct transduction assays assess the impact of continuous stable production and secretion of the transgenic proteins over an extended period of time in a cellular population containing both transduced and non-transduced cells. This ability to assess continuous stable expression would be of increased importance for vectors constructed to direct low level protein expression for long-term therapeutic application. While I could not exclude the possibility that physical transduction with AAV-BDNF or AAV-GDNF induced some phenotypic changes in the transduced cultures, I believe that it is the BDNF or GDNF secreted following AAV-mediated gene delivery that enhanced the differentiation of calbindin- and TH- expressing neurons by acting through cell surface receptors on both transduced and non-transduced cells. Cell-survival assays in vitro using staurosporine to induce apoptosis confirm the expression of functional anti-apoptotic factors. The use of different culture systems – primary embryonic culture versus an engineered cell-line – for the anti-apoptotic factors demonstrated the potential for utilising different cells when the protein under consideration is not cell-type specific. For testing expression from AAV vectors the cells clearly need to be receptive to AAV transduction of which both neuronal cultures and the HT-1080 cells are, however the HT-1080 cells appeared to offer several advantages over primary cultures. Most noticeable were the homogeneity and stability of an immortalised cellline with no confounding culture artefacts as observed along with the considerable level of apoptotic death occurring in the primary cultures possibly due to the high cell density or the length of time in culture. While in vivo studies are required to conclusively determine the physiological activity and therapeutic potential of any expressed molecule, these initial in vitro assessments are important for verifying expected functional activity. 70 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion Chapter 3 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion 3.1 Introduction To investigate preventative in vivo gene therapy for Huntington’s disease I utilised the QA lesion model of striatal cell death. QA, an endogenous glutamate analogue, induces excitotoxic mechanisms following over-activation of the NMDA receptors expressed postsynaptically by the GABAergic medium spiny projection neurons in the striatum. Directly injected into the striatum, QA produces the most accurate neurotoxin-induced representation of HD striatal pathology in both rodents and non-human primates (Beal et al. 1986; Beal et al. 1989). While QA does not replicate the genetic mutation or the slow progressive characteristics of HD neurodegeneration, excitotoxic mechanisms – and possibly endogenous QA – are proposed to be a significant contributor to HD following a mutant huntingtin-mediated increase in the susceptibility of vulnerable neurons to excitatory inputs (Cowan and Raymond 2006). Despite the extensive use of QA as a model of Huntington’s disease there has been no consensus within the relative literature regarding a delivery protocol for QA to the rodent striatum. It was therefore important that I briefly optimise a QA delivery protocol to generate a selective striatal lesion large enough to generate behavioural deficits without causing excessive mechanical damage or causing a physical hole at the site of injection. 3.2 Optimisation and Characterisation Procedures 3.2.1 Animals and surgeries Optimisation of the QA lesion model was undertaken in adult male Wistar rats (230 – 280g) with direct stereotaxic infusion of QA into the striatum on one side of the rodent brain. Stereotaxic injection of QA was performed as listed in Table 3-1 using surgical procedures outlined in Section 4.4. Based on my previous experience with QA administration I performed initial trials with 50 or 100nmol QA delivered in 500nL NaOH, pH 7.4. A second group of animals received either 30 or 71 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion 50nmol QA dissolved in 400nL NaOH, pH 7.4. Behavioural deficits were analysed on a further cohort of animals that received 30nmol QA in 400nL before I undertook a more detailed behavioural and immunocytochemical analysis of the selected 50nmol QA in 400nL lesion. Later development of an additional behaviour task necessitated the use of a final cohort of rats to assess the level of sensorimotor neglect. QA Coordinates from Bregma A-P M-L D-V No. rats Trial 1 100nmol, 500nL 50nmol, 500nL +0.5 +2.8 -6.0 2 2 Trial 2 50nmol, 400nL 30nmol, 400nL +0.5 +2.7 -5.0 3 3 Trial 3: Behaviour Testing Setup 30nmol, 400nL +0.6 +2.6 -5.0 4 Final Characterisation 50nmol, 400nL +0.5 ±2.7 -5.0 10 Additional Behaviour 50nmol, 400nL +0.5 +2.7 -5.0 4 Table 3-1 QA lesion optimisation trials Table detailing the intrastriatal injection of QA to produce unilateral lesioning of the striatum in the series of trials undertaken to optimise and characterise the neuropathological and behavioural changes. The anterior-posterior (A-P) and medial-lateral (M-L) coordinates were measured from Bregma and the dorsal-ventral (D-V) distance from the top of the dura with nose-tooth bar set at zero. 3.2.2 Behavioural assessment It was necessary that the unilateral QA lesion generated enough of a performance imbalance between the two hemispheres of the basal ganglia that resultant functional motor deficits could be reliably detected by an imbalance in the rats left / right side behavioural activity. I setup an array of behavioural tests to assess QA induced deficits covering: non-induced motor control (spontaneous exploratory forelimb use); sensorimotor neglect (“corridor” task); and drug induced activity (apomorphine and amphetamine induced rotations). Detailed procedures for each behavioural assessment are provided in Section 4.5. 3.2.3 Neuropathological analysis Rats were euthanised between seven and 20 days post-QA lesioning, fixed and processed for immunocytochemical assessment as described in Section 4.7.1. To analyse the extent of QA lesioning within the striatum, I used immunocytochemical techniques (Section 4.7.2) to visualise the 72 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion expression of the general neuronal marker NeuN (1:1000, Chemicon), and more specifically for GABAergic striatal neurons containing calbindin (1:5000, Swant) or DARPP-32 (1:750, Chemicon). Further characterisation of the final 50nmol QA lesion within the striatum was undertaken by immunocytochemistry against the interneuron marker proteins: calretinin (1:5000, Swant), parvalbumin (1:5000, Swant), choline acetyl-transferase (ChAT; 1:500, Chemicon) and NOS (1:10000, Piers Emson). 3.3 QA Lesion Optimisation Analysis 3.3.1 Initial QA trial As a starting point to optimise the QA lesion for the neuroprotective studies, I investigated 100 and 50nmol QA dissolved in 500nL 0.4M and 0.2M NaOH respectively, pH 7.4. These quantities of QA were selected based on both published literature (Qin et al. 1992; Arenas et al. 1993; Perez-Navarro et al. 1994; Perez-Navarro et al. 1996) and my own prior experience with QA ((Kells et al. 2004); unpublished data) in which I used these quantities but in significantly greater delivery volumes. With larger injection volumes it is common to observe physical damage to the striatum that can result in a hole at the site of injection. Analysis of NeuN immunopositive cells seven days after QA injection showed both 100 and 50nmol QA injections generated large extensive lesions within the striatum and in some cases extending out into the adjacent cortical regions (Figure 3-1). The 100nmol QA injection resulted in extensive tissue loss in the striatum along the needle tract in both rats which was not observed with the 50nmol injection. Additionally, there appeared to be greater numbers of NeuN-immunopositive cells still present within the “transition zone” surrounding the core of the 50nmol QA lesion indicating the differential selectivity / vulnerability of different populations of striatal neurons to this protocol of QA delivery. Deciding to investigate a slightly smaller lesion with a lower delivery volume and quantity of QA, I did not further assess the specific populations of striatal neurons. 73 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion A B 100nmol QA D C 50nmol QA Figure 3-1 Initial 100 and 50 nmol QA lesion assessment Representative images showing NeuN-positive neurons in the striatum following a unilateral injection of (A,B) 100nmol or (C,D) 50nmol QA (500nL) into the striatum. NeuN immunostaining displays extensive neuron loss within the injected striatum with physical damage surrounding the needle tract in the 100nmol QA injected striatum and loss of some adjacent cortical neurons. Surviving neurons were seen in the peripheral reaches of the lesion indicating a “transition zone” in which small populations of striatal neurons display resistance to the QA-induced excitotoxicity. Dotted lines show rough outline of the extent of QA lesioning in the striatum. Scale bar = 2mm for whole sections (A,C) and 500µm for higher magnification images (B,D) 3.3.2 Trial 2: Lower QA quantity While the 50nmol QA lesion in my initial trial generated an acceptable lesion representative of late stage HD striatal pathology, I wanted to investigate a more restricted lesion that would potentially be more representative of earlier stage HD and possibly more applicable to the neuroprotective gene therapy research. For this trial I reduced the delivery volume by 20% to 400nL to limit the mechanical infusion, investigating injections of 50 and 30nmol QA. Ten days post-QA the lower volume lesions appeared more contained than the 50nmol QA lesions in the initial trial, with both the 30 and 50nmol QA affecting ~50% of the striatum. The 30nmol QA looked to produce a slightly less severe lesion, with greater sparing of NeuN-positive striatal neurons 74 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion throughout the lesion, compared with a small “transition zone” following the 50nmol QA injection (Figure 3-2A, C). Only a small population of calbindin-positive neurons were seen within the lesioned area of the striatum for either concentration of QA (Figure 3-2B, D), suggesting that the spared NeuN-positive cells are probably populations of interneurons and not the projection neurons which generally express calbindin (DiFiglia et al. 1989). Therefore I felt the lesion produced by the 30nmol QA injection was potentially more representative of the selective degeneration of HD, providing a less intensive model of excitotoxic cell death more suited to assessing protective effects following delivery of neuroprotective biotherapeutic molecules. NeuN Calbindin B C D 30nmol QA 50nmol QA A Figure 3-2 QA lesion testing: 50 and 30 nmol intrastriatal QA injections Representative images showing surviving NeuN- and calbindin- immunopositive neurons following injection of (A,B) 50nmol or (C,D) 30nmol QA (400nL) into the striatum. Survival of striatal neurons within the lesioned striatum appeared greater in the lower concentration of QA (C), with some calbindin-positive neurons also seen to be maintained post-QA (D). Dotted lines show rough outline of the extent of QA lesioning in the striatum. Scale bar = 500µm 75 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion 3.3.3 Trial 3: Behaviour testing – 30nmol QA In order to later investigate functional protection against the development of motor impairments, I needed to ensure that the QA lesion was sufficient to generate behavioural deficits that could be readily observed and quantified by behaviour tests designed to assess imbalances in basal ganglia function following unilateral lesioning. Therefore to further assess the suitability of the less severe 400nL, 30nmol QA injection from the second QA lesion trial, I took another small cohort of rats (n = 4) and assessed their performance in behaviour tests seven days post-QA lesioning. Following QA lesioning of the right striatum, all of the rats displayed preferential use of their right (ipsilateral) forelimb for spontaneous exploration of the cylinder, with overall asymmetry scores recording 54 – 71% ipsilateral forelimb use. However the administration of apomorphine or amphetamine only stimulated strong rotational behaviour in two of the four rats. The rats that did respond displayed contrasting rotational effects arising from the two drugs. Apomorphine induced the rats to rotate towards the lesioned striatum (94% ipsilateral), while amphetamine stimulated rotations away from the lesioned striatum (96% contralateral). While these results were not expected, previous assessment of striatal lesioning has shown similar results when the anterior portion of the striatum alone is lesioned (Norman et al. 1992; Fricker et al. 1996). Overall these rats showed that the 30nmol QA lesion is severe enough to cause measurable behavioural deficits, however the lack of significant drug-induced rotational behaviour in half of the rats, and the differentially induced direction of rotation suggest that the ability to generate a lesion that gives consistent behavioural impairments over a large cohort of rats may be difficult with this quantity of QA. Larger lesions generally will provide greater consistency in overall behavioural performance between subjects, therefore I elected to not proceed with this 30nmol QA lesion but to use the higher concentration of 400nL, 50nmol QA for the neuroprotective investigations. 3.3.4 QA lesion model characterisation – 50nmol QA After assessment of all the QA lesion trials I decided to proceed with 50nmol QA in 400nL NaOH, as this protocol generated a large striatal lesion without obvious mechanical damage, while sparing a population of non-calbindin containing neurons within the lesioned area suggestive of interneuron sparing. To more comprehensively characterise the neuropathological and behavioural changes occurring following the 50nmol QA injection I assessed the development of quantifiable functional behaviour deficits and performed immunocytochemical staining to positively identify sub- 76 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion populations of striatal neurons to determine more specifically which neurons are maintained in this particular QA lesion model. 3.3.4.1 Functional Behaviour Assessment of Unilateral QA Lesioned Rats To ensure reliable assessment of the QA lesion and associated functional impairments I took a larger group of rats (n = 10) for investigation. Baseline measurements of exploratory forelimb use and amphetamine- / apomorphine-induced rotations were undertaken one week prior to stereotaxic QA injection. The rats were left for one week post QA injection to allow the acute lesion development to occur before proceeding with behavioural assessment. I analysed spontaneous exploratory forelimb use seven days post-QA lesioning followed directly by amphetamine-induced rotational assessment. Apomorphine-induced rotational behaviour was assessed ten days post-lesion before the rats were euthanised and processed for neuropathological analysis. Functional impairments were measurable by behaviour tests one week following QA lesion. The unilateral QA lesion rats developed a significant 63% ± 3 ipsilateral forelimb bias for exploratory behaviour compared with the 53 ± 3% baseline assessment (paired t-test P < 0.05; Figure 3-3C). Post-QA drug-induced rotational behaviour was preferentially towards the lesioned hemisphere with apomorphine-inducing 91% ± 5 of rotations to be ipsilateral and amphetamine 82 ± 9% ipsilateral, showing a significant shift from the proportion of baseline ipsilateral rotations of only 48% ± 9 and 34% ± 9 respectively (paired t-tests P < 0.05; Figure 3-3A, B). The extent of drug-induced rotational behaviour following the QA lesion with apomorphine administration significantly increasing total rotational behaviour from 2.2 ± 0.6 rotations per minute to 4.7 ± 0.7 (paired t-test P < 0.01), however amphetamine only induced a rotation rate of 1.6 ± 0.3 rotations per minute that was non-significantly increased to 2.7 ± 0.6 following the QA lesion (paired t-test P = 0.08). The amphetamine induction of ipsilateral rotations was in contrast to that induced following the smaller 30nmol lesion which caused the rats to rotate contralaterally, suggesting the extent of striatal lesioning determines the direction of rotation (Norman et al. 1992). 77 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion A Apomorphine B Amphetamine C Forelimb Use D Sensorimotor Neglect Figure 3-3 QA lesioning induced behavioural deficits Plots showing the development of measurable functional deficits following 50nmol QA lesion assessed by behavioural tests before and after 50nmol QA lesioning. Paired t-test: * P < 0.05, ** P < 0.01 3.3.4.1.1 Additional sensorimotor behavioural testing The inclusion of the recently developed “corridor” task (Section 4.5.2; (Dowd et al. 2005)) to assess sensorimotor neglect in the neuroprotective investigations required a small group of rats (n = 4) to facilitate the setting up of the corridor apparatus, determination of an assessment protocol, and to ensure that the 50nmol QA lesion induced a measurable sensorimotor deficit. Behavioural performance was observed on days one and two prior to QA lesion and on days 12 and 13 after injection. Two of the rats displayed right side bias for sugar pellet retrieval in the baseline testing prior to the QA lesion indicating a dominant left hemisphere. Unfortunately the QA was administered to the right striatum of each rat, rather than their dominant side, resulting in an ipsilateral group bias prior to QA. Following QA, three of the four rats showed an increased preference for ipsilateral (right side) retrievals with an overall group shift from 59 ± 7% to 76 ± 7% ipsilateral retrievals (Figure 3-3D). This did not reach statistical significance (Wilcoxon signed rank test: P = 0.25), however this 78 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion was possibly due to the small group size and lesioning of the non-dominant hemisphere in half the subjects. Reanalysis of the data using the duplicate trail results for each rat in an unpaired t-test did indicate the significant development of sensorimotor neglect after QA lesion (P < 0.05). 3.3.4.2 Neuropathological Assessment of QA Lesion Immunocytochemistry was performed on coronal sections through the striatum from each rat brain to assess the extent of the QA-induced striatal lesioning by NeuN-, calbindin- and DARPP-32immunopositive staining. More specifically the surviving striatal interneurons were visually identified with antibodies directed against calretinin, parvalbumin, NOS and ChAT (Section 3.2.3). Using the general neuronal marker NeuN, the loss of neurons following the 50nmol QA intrastriatal injections was generally confined to the striatal region causing extensive neuronal loss without obvious structural damage ten-days post injection (Figure 3-4A). In some cases where the lesion was situated in the dorsal or lateral portion of the striatum, a loss of neurons was also observed in the deep layers of the cortex overlying the lesioned striatum. A number of striatal neurons within the lesioned area were however resistant to QA excitotoxicity (Figure 3-4C) as also observed in the earlier testing (Figure 3-2). While a scattering of DARPP-32-positive neurons (Figure 3-4F) and calbindin-positive neurons (not shown) were still observed within the QA lesioned area, the majority of the QA resistant neurons were assumed to be striatal interneurons which normally make up ~5% of the striatal neuron population. Partial sparing of striatal neurons was particularly enhanced in the dorsolateral portion of the striatum with the medial extent of the lesion usually showing a distinct border between the lesioned and non-lesioned striatum (Figure 3-4D, F). Immunocytochemistry for markers of subpopulations of the striatal interneurons showed that only the small aspiny calretininpositive neurons (Figure 3-5C, D) and the large aspiny cholinergic ChAT-positive neurons (Figure 3-5G, H) were largely spared. The GABAergic parvalbumin (Figure 3-5A, B) and NOS (Figure 3-5E, F) immunopositive neurons were vulnerable to QA induced cell death. I also observed that parvalbumin expressing neurons of the globus pallidus were affected by the QA lesion (Figure 3-4G, H) suggesting a potential process of secondary cell death in the striatal output nuclei which may have occured following the loss of afferent projections from the striatum. No assessment was performed of the neurons in the substantia nigra following degeneration of the striatonigral projections. 79 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion A B NeuN Intact DARPP-32 Lesioned D E F G H Parvalbumin DARPP-32 NeuN C Figure 3-4 Pathological characterisation of the 50nmol QA lesion Immunocytochemical staining for (A,C,D) NeuN or (B,E,F) DARPP-32 positive neurons show the extent of the QA induced neuronal cell loss within the injected striatum compared to the intact contralateral hemisphere. (G,H) Loss of parvalbumin positive cells in the globus pallidus indicates possible QA induced cell death outside of the striatum. Dotted lines show a rough outline of the extent of QA lesioning in the striatum (D,F) and outline of the globus pallidus (G,H). Scale bar = 3.5mm for whole sections (A,B), 500µm for (C-F) and 900µ m for (G,H). 80 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion Intact Lesioned B C D E F G H ChAT NOS Calretinin Parvalbumin A Figure 3-5 Striatal interneurons following QA lesioning Representative images of the striatal interneurons within intact and QA lesioned striatum. GABAergic interneurons expressing (A,B) parvalbumin, (C,D) calretinin or (E,F) NOS are present in the striatum. The small population of calretinin neurons appeared to be fully maintained throughout the lesioned striatum, with the large ChAT-positive cholinergic interneurons also appearing to be relatively spared by QA. A few parvalbumin (B) and NOS (F) positive neurons were still present in the “transition zone” more distal from the QA injection site (right side of images). Scale bar = 200µ m 81 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion 3.4 Discussion Optimisation and brief characterisation of the QA lesion model of HD was undertaken to ensure the model generated the major pathological neurodegeneration of striatal projection neurons representative of HD and produced quantifiable functional deficits in the rats’ behaviour. While the QA model cannot be claimed to replicate the slow progressive neurodegenerative mechanisms occurring in the HD brain, the selective vulnerability of striatal neurons is suggestive of excitotoxic involvement. By restricting the quantity of QA and volume injected into the striatum I was able to generate a partially selective lesion of the striatal neurons without inflicting gross structural changes of the brain tissue. While the lower quantity of 30nmol QA resulted in a slightly less intensive lesion and would have debatably provided a more realistic representation of excitotoxic insult for my preventative gene delivery studies, concern over the reliable level of measurable behavioural deficits meant that the 50nmol QA lesion protocol was ultimately selected. 3.4.1 QA-induced neuropathology Previous investigations involving the intrastriatal injection of QA to model HD pathology in adult rats have been conducted with a diverse range of quantities and delivery volumes including: 300nmol, 1µL QA (Popoli et al. 1994; Scattoni et al. 2004); 225nmol, 1µL (Emerich et al. 1996; Borlongan et al. 2004; Emerich 2004b); 180nmol, 1µL (de Almeida et al. 2001; Escartin et al. 2004); 120nmol, 1µL (Dobrossy and Dunnett 2003); 120nmol, 0.5µL (Beal et al. 1986); 100nmol, 2µL (Alexi et al. 1999); 85nmol, 42.5nL (Brickell et al. 1999); 60nmol, 0.5µL (Bordelon et al. 1999); 50nmol, 2µL (Kells et al. 2004); 50nmol, 1µL (Figueredo-Cardenas et al. 1998); 15-60nmol, 1µL (Ryu et al. 2003). In my personal experience the larger quantities of QA have generally resulted in physical damage to striatum, often causing a physical loss of striatal tissue at the site of injection as observed in the initial 100nmol QA trial (Figure 3-1). Although neuroprotection has been achieved against some of the larger doses of QA, the protracted but inevitable neurodegeneration induced by the poly-Q tract expansion in huntingtin would indicate a more subtle contribution of excitotoxicity to striatal neuronal death, better replicated by lower quantities of QA. It is possible that spreading the QA across multiple injection sites in the striatum as performed by Dobrossy et al. (2003) may further reduce the intensity of excitotoxicity directly surrounding the injection sites, and thereby avoid inducing such severe damage. However the lower quantities of QA are still capable of inducing neuronal cell death over a considerable portion of the striatum following a single intrastriatal delivery site, indicating that QA can readily diffuse through the striatal parenchyma. Multiple injection sites would enable a greater proportion of the striatum to be lesioned and reduce 82 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion the extent of QA concentration gradients presumably responsible for the development of a “transition zone” of partial neuron survival surrounding a core of total neuron loss (Brickell et al. 1999). A region of complete neuronal death proximal to the site of injection was often not clearly distinguishable following the selected 50nmol QA lesion although there was still a gradient of increasing neuron survival away from the injection site. The selective sparing of small calretinin-positive interneurons and the large cholinergic neurons from QA-induced cell death is representative of their maintenance in the HD brain (Ferrante et al. 1987; Cicchetti and Parent 1996a; Cicchetti et al. 1996b) and in agreement with earlier QA lesion assessment (Figueredo-Cardenas et al. 1994). However the apparent loss of medium aspiny NADPHd / NOS or parvalbumin containing striatal interneurons contrasts with their relative invulnerability to HD neurodegeneration (Ellison et al. 1987; Harrington and Kowall 1991), but has previously been reported following QA lesioning (Davies and Roberts 1987; Waldvogel et al. 1991; Figueredo-Cardenas et al. 1994); although others have detected sparing of these NADPHd / somatostatin / neuropeptide Y interneurons (Beal 1992a; Beal 1992b) possibly due to differences in QA injection protocols. The observed selectivity of striatal neuronal sparing of the 50nmol, 400nL QA injection concurs with the quantitative analysis of striatal interneurons I previously conducted following a larger volume 50nmol, 2µL QA lesion (Kells et al. 2004). 3.4.2 QA-induced behavioural impairments A number of behavioural tests have been developed to assess functional deficits in rodent behaviour exhibited following excitotoxic lesioning of the striatum. While disturbances in cognitive function generally require bilateral lesioning, simple deficits in motor performance are readily induced by unilateral lesioning and can be detected by observing imbalances in lateralised behaviour (Nakao and Itakura 2000). Although it is difficult to correlate behavioural deficits in rodent models with HD symptoms, it is possible that some of the motor impairments are resultant from the same neuronal dysfunction responsible for the choreiform movements of HD patients. With symptomatic benefits being the most critical measure of any treatment strategy, the use of behavioural testing in animalbased investigations is a vital measure of therapeutic efficacy. Therefore to characterise the acquirement of motor deficits following the selected QA lesion I set-up four behaviour tests: druginduced rotational behaviour, apomorphine and amphetamine; spontaneous forelimb use; and sensorimotor neglect “corridor” task. Unilateral loss of striatal projection neurons creates a hemispherical imbalance in the populations of dopaminoceptive neurons. Administration of either a dopamine agonist (apomorphine) or dopamine83 Rodent Model of Huntington’s Disease: Quinolinic Acid Lesion releasing drug (amphetamine) following QA injection induced the majority of rats to rotate towards the lesion side (ipsilateral) as previously reported for excitotoxic striatal lesions with the extent of lesioning determining the degree of rotational asymmetry (Schwarcz et al. 1979; Dunnett et al. 1988; Nakao et al. 1996; Nakao and Brundin 1997). The loss of neurons expressing dopamine receptors would conceivably be expected to result in similar motor behaviour following drug induced dopamine release or dopamine agonist administration through greater dopamine receptor stimulation in the intact striatum. However, the brief assessment of a smaller 30nmol QA lesion suggested contrasting effects with amphetamine-inducing rotations away from the lesion side suggesting that smaller lesions may result in other more subtle changes to dopamine signalling, or may have been related to the positioning of the lesion with anterior lesions having been shown to induce contralateral rotational behaviour (Norman et al. 1992; Fricker et al. 1996). More traditionally used for assessing the integrity of the dopaminergic nigrostriatal connections (Choi-Lundberg et al. 1998), drug-induced rotational assessment can also provide some indication of striatal projection neuron maintenance; although inconsistencies in the direction of induced rotation, especially when dealing with small lesions, complicates assessment of deficit in relation to the degree of striatal lesioning. A general assessment of lateralised brain injury causing motor function impairment, the spontaneous forelimb use analysis following the QA lesion showed enhanced overall use of the ipsilateral forelimb. Greater use of the forelimb on the same side as the lesion is induced due to impaired CNS control of the contralateral forelimb, which is proportional to the severity of lateralised damage (Schallert et al. 1997; Schallert et al. 2000). Recent development by Dowd et al. (2005) of the “corridor” task, a simple test of lateralised response selection, to assess the development of sensorimotor neglect following unilateral 6-hydroxydopamine lesioning of dopaminergic neurons also showed contralateral side neglect following the unilateral QA striatal lesion. Although still an assessment of spontaneous motor control, the lateralised selection of food retrieval following balanced left and right side external sensory cues possibly provides a more sensitive assessment of basal ganglia control of sensorimotor coordination than the non-motivated exploratory forelimb use analysis. Overall, the non-drug induced assessments of sensorimotor function displayed significant behavioural deficits following the 50nmol, 400nL QA injection without inducing purely unilateral responses, which was often observed following drug-induced rotational behaviour. Sub-maximal responses are more likely to allow for the detection of subtle changes potentially provided by therapeutic agents against the neuropathological changes induced by QA lesioning. 84 Neuroprotective Study Methods Chapter 4 Neuroprotective Study Methods 4.1 Overview To investigate AAV-mediated delivery of biotherapeutics as a preventative treatment strategy against striatal neurodegeneration and the onset of functional motor control impairments, I surgically injected AAV vectors unilaterally into the rats’ striatum prior to an excitotoxic insult with QA (Figure 4-1). To monitor the development of functional impairments I put each rat through a series of behavioural tests both before and after AAV vector delivery to determine baseline behaviour and then repeatedly for 6-7 weeks following QA lesion. On completion of behavioural testing the rats were euthanised, paraformaldehyde fixed and the brains processed for immunocytochemical analysis. To determine expression levels of the biotherapeutics within the striatum at the time of QA lesion, I injected additional cohorts of Wistar rats with the same AAV vectors, euthanised after three weeks and isolated the striatum for quantitative ELISA analysis. QA injection AAV delivery -5 -4 Baseline behaviour -3 -2 Euthanised -1 0 1 2 Post AAV behaviour 3 4 5 6 … … Functional behaviour assessment Figure 4-1 Neuroprotective study timeline Timeline of the in vivo investigative studies to assess the neuroprotective effects of AAV-mediated gene transfer and prevention of functional behavioural deficits associated with a striatal lesion. Numbers represent weeks prior and post QA injection with rats euthanised at various end-points post-QA. 85 Neuroprotective Study Methods 4.2 Neuroprotective Investigations 4.2.1 In vivo expression testing To test in vivo transduction and transgene expression for each of the AAV treatment vectors, prior to commencing the neuroprotective investigation, I injected 4.0 µL of each AAV vector unilaterally into the striatum at one or two stereotaxic sites (Table 4-1). The rats were housed for three weeks following AAV injection to let transgene expression stabilise in the transduced cells before euthanizing and processing for HA-immunostaining to visualise the spread and extent of transgenic protein expression. Coordinates from Bregma A-P M-L D-V AAV vector delivery First Investigation – Single site +0.6 2.6 -5.0 Second Investigation – Two-sites +1.3 2.4 -5.0 -0.2 3.4 -5.0 Table 4-1 Stereotaxic injection coordinates for AAV vector delivery Injection coordinates were determined from a rat brain atlas (Paxinos and Watson 1986), and in vivo testing with AAV-Luciferase (Section 4.4.2). 4.2.2 Initial investigations: AAV-BDNF (high-titre), AAV-GDNF, AAV-Bcl-xL The first in vivo study I undertook investigated the protective effects of AAV-BDNF, AAV-GDNF and AAV-Bcl-xL administration. Wistar rats were randomly assigned into five groups to receive unilateral intrastriatal AAV vector or sham PBS injection (Table 4-4). Functional motor behaviour was assessed in the spontaneous forelimb use test and by drug-induced rotations (Section 4.5). Baseline behaviour was assessed to determine each rat’s naturally dominant hemisphere. Infusing directly into the dominant hemisphere using stereotaxic surgical procedures (Table 4-1; Section 4.4.1), each rat received 4.0 µL AAV-BDNF, AAV-GDNF, AAV-Bcl-xL, AAV-Luciferase or PBS, mixed with 1.0 µL 20% mannitol to increase the efficiency of transduction (Mastakov et al. 2001; Mastakov et al. 2002). Three weeks later all rats received an intrastriatal injection of QA at the same stereotaxic coordinates (Chapter 3). Functional motor behaviour was assessed weekly with the AAV-BDNF treated rats euthanised five weeks post-QA and the remaining groups seven weeks after QA (Table 4-2). 86 -4 ! Spontaneous Forelimb Use ! Apomorphine Rotations ! Amphetamine Rotations -3 -2 -1 ! ! ! 0 1 2 3 4 5 6 ! ! ! ! ! ! Table 4-2 First neuroprotective investigation timeline 4.2.3 Follow-up ! ! ! ! investigations: ! AAV-BDNF ! 7 Perfused -5 AAV Delivery Week QA Injection Neuroprotective Study Methods (diluted), AAV-Bcl-xL, AAV-XIAP My second in vivo investigation followed-up on the initial results of the previous study to further investigate AAV-BDNF and AAV-Bcl-xL administration, in addition to assessing the newly constructed AAV-XIAP vector. Rats were randomly assigned to receive AAV-BDNF, AAV-Bcl-xL, AAV-XIAP, AAV-Luciferase or PBS (Table 4-4). The dominant brain hemisphere was determined for each rat following baseline behaviour testing in the “corridor” task, spontaneous forelimb use and apomorphine-induced rotations (Section 4.5). Following baseline behaviour assessment, I injected the AAV vectors or PBS into the dominant striatum. In contrast to the first study (Section 4.2.2) I decided to split the vectors over two-injection sites within the striatum (Table 4-1), with each site receiving 4.0 µL vector or PBS plus 1.0 µL 20% mannitol, except the AAV-BDNF rats which only received 2.0 µL vector plus 1.0 µL 20% mannitol per site. The AAV-BDNF and AAV-XIAP vectors were each diluted 1:10 with sterile PBS prior to injection. QA was administered three weeks after vector delivery to all rats except five randomly chosen AAV-BDNF animals which provided nonlesioned AAV-BDNF treated controls. Rats were all euthanised eight weeks after QA lesion with “Corridor” Task Table 4-3 ! ! ! -3 -2 -1 ! ! ! 0 1 2 3 4 5 6 7 ! ! ! ! ! ! ! ! ! Second neuroprotective investigation timeline 87 ! ! ! ! ! 8 Perfused Apomorphine Rotations -4 QA Injection Spontaneous Forelimb Use -5 AAV Delivery Week Commenced Dietary Restriction behaviour assessed weekly or biweekly throughout (Table 4-3). Neuroprotective Study Methods 4.3 Animals Number of Rats 29 (Section 3.2.1: Table 3-1) QA Lesion Development AAV vector and Coordinate Testing AAV-BDNF 10 AAV-GDNF 6 AAV-Bcl-xL 6 AAV-XIAP 6 AAV-Luciferase 12 Functional Protection Transgene Expression AAV-BDNF 7 5 AAV-GDNF 10 5 AAV-Bcl-xL 10 5 AAV-Luciferase 8 5 PBS 10 - AAV-BDNF - no QA 9 5 5 - AAV-Bcl-xL 11 5 AAV-XIAP 9 5 AAV-Luciferase 5 - PBS 5 - First Investigation Second Investigation Table 4-4 Allocation of animals Cohorts of male Wistar rats analysed for QA lesion development, AAV vector testing and neuroprotective gene transfer studies. All of the in vivo studies were conducted in adult male Wistar rats (200 – 300g) supplied by the University of Auckland Animal Resources Unit with strict adherence to approvals granted by the University of Auckland Animal Ethics Committee in accordance with the NZ Animal Welfare Act (1999). Rats were housed in a temperature and humidity controlled environment on a 12hr light / 12hr dark regime. Depending on experimental design, housing was either in groups of 4-5 with food and water available ab libitum, or individually when dietary restriction was required to maintain the rats at 85% of their expected free feeding weight for incentive based behaviour testing – water was available ab libitum. 88 Neuroprotective Study Methods Following immunocytochemical analysis of the transgene expression and visualisation of the QA lesion in the main neuroprotective investigations I removed from the analysis any rats that lacked transgenic protein expression in the striatum, had no evidence of QA lesioning, or showed misplacement of either AAV vector or QA injection. 4.4 Stereotaxic Surgeries: AAV Vector Delivery and QA Injection 4.4.1 Operating procedure Direct intrastriatal injection of the AAV vectors and QA was performed by stereotaxic surgery techniques using Kopf® small animal stereotaxic frames paired with a MicroSyringe Pump Controller (Micro 4; World Precision Instruments) to control the injections via Hamilton® syringes. Rats were systemically anaesthetised with an initial intraperitoneal (i.p.) injection of 60 mg/kg sodium pentobarbital (National Veterinary Supplies Ltd, NZ) plus additional dosing as required to maintain anaesthesia throughout the surgery. Marcain (AstraZeneca) was administered (0.5 mg/kg) by subcutaneous (s.c.) injection, directly above the rat’s skull, prior to beginning surgery to provide local analgesic at the site of incision. The rats were firmly secured into a Kopf® stereotaxic frame by nose/tooth bar and ear bars with the nose/tooth bar zeroed to hold the skull level. The skin above the skull was opened with a single longitudinal incision to expose the skull and positively identify Bregma from which the A-P and M-L stereotaxic injection site coordinates were measured. A burr hole was drilled with a 0.5mm tungsten carbide bit (Sunshine®-Dental) in the skull at the A-P, M-L injection coordinates through which a 5 or 10µL Hamilton® syringe (32G needle) was carefully lowered 1mm/min to the desired depth below the dura. Once the syringe was correctly positioned the MicroSyringe Pump Controller was programmed to slowly infuse either an AAV vector / PBS vehicle control at 150 nL/min or QA at 100 nL/min. After injection the syringe was left in place for an additional 5mins before withdrawing to minimise backflow up the needle tract while the syringe was slowly withdrawn. On completion of the stereotaxic injections the skin incision was sutured closed (Silk 4/0, Resorba) and Xylocaine® 2% Jelly (AstraZeneca) was topically applied to the wound. The rats were placed under a heat-lamp and monitored post-operatively until they recovered from the systemic anaesthesia before returning to their home cages. 89 Neuroprotective Study Methods 4.4.2 Determination of initial AAV vector injections parameters To test the striatal coordinates and the required volume for AAV vector delivery I performed a small test using the AAV-Luciferase vector injecting either: 3.0 µL (n = 3) or 4.5 µL (n = 3) at a single site (A-P +0.5, M-L 2.7, D-V -5.0 mm); or 5.0 µL evenly split between two injection sites (n = 3; Site 1: A-P +1.0, M-L 2.7, D-V -5.0; Site 2: A-P -0.2, M-L 3.0, D-V -5.0). All vectors were injected along with 1.0 µL 20% mannitol to increase the efficiency of transduction (Mastakov et al. 2001; Mastakov et al. 2002). The Rat Brain in Stereotaxic Coordinates (Paxinos and Watson 1986) was used as a guide to determine initial stereotaxic coordinates for all injection sites. Intrastriatal injection of AAV-Luciferase led to transduction of the targeted striatal cells plus pyramidal cortical neurons surrounding the needle tract, and pallidal neurons (Figure 4-2). The lower volume injection tended to result in transduction of striatal neurons in a broad band surrounding the needle tract in contrast to the more extensive spread of transduction throughout the striatum following the single 4.5 µL or two-site 5.0 µL injection protocols. Cortical neurons surrounding the needle tract were extensively transduced indicating some backflow of the vectors up the needle tract and also along the top of the corpus callosum. It is also possible that some of the cortical transduction was facilitated by retrograde transport of the AAV vectors as previously observed (Kaspar et al. 2002; Burger et al. 2004). Transduction of pallidal neurons within the globus pallidus, a target nucleus of the striatal projection neurons, is indicative of anterograde vector transport and was observed following all injection protocols. Transduction of the nigral neurons seen in the main investigation was not assessed in these initial AAV-Luciferase transduced animals. The anterograde transport and transduction of target nuclei may well be of therapeutic benefit for delivering biotherapeutics to the terminal axonal projection fields of the medium spiny neurons and supply target derived support of the neuronal projections against cellular insult. Given the extensive spread of striatal transduction achieved following a single injection I decided to proceed initially with a single site 4.0 µL delivery protocol for my neuroprotective studies to minimise any surgical-related trauma associated with the vector delivery and generally restrict transduction to the striatum. The 4.0µL vector plus 1.0µL mannitol injection allowed the use of 5.0µL Hamilton syringes. In later investigations I elected to use a two-site injection to enhance the distribution of transduced neurons away from the direct site of QA injection. 90 Neuroprotective Study Methods A B C D Figure 4-2 Intrastriatal AAV vector delivery (A) Visualisation of in vivo Luciferase expression following trials of various intrastriatal AAVLuciferase injection protocols. Intrastriatal injection of AAV-Luciferase resulted in transduction of (B) cortical neurons, (C) striatal neurons, and (D) neurons within the globus pallidus. Scale bar = 2mm for whole sections (A), 100µm for (B) and 50µ m for (C,D). 91 Neuroprotective Study Methods 4.5 Functional Behavioural Analysis To assess the physical impact of AAV-mediated treatment on the rat’s behaviour I observed their motor performance in a range of behavioural tests designed to show preferential biasness between the left and right side of the rats following a unilateral lesion in the basal ganglia. All assessment of behavioural testing was performed blinded to the specific AAV vector / PBS delivery and the treated hemisphere. 4.5.1 Spontaneous exploratory forelimb use Spontaneous exploratory forelimb use assessed the development of a preference for the QA lesioned rat to use its forepaw ipsilateral to the lesioned hemisphere when exploring a novel environment (Schallert et al. 2000). Rats were transferred from their home cage to a clear acrylic cylinder (200mm internal diameter; 400mm high) where they were free to explore by rearing up on their hindpaws and using their forepaws to support themselves against the cylinder walls. Video recording of 5mins exploration was analysed to quantify left and right forepaw usage while rearing off the floor, initial placement on the cylinder wall and for landing back on the floor. A single asymmetry score was calculated by averaging the percentage of time each rat used its ipsilateral forepaw for rearing, initial cylinder wall placement and landing. A B Figure 4-3 Spontaneous exploratory forelimb use Each rat was placed into clear cylinder and video recorded for 5 mins for later analyse of left and right forelimb usage while exploring the cylinder. The placement of two-mirrors behind the cylinder allowed a full 360° view of the rats. Rat in (A) is shown initiating an exploratory rear off its right forelimb, and in (B) contracting the cylinder wall with its left forelimb. 92 Neuroprotective Study Methods 4.5.2 “Corridor” task The “Corridor” Task recently developed by Dunnett and colleagues to study 6-hydroxydopamine induced lesions assesses left / right preference for food retrieval (Dowd et al. 2005). Rats are placed into a long narrow corridor with caps containing sugar pellets located on each side of the corridor floor in evenly spaced adjacent pairs. The rat is free to explore up and down the corridor but when retrieving sugar pellets it is required to make a left or right selection or move onto the next pair of sugar pellet containing caps. Although not previously reported to assess QA lesions I choose to investigate using this test as it introduces an element of decision making and assesses the development of any sensorimotor neglect. To undertake this task the rats were diet restricted to 85% free feeding body weight in accordance with our ethical approval. In an effort to restrict exploratory behaviour during assessment the rats were habituated to the testing apparatus one day prior to testing by placing each animal into the corridor for 10mins with a few sugar pellets scattered along the floor of the corridor. Then immediately prior to testing the rats were placed into an empty corridor for 5mins before transferring directly into a corridor setup to run the task. Retrievals were scored whenever the rat placed its nose into a cap regardless of the number of sugar pellets actually retrieved. The first 20 retrievals (max 5mins) were recorded as being either from the left- or rightside. Rats were tested on two consecutive days with the percentages of retrievals ipsilateral to the lesion side averaged to ensure consistency. 93 Neuroprotective Study Methods 2400 70 350 ø 20 130 depth: 200 Figure 4-4 Sensorimotor “corridor” task Dietary restricted rats were allowed to freely explore up-and-down the corridor retrieving sugar pellets from evenly spaced containers along each wall of the corridor. A clear Perspex lid dissuaded the rats from vertical exploration. Corridor dimensions in mm. 4.5.3 Drug-induced rotational analysis Drug-induced rotational behaviour analysis was used to measure bilateral imbalances in the dopamine system. Apomorphine – a dopamine agonist – and amphetamine – a dopamine releasing agent – are commonly used to assess functional performance in Parkinson’s disease models but have also been employed in QA lesion studies (Schwarcz et al. 1979; Dunnett et al. 1988). I investigated the used of both apomorphine (1mg/kg injected s.c.) and amphetamine (5mg/kg injected i.p.) dissolved in 0.9% saline. Following drug administration the rats were placed into rotational bowls and assessed for 1hr either by video analysis or in real time using an automated Rotomax Rotometer (AccuScan Instruments) system. 94 Neuroprotective Study Methods Figure 4-5 Drug-induced rotational behaviour Rats were fitted with a harness, placed into clear rotational cylinder and tethered to a Rotomax Rotometer system used to quantify rotational behaviour for 1 hour following apomorphine or amphetamine administration. 4.6 Quantitative Transgene ELISA Quantification of the transgene expression within the striatum following AAV vector gene delivery was undertaken to determine the level of biotherapeutic protein expression at the time of QA injection. Three weeks after AAV vector delivery the rats were deeply anesthetised with an overdose of sodium pentobarbital (~300mg/kg i.p.), decapitated and their brains immediately removed. The brains were split into two hemispheres down the mid-line and the striatal region from each hemisphere dissected out, snap-frozen in liquid nitrogen and stored at -80°C. Homogenisation of the striatal tissue in preparation for ELISA quantification was performed in a 50mM Tris buffer pH 6.8 containing 0.5% Tween-20, 0.1% sodium azide, 1.5g/L EDTA, 5mg/L Pepstatin A and 10mg/L PMSF. Frozen striatal tissue was transferred to a 15mL tube along with 600µL of homogenisation buffer and mechanically homogenised using a motorised cylindrical Teflon pestle until tissue fragments were no longer visible. Homogenates were centrifuged at 3,000rpm for 15mins at 4°C. The protein containing supernatant was divided into aliquots and stored at -80°C to prevent protein degradation. Quantitative ELISA assays were undertaken in accordance with the appropriate ELISA kit instructions. Promega Emax® ImmunoAssay Systems were used for BDNF (G7610) and GDNF (G7620), and R&D Systems Duoset® IC for XIAP (DYC822) and Bcl-xL (DYC894). To perform quantification the sample supernatants were each run in triplicate at two dilutions (Table 4-5) along with samples from AAV-Luciferase treated rats as AAV-vector transduction controls (1:10 and 1:50 dilutions) and each kits recommended eight-point standard series. Optical absorbance of the ELISA plate was read on a Spectra max plate reader (Molecular Devices). Protein concentrations calculated from the standard curve were subsequently standardised against total protein in each sample. 95 Neuroprotective Study Methods Total protein was measured using bicinchinonic acid (BCA; Sigma-Aldrich) and bovine serum albumin (BSA; Invitrogen) as a protein standard. A 4000 µg/mL BSA standard was prepared in 1N sodium hydroxide and serially diluted 1:2 to produce an eight point standard curve from 2000 – zero µg/mL. Sample supernatants were diluted 1:10, 1:20 and 1:40 and plated into a 96 well ELISA plate in duplicate alongside the BSA standards also plated in duplicate (50 µL/well). 100µL BCA reagent (1mL 4% copper sulphate solution per 50mL BCA) was added to each well and incubated at room temperature for 2-3 hours before reading on a Spectra max plate reader at 562nm. Total protein was calculated from the BSA standard curve. Sample Dilutions Initial Investigations Ipsilateral Contralateral AAV-BDNF 1:1000, 1:5000_ 1:10, 1:50 AAV-GDNF 1:500, 1:2500 1:10, 1:50 AAV-Bcl-xL 1:50, 1:250 1:10, 1:50 AAV-BDNF (1:10) 1:500, 1:1000 1:10, 1:50 AAV-Bcl-xL (Two-site) 1:500, 1:1000 1:10, 1:50 AAV-XIAP 1:500, 1:1000 1:10, 1:50 Follow-up Investigations Table 4-5 Dilutions used for ELISA quantification of in vivo transgenic protein expression. Transgenic protein containing supernatants from the striatal tissue homogenates were diluted in supplied buffers to appropriate levels for ELISA quantification analysis. The ipsilateral hemisphere required greater dilution to fit standard curves. 4.7 Immunohistochemical Analysis Immunocytochemistry was performed to visualise transgene expression following AAV gene delivery, analyse QA induced neurodegeneration and to correlate the attenuation of behavioural impairments with structural maintenance of brain anatomy. 4.7.1 Brain tissue collection and processing Paraformaldehyde fixation of the rat brains was used throughout the research. The rats were deeply anesthetised with an overdose of sodium pentobarbital (~300mg/kg i.p.) and perfused transcardially with 100mL chilled 0.9% saline followed by 500mL 4% paraformaldehyde in 0.1M phosphate buffer, pH 7.4. Following ~30min perfusion the brains were removed, post-fixed overnight in 4% 96 Neuroprotective Study Methods paraformaldehyde at 4°C and then cyroprotected for sectioning in 30% sucrose 0.1M phosphate buffered solution. A sliding microtome with freezing stage was used to cut 40µm coronal sections through the striatum from frozen brains. Eight sets of sections were collected from each brain (distance of 320µ m between consecutive sections in each set) and stored in a cyroprotectant solution (30% sucrose, 30% ethylene glycol, 0.5M phosphate buffer) at -20°C. 4.7.2 DAB-staining immunocytochemistry protocol The visualisation of specific cellular populations and / or structure is possible with immunocytochemistry using antibodies raised against specific proteins or peptides uniquely expressed by the cells of interest. I utilised a DAB (3,3’-diaminobenzidine tetrahydrochloride)staining procedure to visualise various neuronal populations within the basal ganglia via the binding of primary antibodies raised against specific marker proteins (Table 4-6). Immunocytochemical staining was performed on individual sets of free floating coronal sections spanning the region of interest. Sections were removed from the cyroprotectant solution and washed overnight in PBS. To block any endogenous peroxidase activity I first incubated the sections in a solution of 1% hydrogen peroxide, 50% methanol for 10mins and then washed four times in PBSTriton (PBS with 0.2% Triton X-100). The primary antibodies (Table 4-6) were diluted in PBSTriton, either with goat serum as a blocking agent or without any blocking serum. Goat serum (GIBCO®) was used as a blocking agent for all antibodies except for when the primary antibody was raised in goat. Sections were incubated in the diluted primary antibody overnight at room temperature with gentle agitation on a rocking table. Following overnight incubation the sections were washed four times in PBS-Triton and incubated with an appropriate secondary antibody diluted 1:500 in PBS-Triton with or without goat serum. All secondary antibodies were biotinlyated antibodies raised against the host species of the primary antibody (Appendix B.4). The sections were incubated with the secondary antibodies for 2-3 hours at room temperature on a rocking table, washed four times with PBS-Triton and incubated for a further 2-3 hours with Extravidin peroxidise (Sigma-Aldrich) diluted 1:500 in PBS-Triton. Sections were washed four times in PBS before staining in DAB or DAB-nickel sulphate solution. After staining the sections were washed in PBS and mounted onto polysine-coated glass microscope slides (Biolab Scientific, NZ). The slides were dried overnight, serially dehydrated in 70%, 90% and 100% ethanol, delipidified in xylene and coverslipped with histomount mounting medium (DPX; Sigma-Aldrich). Marker Antibody Dilution Supplier HA-epitope HA.11 1:1000 Covance 97 Neuroprotective Study Methods Luciferase Luciferase 1:10000 Chemicon Neurons NeuN 1:1000 Chemicon Striatal Neurons DARPP-32 1:750 Chemicon Striatal Neurons Egr-1 (krox-24) 1:1000 Santa Cruz Dopaminergic Neurons Tyrosine hydroxylase 1:500 Chemicon Pallidal Neurons Parvalbumin 1:5000 Swant Cortical Neurons SMI32 1:10000 Sternberger Monoclonals Table 4-6 Primary antibodies for DAB-staining immunocytochemistry Primary antibodies and working dilutions for immunocytochemical analysis of different neuronal populations and the localisation of transgene products in fixed free-floating brain sections. Additional antibody details in Appendix B.5. 4.7.3 Fluorescent immunocytochemistry analysis Double labelling of striatal sections was performed by fluorescent immunocytochemistry to visualise the expression of cellular markers – NeuN, DARPP-32, krox-24 and GFAP – and the relative localisation of transgenic protein expression. Striatal sections were removed from the cyroprotectant solution and washed overnight in PBS. The sections were then incubated overnight at room temperature with pairs of primary antibodies diluted in PBS-Triton with 1% blocking serum (Table 4-7). Following overnight incubation the sections were washed four times in PBS-Triton and incubated in Alexa Fluor® secondary antibodies (Invitrogen) diluted 1:500 in PBS-Triton with 1% goat or donkey serum (Table 4-7). After washing in PBS the sections were mounted onto uncoated glass slides and coverslipped with Cytifluor (Agar Scientific) for analysis with confocal microscopy. Primary antibodies Dilution NeuN 1:1000 DARPP-32 or krox-24 1:500 / 1:1000 Luciferase 1:20000 NeuN or GFAP 1:1000 / 1:500 Luciferase 1:20000 DARPP-32 or krox-24 1:500 / 1:1000 HA 1:1000 DARPP-32 or krox-24 1:500 / 1:1000 Serum Goat Donkey Donkey Goat 98 Alexa Fluor® 2° antibodies 594 anti-mouse 488 anti-rabbit 594 anti-goat 488 anti-mouse 594 anti-goat 488 anti-rabbit 594 anti-mouse 488 anti-rabbit Neuroprotective Study Methods Table 4-7 Antibodies for co-immunofluorescent labelling Pairs of primary antibodies used for co-labelling of neurons, working antibody dilutions, blocking serum (Goat, GIBCO®; Donkey, Sigma-Aldrich), secondary Alexa Fluor® conjugated antibodies raised in goat (Molecular Probes™, Invitrogen). Further antibody details in Appendix B.5. 4.7.4 Microscopy and stereology Light microscopy analysis and stereological quantification was performed using a Nikon Eclipse E800 microscope coupled with StereoInvestigator® software (MicroBrightField) and MicroFire™ S99808 digital camera (Optronics). Staining intensity analysis was performed on captured images using ImageJ (National Institutes of Health, USA). Presented images were compiled in Adobe® Photoshop® (Adobe). Confocal analysis of NeuN, DARPP-32 and HA staining confirmed full penetration of the antibodies through the tissue slices (data not shown). 4.7.4.1 Striatal Neuron Stereology DARPP-32 immunopositive neurons within the striatum were counted stereologically by unbiased optical fractionator sampling procedures. For the first investigation I assessed the striatum over twelve equispaced (320µm) coronal sections (sectioned at 40µm) extending from the head of the striatum at bregma +2.4 mm to -1.8 mm. The striatum was delineated on each section under a 2× objective using the lateral ventricle, corpus callosum, internal capsule and globus pallidus to help define the striatal borders. Medium spiny projection neurons expressing DARPP-32 were estimated by systematic sampling from a random starting point using a 60× objective lens. The optical fractionator was set to sample at intervals of x = 600µm, y = 700µm with a counting frame x = 90µ m, y = 70µm, dissector height z = 7µm; giving a section sampling fraction (ssf) of 1/8, area sampling fraction (asf) of 1/67 and thickness sampling fraction (tsf) of 1/1.4. In the second investigation I restricted the striatal neuronal estimates to six serial coronal sections (40µ m sections equispaced at 320µ m) covering the site of AAV vector and QA striatal injection (bregma +1.8mm to -0.5mm). Optical fractionators were set with grid spacing x = 650µm, y = 650µm and counting frame x = 90µ m, y = 70µm, z = 8µm (ssf = 1/8, asf = 1/67, tsf = 1/1.25). Coefficient of Error values were calculated using Gundersen equations, m = 1. Subsequent stereological analysis of krox-24 expressing striatal neurons in the second investigation was undertaken over eight coronal sections (40µm sections equispaced at 320µm) through the striatum (bregma +2.1mm to -0.8mm). Optical fractionators were set with grid spacing x = 650µ m, y = 650µm and counting frame x = 70µm, y = 70µm, z = 7µm (ssf = 1/8, asf = 1/86, tsf 1/1.4). 99 Neuroprotective Study Methods 4.7.4.2 Striatal Atrophy The extent of striatal atrophy was determined from the coronal section contours outlined for stereological counting of DARPP-32 neurons (Section 4.7.4.1). Total cross-sectional area of the ipsilateral striatum was calculated as a proportion of the contralateral striatal area. 4.7.4.3 Striatal TH-Staining Intensity Maintenance of TH in the striatum was quantified by density measurements of TH-immunostained sections using ImageJ. Images of the ipsilateral and contralateral hemispheres from eight equispaced (320µm spacing) coronal sections through the striatum (bregma +2.1mm to -0.8mm) were captured under a 2× objective and the striatal regions outlined as above (Section 4.7.4.1). Integrated areadensity measurements of TH-staining in the treated striatum relative to the adjacent contralateral striatum were averaged across the eight sections to give the relative maintenance of striatal TH expression. 4.7.4.4 Maintenance of Striatonigral Projections Innervations of the substantia nigra by DARPP-32 containing fibres were also quantified by density measurements. Three equispaced (320µm spacing) coronal sections through the substantia nigra (bregma -4.9mm to -5.9mm) were photographed under a 4× objective and both ipsilateral and contralateral nigral regions outlined with ImageJ. Integrated area-density measurements of DARPP32 staining in the treated nigra relative to the adjacent contralateral nigra were averaged across the three sections to determine relative maintenance of the DARPP-32 positive fibre innervations. 4.7.4.5 Pallidal Neuron Stereology Parvalbumin expressing neurons in the globus pallidus were estimated by optical fractionator following delineation of the GP region on four equispaced (320µm spacing) coronal sections (sectioned at 40µm) between bregma -0.5mm to -1.8mm. Optical fractionator sampling parameters were set to grid spacing of x = 200µm, y = 300µm and counting frame x = 100µm, y = 100µm, z = 7µ m (ssf = 1/8, asf = 1/6, tsf = 1/1.4). 4.7.4.6 Confocal Microscopy To assess co-labelling of striatal cells following immunofluorescent staining I used a Leica TCS SP2 laser scanning confocal microscope (Biomedical Imaging Resource Unit, The University of Auckland) to captured single optical slice images of fluorescently stained striatal cells under a 63× objective lens. 100 Neuroprotective Study Methods 4.8 Statistical Analysis All statistics were performed with GraphPad Prism® (version 4). Unless otherwise stated all data presented represents group mean ± standard error of the mean (SEM). Non-parametric Mann-Whitney tests were performed to assess the level of therapeutic transgenic protein expression following AAV-vector delivery relative to endogenous protein expression in AAV-Luciferase treated control striata and non-treated contralateral striata. Data collected from behavioural testing was analysed by paired t-test to assess development of QA induced functional deficits relative to baseline testing (Section 3.3.4.1), or two-way repeated measures (RM) ANOVA to gauge the impact of therapeutic AAV vector delivery on QA-induced deficits over the post-QA assessment period of the studies relative to controls. Un-paired t-tests were used to assess any significant alterations imparted by AAV-vectors relative to controls prior to QAlesioning. Statistical analysis of all neuropathological changes – stereological cell counts, striatal atrophy, and immunostaining density – were all undertaken by un-paired t-tests between the treatment groups and control rats, thereby treating each individual vector as an independent investigation. Correlations between functional behaviour and neuropathological alterations were performed by calculating each rats mean forelimb asymmetry score and “corridor” task retrieval score across all post-QA trials to give each rat an overall behavioural score for each test that could be correlated with the rats’ end-point pathological assessments. For apomorphine-induced rotational behaviour all individual post-QA trials were included in the correlation analysis. Pearson correlation coefficients were performed to determine any overall trends independent of treatment. Linear regression or Deming (Model II) linear regressions were calculated for the individual treatment groups to assess individual correlations and any significant alterations due to vector delivery. In all assessments the AAV-Luciferase vector control and PBS vehicle control data was grouped for analysis provided they were not significantly different at the 5% level (P > 0.05). One-way ANOVA with Bonferroni’s post-hoc tests or non-parametric Mann-Whitney analysis were used to analyse the earlier functional protein assays verifying transgenic protein expression following AAV-vector transduction. 101 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Chapter 5 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.1 Overview BDNF and GDNF were investigated as potential neuroprotective agents against excitotoxic insult by infusing an AAV vector encoding either BDNF or GDNF directly into the rodent striatum to enhance localised neurotrophic factor expression prior to a unilateral intrastriatal injection of the excitotoxic glutamate analogue QA. Building on previous literature that has reported protective benefits associated with BDNF (Martinez-Serrano and Bjorklund 1996; Bemelmans et al. 1999; PerezNavarro et al. 2000a; Kells et al. 2004) and GDNF (Perez-Navarro et al. 1996; Araujo and Hilt 1997; McBride et al. 2003; Kells et al. 2004), but which has indicated limitations with achieving efficient in vivo delivery procedures, I constructed AAV vectors with the intention of directing high level striatum targeted transgenic protein expression to support the striatal projection neurons and prevent associated functional deficits. Unfortunately following AAV-BDNF delivery some of the rats displayed detrimental behaviour, presumably due to excessive BDNF expression, resulting in early termination of this trial. Despite the adverse reaction I felt AAV-BDNF delivery still had sufficient merit as a therapeutic agent if these side effects could be avoided so conducted a second trial with a lower-titre of the AAV-BDNF vector. Continuous expression of the neurotrophic factors following AAV-mediated delivery showed some attenuation of the behavioural deficits resultant of QA lesioning however these functional benefits only had limited correlation with BDNF-derived maintenance of the basal ganglia. Overall these investigations demonstrated controlled delivery of neurotrophic factors could provide symptomatic relief without necessarily supplying significant neuroprotection to reduce the extent of neuronal cell death. 5.2 Procedures Chimeric AAV1/2 vectors were constructed containing cDNA encoding BDNF or GDNF and were assessed with in vitro assays to verify they correctly directed the expression of biologically functional neurotrophic factors (Section 2.3; (Kells et al. 2006)) prior to checking in vivo 102 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF transduction and transgene expression in the rodent striatum. Following the AAV vector testing I undertook two in vivo studies, conducted as described (Chapter 4) to assess the influence of BDNF and GDNF expression on QA-induced striatal excitotoxicity and associated functional behavioural deficits. In the initial study I assessed the delivered of AAV-BDNF (n = 7*) or AAV-GDNF (n = 10) to a single stereotaxic site within the rodent striatum prior to QA injection at the same stereotaxic coordinates. Following adverse reactions resulting from the AAV-BDNF delivery I conducted a second follow-up investigation with a ten-fold lower titre of AAV-BDNF (n = 9) delivered to two separate striatal sites flanking the QA injection site. An additional group of rats (n = 5) that received reduced-titre AAV-BDNF in this second study were assessed without QA lesioning to directly monitor the functional impact of AAV-mediated BDNF expression on sensorimotor performance. Control rats for each study received either AAV-Luciferase (Initial: n = 8; Follow-up: n = 5) or sterile PBS (Initial: n = 10; Follow-up: n = 5) prior to QA lesioning. 5.3 Animal Welfare Throughout all in vivo investigations the only rats that were observed to display any abnormal health issues were rats that received AAV-BDNF in the initial neuroprotective study. Starting five weeks after AAV-BDNF infusion – two-weeks post-QA – a number of these rats began to display excessive hyperactivity and aggression in their home cages, weight loss and seizure activity. In accordance with predetermined animal welfare guidelines the rats were euthanised if they had greater than 20% weight loss or ongoing seizures. The AAV-BDNF study was prematurely terminated seven weeks post AAV delivery. No indication of any negative effects were noted during earlier in vivo vector testing trials suggesting the health concerns were resultant of consistent long-term BDNF overexpression and therefore were not evident during the first three weeks post-AAV vector delivery in which I had conducted all prior assessment. It was not determined whether these welfare issues were due to the level of BDNF expression, the distribution within the brain, or a combined effect, but they nevertheless highlight the challenges associated with the administration of neurotrophic factors as therapeutic agents for neurological disorders. To avoid further welfare concerns but proceed with investigation of BDNF as a neuroprotective agent using the existing AAV-BDNF vector, I reduced the intensity of BDNF expression by lowering the vector titre 10-fold and splitting gene delivery over two separate stereotaxic sites within the striatum. * n values here represent the final number of rats included in the analysis of the investigations (Section 4.3). 103 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.4 In Vivo AAV Vector Testing To assess the spread of in vivo AAV vector transduction and neurotrophic factor transgene expression prior to investigating the therapeutic efficacy of BDNF or GDNF expression, I injected naïve rats using the same stereotaxic surgical delivery procedures to be performed on the main cohorts of rats in the neuroprotective investigations. In the first instance, the surgical vector delivery protocol was determined from the earlier AAV-Luciferase delivery testing (Section 4.4.2), which I then modified for the second lower titre AAV-BDNF two-site injection study designed to reduce the expression level but maintain a wide distribution of transduced cells within the striatum. Three weeks post-AAV vector delivery transgenic protein expression was visualised by immunocytochemical staining against an HA-epitope attached to the C-terminal of the expressed neurotrophic factors. A B C D Figure 5-1 Spread of undiluted AAV-BDNF transduction and BDNF expression (A) Representative coronal sections of a rat brain showing the main regions of HA-tagged BDNF expression three weeks after a single site 4.0 µL striatal injection of the undiluted AAV-BDNF vector. Cells expressing HA-tagged BDNF were largely contained within the injected striatum where a large population of the striatal neurons appeared to be transduced. Higher-power images of AAV-BDNF transduced neurons in (B) the striatum, (C) globus pallidus, and (D) substantia nigra. Scale bar = 2mm for whole sections (A); 100µm for higher magnification images (B-D). 104 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Following the high-titre AAV vector injections, transduced cells were observed in an extensive but often irregularly dispersed distribution throughout the rostral-caudal extent of the striatum (Figure 5-1, Figure 5-2). A population of cells within the globus pallidus and substantia nigra pars compacta ipsilateral to the injected striatum were HA-positive indicating transportation of the AAV-vectors and transduction of cells in the striatal target nuclei. The ipsilateral substantia nigra pars reticulata also displayed HA-immunostaining, although this was mainly restricted to the striatonigral axonal fibres with very few identifiable HA-positive cell bodies. Ten-fold dilution of the AAV-BDNF vector clearly restricted the extent and distribution of cellular transduction with the majority of HA-positive cells located close to the site of injection (Figure 5-3). The transduction of cells within the substantia nigra and globus pallidus was also diminished with only a few weakly HA-immunopositive cell bodies observed (Figure 5-3 C, D). A greater 100-fold A B C D Figure 5-2 Spread of AAV-GDNF transduction and GDNF expression (A) Representative coronal sections of a rat brain showing the main regions of HA-tagged GDNF expression three weeks after a single site 4.0 µL striatal injection of the AAV-GDNF vector. Cells expressing HA-tagged GDNF were largely contained within the injected striatum where a large population of the striatal neurons appeared to be transduced. Higher-power images of AAV-GDNF transduced neurons in (B) the striatum, (C) globus pallidus, and (D) substantia nigra. Scale bar = 2mm for whole sections (A); 100µ m for higher magnification images (B-D). 105 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF A B C D Figure 5-3 Spread of the reduced-titre AAV-BDNF transduction and BDNF expression (A) Representative coronal sections of a rat brain showing the main regions of HA-tagged BDNF expression three weeks after striatal injection of the AAV-BDNF vector diluted ten-fold. Extensive transduction of the striatal cells was seen in the proximity of the intrastriatal injection site and directly adjacent to the corpus callosum. Higher-power images of AAV-BDNF transduced neurons in (B) the striatum, (C) globus pallidus, and (D) substantia nigra. Scale bar = 2mm for whole sections (A); 100µm for higher magnification images (B-D). dilution of the AAV-BDNF vector almost completely eliminated striatal transduction detectable by HA-immunocytochemistry, with only a few isolated HA-positive cells visually detectable close to the site of injection. 5.5 BDNF and GDNF Expression Level To quantify the level of BDNF and GDNF expression generated following AAV-mediated gene transfer, cohorts of rats were euthanised three weeks after AAV vector delivery and their ipsilateral and contralateral striatal tissue separately isolated for ELISA-based quantification of the total BDNF and GDNF protein. Expression was assessed three weeks post-AAV delivery to provide a measure of neurotrophic factor expression equivalent to that expected to be present in the neuroprotective 106 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF study group of rats at the time of QA injection. BDNF expression in the striatum following undiluted AAV-BDNF injection was enhanced ~1000 fold to 46 ± 7 ng/mg total protein compared with the 0.049 ± 0.006 ng/mg total protein detected in control AAV-Luciferase injected striatum (P < 0.01; Figure 5-4A). The contralateral striatum displayed a slight increase in BDNF expression to 0.12 ± 0.02 ng/mg total protein (P < 0.05 compared to AAV-Luciferase contralateral striatum 0.064 ± 0.007 ng/mg total protein) suggesting a small amount of transgene expression may have crossed to the non-injected hemisphere. The lower titre AAV-BDNF injections resulted in a ~150-fold increase to 4.2 ± 0.7 ng/mg total protein compared to 0.026 ± 0.002 ng/mg total protein in the contralateral striatum. AAV-GDNF administration induced a similar increase in GDNF expression with GDNF levels increased to 26 ± 6 ng/mg total protein in the injected striatum and 0.063 ± 0.005 ng/mg total protein in the contralateral striatum (Figure 5-4B). Striatal expression of GDNF protein within the control AAV-Luciferase treated rats was below the reliable detection limit of the ELISA assay. A B Figure 5-4 ELISA quantification of BDNF and GDNF expression in the striatum The level of BDNF (A) or GDNF (B) protein expression in striatal tissue isolated three weeks after infusion of AAV-BDNF, AAV-GDNF or AAV-Luciferase was quantified by ELISA. Total protein in the samples was measured by BCA assay to normalise the individual samples. The GDNF expression in the AAV-Luciferase treated rats was considered to be below reliable detection levels. Mann-Whitney tests: comparison with AAV-Luciferase * P < 0.05, ** P < 0.01; comparison with high-titre AAV-BDNF ‡ P < 0.01. No statistics were performed on the GDNF expression levels. 5.6 Functional Behaviour Assessment Throughout the neuroprotective studies I assessed the development of functional deficits by analysing the performance of each rat in behavioural tests designed to show hemispherical imbalances in brain function arising as a result of unilateral lesioning in the basal ganglia. The functional behaviour tests undertaken as described earlier (Section 4.5) involved spontaneous 107 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF forelimb use analysis, drug-induced rotational behaviour following either apomorphine or amphetamine dosing and preferential left / right side food selection. All the rats were assessed with baseline measurements taken both before and after AAV delivery and for 5-7 weeks post-QA injection (Table 4-2 and Table 4-3). 5.6.1 Spontaneous forelimb use Deficits in functional motor performance were assessed by observing spontaneous forelimb usage during exploratory rearing behaviour. Delivery of the AAV vectors or PBS did not induce any significant alterations in baseline forelimb usage assessed prior to the QA injection, although both neurotrophic factor treated groups – AAV-BDNF and AAV-GDNF – were tending towards an increased preference to use their contralateral forelimb (Figure 5-5A,B). This slight contralateral preference resulted in a small divergence in forelimb use between the control and neurotrophic factor treated rats reaching statistical significance with lower-titre AAV-BDNF treated rats in the follow-up study (Unpaired t-test P < 0.05). Following unilateral QA administration the AAV-Luciferase and PBS treated control rats showed an immediate shift towards preferentially using their ipsilateral forelimb for spontaneous exploration as expected from the earlier testing (Section 3.3.4). This ipsilateral bias was however only observed during the first two-weeks following QA injection in the initial study (Figure 5-5A), but maintained throughout the seven-week post-QA assessment period in the second study (Figure 5-5B). In contrast, the preferential forelimb use by AAV-BDNF or AAV-GDNF treated rats was not significantly affected by the QA injection in either investigation (Figure 5-5A,B). AAV-GDNF treated rats maintained baseline preferences of non-biased spontaneous forelimb use, significantly ameliorating the QA-induced ipsilateral bias (Two-way RM ANOVA P < 0.05; Figure 5-5A). Following QA injection the AAV-BDNF treated rats in the initial high expression study continued to develop a persistent contralateral forelimb preference in contrast to the immediate preferential ipsilateral forelimb shift in the control rats (Two-way RM ANOVA P < 0.001; Figure 5-5A). Subsequent BDNF investigation following ten-fold dilution of the AAV-BDNF vector eliminated the ongoing development of a contralateral forelimb bias beyond the initial divergence from the control group induced by AAV-BDNF transduction prior to QA injection (Figure 5-5B). Following QA injection into the striatum of reduced-titre AAV-BDNF treated rats, the acquirement of a slight contralateral forelimb use preference was reversed with a small shift towards greater ipsilateral forelimb usage, such that overall the group remained largely unbiased in contrast to the significant ipsilateral preference of control rats (Two-way RM ANOVA P < 0.05; Figure 5-5B). The extra 108 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF cohort of non-lesioned AAV-BDNF treated rats displayed a small shift towards contralateral forelimb use, but with no on-going changes the continuous induced expression of BDNF at this lower level did not appear to directly influence forelimb motor control (Figure 5-5B). A B Figure 5-5 Spontaneous ipsilateral forelimb use asymmetry score Plots displaying changes in group ipsilateral asymmetry scores quantified from forelimb use during exploratory rearing behaviour in (A) the initial AAV-BDNF / AAV-GDNF and (B) AAV-BDNF follow-up investigations. Ipsilateral asymmetry scores represent the combined usage of the ipsilateral forelimb, as a percentage of the total left and right forelimb use, for rearing, initial contact of the cylinder wall during an exploratory rear and landing. Un-paired t-test between treatment groups: * P < 0.05. Two-way RM ANOVA relative to the control group: † BDNF P < 0.001, GDNF P < 0.05; ‡ BDNF P < 0.05, BDNF – no QA P < 0.001. 109 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF In addition to the forelimb asymmetry scores, the level of exploratory activity in the cylinder was assessed showing general reduction in the number of exploratory rears made during the five minute observational period with sequential trials. Both AAV-BDNF and AAV-GDNF treated rats were observed to maintain a significantly higher level of exploratory activity following QA administration with the initial high-titre AAV-BDNF treated rats displaying full maintenance of exploratory behaviour (Figure 5-6A). A higher level of activity was recorded in the second study for both control and neurotrophic factor treated rats, which was likely to have been consequential of the dietary restriction regime in the second investigation or adjustment to the home cage environment which enhanced their level of exploratory behaviour (Figure 5-6B). A B Figure 5-6 Spontaneous exploratory rearing activity Plots displaying the number of exploratory rears in the assessment cylinder during the five minute analysis of spontaneous forelimb use in (A) the initial AAV-BDNF / AAV-GDNF and (B) AAVBDNF follow-up investigations. No statistics were performed on the pre-QA baseline data. Two-way RM ANOVA relative to the control group: † GDNF P < 0.05, BDNF P < 0.01; ‡ BDNF P < 0.05, BDNF – no QA P < 0.01. Overall the single administration of either AAV-BDNF or AAV-GDNF resulted in a significant amelioration of the ipsilateral forelimb use bias induced by a unilateral striatal lesion with high BDNF expression actually leading to predominant use of the contralateral forelimb. The apparent recovery of the control group towards un-biased forelimb use in the initial study was in contrast to the increasing bias in the second follow-up investigation. An explanation for this conflicting observation was not evident although the rats in the second study also exhibited a higher level of exploratory rearing behaviour, potentially due to the dietary restriction of the rats in the second study. 110 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.6.2 Drug-induced rotational behaviour The extent of rotational behaviour induced by apomorphine or amphetamine was undertaken to assess imbalances in the dopamine system following unilateral intrastriatal QA injection and the subsequent loss of striatal neurons expressing dopamine receptors. Results of both the apomorphine and amphetamine induced rotational behaviour were difficult to interpret due to the inconsistency of rotational direction between individual rats within the treatment groups which was possibly resultant from slight variations in the positioning of the striatal lesion. 5.6.2.1 Apomorphine Apomorphine administered subcutaneously acts as a non-specific dopamine receptor agonist stimulating dopaminoceptive neurons and thereby inducing rotational motor behaviour generally towards the lesioned striatum (Section 3.3.4). The prevalence of apomorphine induced rotational behaviour varied greatly between individual rats with rotation rates ranging from no rotational behaviour through to 20 rotations per minute. Within each group some rats displayed a contralateral rotational dominance in contrast to the majority of control treated rats that rotated towards the lesioned hemisphere (Figure 5-7). Although there were no significant differences between the treatment groups in terms of total rotations, I did observe an overall trend towards increased rotational behaviour after QA injection (Figure 5-8). The additional non-lesioned AAV-BDNF rats in the follow-up study also tended to display increased rotational behaviour in spite of not being lesioned and in contrast to the AAV-BDNF treated rats that were QA lesioned. For reliable assessment of rotational behaviour I restricted the analysis of ipsilateral rotation percentage to rats that had greater than 0.5 rotations per minute for apomorphine. Having selectively treated the rats on their naturally dominant striatal hemisphere – determined following the baseline testing – the initial baseline rotational behaviour was generally in an ipsilateral direction less than 50% of the time (Figure 5-7). Apomorphine administration after AAV vector delivery but prior to QA injection resulted in a large distribution in the percentage of ipsilateral rotations for both neurotrophic factor – AAV-BDNF and AAV-GDNF – and the AAVLuciferase or PBS control treated groups. The majority of AAV-BDNF treated rats were however observed to switch rotational direction following AAV-BDNF injection with the increase in striatal BDNF protein expression causing the rats to predominantly rotate towards their treated hemisphere. This change in rotational direction was particularly pronounced in the initial AAV-BDNF trial which had the highest BDNF expression levels (Figure 5-7A). The induction of ipsilateral rotational behaviour following BDNF expression was also observed in the smaller cohort of reduced-titre 111 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF AAV-BDNF treated rats in the follow-up study which were not selected for QA lesioning. However, five weeks following AAV-BDNF delivery the non-lesioned rats had reverted to a contralateral rotational dominance suggesting modifications in the dopamine system following continuous expression of BDNF in the striatum (Figure 5-7B). Following the QA lesion apomorphine-induced rotational behaviour became more uni-directional in all animals although it was not always in an ipsilateral direction. While the majority of the control treated rats exhibited persistent ipsilateral rotational behaviour, the AAV-BDNF and AAV-GDNF treated groups were more evenly divided between predominantly displaying contralateral or ipsilateral rotational behaviour. With the lack of a consistent rotational direction between similarly treated rats I did not perform any statistical analysis on the rotational data. A B Figure 5-7 Apomorphine-induced rotational behaviour Plots displaying each rats rotational behaviour towards the treated side of their brain as a percentage of total rotations following s.c. administration of 1mg/kg apomorphine in (A) the initial AAV-BDNF / AAV-GDNF and (B) AAV-BDNF follow-up investigations. Rats that had less than 0.5 rotations per minute were excluded from this analysis. Lines represent the median result for each group. 112 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF A B Figure 5-8 Total apomorphine-induced rotations Plots displaying the mean rate of apomorphine-induced rotational behaviour for each group in (A) the initial AAV-BDNF / AAV-GDNF and (B) AAV-BDNF follow-up investigations following s.c. administration of 1mg/kg apomorphine. No significance was found between treatment groups and controls post-QA. 5.6.2.2 Amphetamine Amphetamine was only used to induce rotational behaviour in the initial high-titre AAV-BDNF and AAV-GDNF study. Wide variation in the rotational response of individual rats was observed during baseline testing with the selection of the dominant hemisphere for treatment skewing this into an overall contralateral rotational preference (Figure 5-9A). The average rate of rotation also varied greatly with many rats not showing any induction of rotational behaviour, therefore I restricted the analysis of rotational direction to rats that displayed greater than an average 0.1 total rotations per minute over the 60 minute testing period. Following AAV-vector delivery, but prior to QA injection, the enhanced expression of BDNF and GDNF significantly increased the amphetamine induced rotation rate compared with the control treated rats (BDNF P < 0.05; GDNF P < 0.01; Figure 5-9B) A majority of the AAV-BDNF and AAV-GDNF treated rats preferentially rotated towards their treated side following amphetamine administration, while control treated rats showed very little rotational behaviour. Post-QA injection the majority of rats from all treatment groups immediately displayed largely unidirectional rotational behaviour – predominantly in an ipsilateral direction although a few rats preferentially rotated away from their lesioned hemisphere. Over the sequential trials amphetamine-induced rotational behaviour of the control and AAV-GDNF treated rats became more polarised becoming purely unidirectional at week six. In contrast, AAV-BDNF treated rats displayed much greater variation in rotation direction than the control group with only a few rats acquiring complete unidirectional behaviour and displaying a return towards bidirectional rotational behaviour at four weeks post-QA. The BDNF rats were removed from this initial study five weeks after QA administration and amphetamine-induced rotations were not assessed in the lower AAVBDNF follow-up study so I was unable to determine whether this observed return towards equal 113 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF directional rotational behaviour was enhanced or maintained at later time points. Statistical analysis was not performed on the directional rotation data due to the inconsistency in the direction of induced rotation following QA lesioning. A B Figure 5-9 Amphetamine-induced rotational behaviour Plots displaying the rotational behaviour induced following an i.p. injection of 5mg/kg of amphetamine in the initial AAV-BDNF / AAV-GDNF investigation. (A) Individual rats’ rotational behaviour towards the treated (ipsilateral) side of their brain as a percentage of total rotations. (B) Mean rate of rotational behaviour for each group. Rats that had less than 0.1 rotations per minute were excluded from the directional analysis. Lines in (A) represent the median result for each group at each time point. Post-AAV – Un-paired t-test: * P < 0.05, ** P < 0.001. Post-QA – Two-way RM ANOVA: † BDNF P < 0.05; GDNF P = 0.055. 5.6.3 Sensorimotor “corridor” task The impact of AAV-BDNF delivery on the development of sensorimotor neglect was investigated in the second in vivo study by analysing the rats’ performance in the “corridor” task. During baseline assessment the rats displayed a fairly even distribution of sugar pellet retrievals from the left and right sides of the corridor with the selection of the dominant hemisphere for treatment skewing the group data slightly towards a preferential retrieval of pellets from the contralateral side of the 114 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF corridor (Figure 5-10). Delivery of the AAV vectors did not alter the percentage of ipsilateral retrievals for the control rats or the main group of reduced-titre AAV-BDNF treated rats. However, in contrast to the main treatment groups, the additional group of rats included to assess AAV-BDNF treatment in the absence of a QA lesion did display a small but significant increase in the retrieval of sugar pellets located on the ipsilateral side of the corridor post-AAV delivery (Paired t-test P < 0.05), although this shift did not result in a significant ipsilateral retrieval bias (Not significantly above 50%; Wilcoxon signed-rank test P > 0.05) and remained fairly consistent throughout the full testing period. Following QA administration the control rats rapidly acquired a strong ipsilateral retrieval bias that persisted throughout the seven week investigative period. In stark contrast the AAV-BDNF treated group did not show significant development of an ipsilateral retrieval preference post-QA remaining unchanged from baseline preferences and significantly separated from the control group (Two-way ANOVA post-QA P < 0.01). Figure 5-10 Preferential food selection in the sensorimotor “corridor” task Plot displaying group changes in the number of sugar pellet retrievals from the same side as the treated striatum as a percentage of the 20 retrievals per trial. Two-way RM ANOVA relative to the control group: † BDNF P < 0.01; BDNF – no QA P < 0.05. 5.7 Immunocytochemical Analysis On completion of the functional behavioural assessment I analysed the post-mortem brain tissue to visualise and quantitatively determine the extent of any neuroprotection imparted by the AAVBDNF or AAV-GDNF vector delivery prior to QA-induced excitotoxic insult. Immunocytochemical staining was performed on individual series of coronally sliced sections equispaced at 320µ m intervals through the caudate-putamen portion of the striatum, the globus pallidus and substantia nigra to identify specific populations of basal ganglia neurons affected by the QA striatal lesion. 115 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.7.1 AAV-mediated transgene expression and QA lesioning of the striatum To assess the level of neuroprotective support imparted by AAV-mediated neurotrophic factor expression against QA-induced excitotoxic insult, I initially visualised the distribution of transgene expression by HA or Luciferase immunocytochemical staining and quantified the extent of QA striatal lesioning using DARPP-32 as a marker for the GABAergic striatal projection neurons. A * B C E F H I * * * HA D * * * * DARPP-32 G * * * * NeuN Figure 5-11 High-titre AAV-BDNF transduction and striatal cell maintenance Representative images from adjacent striatal sections showing AAV-mediated BDNF expression and neuronal cell loss five weeks post-QA. (A-C) HA-immunostaining displayed an irregular distribution of BDNF expression within the striatum resultant of undiluted AAV-BDNF transduction. Maintenance of striatal neurons as detected by (D-F) DARPP-32 and (G-I) NeuN-immunostaining was shown to inversely reflect the BDNF expression as identified by the arrows pointing to areas of high BDNF expression and asterisks in areas with no transgenic BDNF expression. Regions outlined by boxes are shown at higher magnification in figures immediately to the right. Scale bar = 1mm for (A,D,G), 200µm for (B,E,H), 50µm for (C,F,I). 116 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.7.1.1 Initial Study – AAV-BDNF and AAV-GDNF Delivery Using adjacent sections to compare the distribution of neurotrophic factor expression and the maintenance of striatal projection neurons identified by their intense expression of DARPP-32, I observed an inverse relationship between areas of extensive HA-tagged neurotrophic factor expression and DARPP-32 positive neurons. This inverse correlation was particularly strong in the AAV-BDNF treated rats as shown in Figure 5-11 where a well-defined portion of striatum is devoid of HA-immunoreactivity but displays complete maintenance of DARPP-32 positive neurons, despite its close proximity to the site of QA injection. Although less apparent, the opposite was also seen A B C E F H I * HA D * DARPP-32 G * NeuN Figure 5-12 AAV-GDNF transduction and striatal cell maintenance Representative images from adjacent striatal sections showing AAV-mediated GDNF expression and neuronal cell loss seven weeks post-QA. (A-C) HA-immunostaining displayed the distribution of GDNF expression within the striatum resultant of AAV-GDNF transduction. Maintenance of striatal neurons was detected by (D-F) DARPP-32 and (G-I) NeuN-immunostaining. A large number of HA-positive GDNF expressing cells were observed within regions of the striatum that exhibited a complete loss of DARPP-32 staining (asterisks). Regions outlined by boxes are shown at higher magnification in figures immediately to the right. Scale bar = 1mm for (A,D,G), 200µm for (B,E,H), 50µ m for (C,F,I). 117 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF with a reduction of DARPP-32 immunopositive cells in the dispersed areas of intensive BDNF expression, but more maintained in adjacent portions of the striatum raising the question of DARPP32 down-regulation in AAV-BDNF transduced neurons. Overall this effect resulted in the AAVBDNF treated rats not having a clearly defined “lesion” area based on the absence of DARPP-32 immunopositive cells as I observed with the PBS and AAV-Luciferase control rats (Figure 5-13). Immunostaining for NeuN in an adjacent section appeared to indicate that while there was a definite reduction in the number of neurons it was not to the extent indicated by DARPP-32. A B C E F H I HA D DARPP-32 G NeuN Figure 5-13 AAV-Luciferase transduction and striatal cell maintenance Representative images from adjacent striatal sections showing AAV-mediated Luciferase expression and neuronal cell loss seven weeks post-QA. (A-C) Immunostaining for Luciferase shows the striatal expression resultant of AAV-Luciferase transduction. Maintenance of striatal neurons was detected by (D-F) DARPP-32 and (G-I) NeuN-immunostaining showing a fairly well defined region of QA-induced lesioning. The vast majority of Luciferase expressing cells were located outside of the lesion borders within intact segments of the striatum. Regions outlined by boxes are shown at higher magnification in figures immediately to the right. Scale bar = 1mm for (A,D,G), 200µm for (B,E,H), 50µm for (C,F,I). 118 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Similar to BDNF the AAV-GDNF treated rats displayed extensive HA-immunostaining of cells within areas of the striatum devoid of DARPP-32 cells, however the area of “lesion” was more clearly delineated and, in contrast to the BDNF rats, extended into regions of striatal tissue not showing any HA staining (Figure 5-12). Surprisingly, staining for the neuronal marker NeuN also displayed a lack of immunopositive neurons in the region of transgenic GDNF expression displaying a similar pattern of cell loss to DARPP-32. In general the maintenance of NeuN-positive cells appeared greater than DARPP-32-positive cells although I did not confirm this observation with stereological quantification. In AAV-Luciferase control treated rats the Luciferase expression was generally restricted to the areas of DARPP-32 staining although there was a scattering of Luciferasepositive cells remaining within the lesioned area that roughly correlated with the remaining NeuNpositive neurons (Figure 5-13). As with the PBS treated rats, AAV-Luciferase control rats displayed more uniformity in DARPP-32 striatal cell loss than the neurotrophic factor expressing rats. 5.7.1.2 Study 2 – Reduced Titre AAV-BDNF Delivery The inverse correlation between HA-detected BDNF expression and DARPP-32 expression seen in the initial high-titre study was still observed following the 10-fold dilution of the AAV-BDNF vector A HA B C E DARPP-32 NeuN D F Figure 5-14 Diluted AAV-BDNF administration and striatal cell maintenance Adjacent sections through the striatum showing AAV-mediated BDNF expression and the loss of striatal neurons eight weeks post-QA. (A,B) HA-immunostaining displayed the distribution of BDNF expression within the striatum resultant of reduced-titre AAV-BDNF transduction. Maintenance of striatal neurons was detected by (C,D) DARPP-32 and (E,F) NeuN-immunostaining. Regions outlined by boxes are shown at higher magnification in figures immediately below. Scale bar = 2mm for whole sections (A,C,E) and 200µm for higher magnifications (B,D,F). 119 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF and the two-site delivery procedure, although the correlation was less distinct with substantial areas of the striatum containing both HA-immunoreactive cells and DARPP-32 expressing neurons (Figure 5-14A-D). Additional staining for NeuN confirmed the presence of some neurons within areas of DARPP-32 loss further suggesting that the transduction with AAV-BDNF is leading to long-term down-regulation of DARPP-32 expression (Figure 5-14E, F). The control AAV-Luciferase and PBS treated rats continued to display a more regularly defined region of striatal lesioning, within which only a small population of DARPP-32 and NeuN-expressing cells remained (Figure 5-15). A HA B C E DARPP-32 NeuN D F Figure 5-15 Two-site AAV-Luciferase injection and striatal cell maintenance Adjacent sections through the striatum showing AAV-mediated Luciferase expression and the loss of striatal neurons eight weeks post-QA. (A,B) Immunostaining for Luciferase shows the striatal distribution of Luciferase expression resultant of the AAV-Luciferase transduction following twosite delivery. Maintenance of striatal neurons was detected by (C,D) DARPP-32 and (E,F) NeuNimmunostaining, displaying a reasonably well defined region of QA lesioning. The majority of Luciferase expressing cells were located outside of the lesion borders within the intact segments of the striatum. Regions outlined by boxes are shown at higher magnification in figures immediately below. Scale bar = 2mm for whole sections (A,C,E) and 200µm for higher magnifications (B,D,F). 5.7.1.3 Stereological Analysis – DARPP-32 To quantify the striatal damage induced by QA lesioning following delivery of AAV vectors encoding BDNF, GDNF, Luciferase or PBS I employed stereological cell counting procedures to estimate the number of DARPP-32 immunopositive neurons remaining within both the treated striatum and the intact contralateral striatal hemisphere. In the initial high-titre AAV-BDNF and 120 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF AAV-GDNF investigations, optical fractionator probes estimated the number of DARPP-32 positive neurons in the sampled region of the caudate-putamen at 1,400,000 ± 120,000 in high-titre AAVBDNF treated rats, 1,210,000 ± 90,000 in AAV-GDNF rats and 1,150,000 ± 120,000 in the AAVLuciferase / PBS control rats compared with an overall average of 2,180,000 ± 50,000 DARPP-32 positive cells in the contralateral intact striatum (Figure 5-16A; Coefficient of Error: Initial High-titre Neurotrophic Factor Investigation A B Follow-up Reduced-titre AAV-BDNF Investigation C D Figure 5-16 Maintenance of DARPP-32 positive striatal projection neurons Graphs displaying the actual and relative numbers of DARPP-32 positive neurons in the striatum as estimated by optical fractionator stereological analysis of the treated (ipsilateral) and intact (contralateral) striatal hemispheres in (A,B) the initial AAV-BDNF and AAV-GDNF investigation and (C,D) reduced-titre AAV-BDNF follow-up study. Population estimates were determined from 12 sections spanning 3.56mm of the striatum for the initial study and 7 sections spanning 1.96mm of the anterior striatum for the follow-up study. Relative cell counts represent the number of DARPP32 cells within the lesioned striatum as a proportion of cells present in the contralateral intact striatum. Statistical analysis revealed no significance differences between treatment groups at the 5% level (Unpaired t-test P > 0.05). 121 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF ipsilateral ≤ 0.04, contralateral ≤ 0.02). A smaller portion of the caudate-putamen centred around the QA lesion was analysed in the second investigation containing an estimated 1,530,000 ± 40,000 DARPP-32 positive neurons in the intact contralateral striatum that was reduced to 930,000 ± 100,000 in the reduced-titre AAV-BDNF treated striatum and 760,000 ± 60,000 in the AAVLuciferase / PBS controls (P = 0.18; Figure 5-16C; Coefficient of Error: ipsilateral ≤ 0.06, contralateral ≤ 0.03). Overall the proportion of cells expressing DARPP-32 in the QA lesioned striatum relative to the intact contralateral striatal hemisphere was reduced in control AAV-Luciferase / PBS treated rats to 52 ± 4 % in the initial study and 47 ± 4 % in the second follow-up investigation. Prior delivery of the AAV vectors to enhance neurotrophic factor expression at the time of QA lesioning was unsuccessful in significantly increasing the maintenance of DARPP-32 positive neurons. The proportion of DARPP-32 positive neurons remaining in the initial high-titre AAV-BDNF injected rats was estimated at 60 ± 4 % (t-test P = 0.28) and 58 ± 5 % in the AAV-GDNF treated group (t-test P = 0.35; Figure 5-16B). Dilution of the AAV-BDNF vector still failed to generate sufficient assistance to significantly protect the striatal neurons from QA-induced insult, despite increasing the relative maintenance of DARPP-32 neurons to 65 ± 8 % of the contralateral striatum (t-test P = 0.054; Figure 5-16D). 5.7.1.4 Striatal Atrophy In addition to the loss of striatal neurons I observed an apparent atrophy of the striatum and enlargement of the adjacent lateral ventricle (e.g. Figure 5-15). By analysis of the striatal area using the same contours drawn to perform stereological cell estimates, I quantified the relative volume of the striatum in the control rats to be 73 ± 2% of the contralateral striatum in the initial study (Figure 5-17A) and 75 ± 3% in the second follow-up investigation (Figure 5-17C). AAV-BDNF treatment significantly prevented striatal atrophy with complete maintenance of the striatal area in the initial high-titre study (103 ± 3%, t-test P < 0.0001) and only a 9% reduction in the follow-up reduced-titre AAV-BDNF investigation (91 ± 6%, t-test P < 0.05). In contrast, AAV-GDNF delivery did not significantly influence QA-induced atrophy compared with the control rats (79 ± 2%, t-test P = 0.12). Comparison between the striatal atrophy and the loss of DARPP-32 expressing striatal projection neurons showed a direct correlation with the striatum contracting in proportion to the loss of striatal neurons (Figure 5-17B,D). The correlation was significantly shifted by AAV-BDNF delivery in the initial high-titre investigation, with the striatum maintaining its full volume despite DARPP-32 cell loss equivalent to some of the control and AAV-GDNF treated rats (P < 0.001). 122 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Initial Neuroprotective Study Follow-up Reduced-titre AAV-BDNF Study A C B D Figure 5-17 Atrophy of the lesioned striatum Graphs displaying the size of the lesioned striatum as a proportion of the intact contralateral striatal area, and plots correlating striatal atrophy with DARPP-32 positive cell loss in the striatum. (A,B) Initial high-titre AAV-BDNF and AAV-GDNF investigation and (C,D) the follow-up reducedtitre AAV-BDNF study. Cross-sectional striatal area was measured on 12 coronal sections for the initial study and 7 sections for the second study. Unpaired t-tests: * P < 0.05, ** P < 0.0001. Linear regression analysis: R values represent the overall Pearson correlation coefficient; Solid lines represent significant non-zero linear correlations for individual treatment groups (P < 0.05). Comparison with controls: (A) BDNF: slope P = 0.81, elevation P < 0.001 (†); GDNF: slope P = 0.43, elevation P = 0.12; (B) BDNF: slope P = 0.88, elevation P = 0.34. 5.7.2 Krox-24 expression As an alternative to the DARPP-32 protein dependent analysis of neuronal survival, I also visualised striatal neurons in the reduced-titre AAV-BDNF follow-up investigation by their expression of krox24 / early growth response protein 1 (Egr1) (Herdegen et al. 1995; MacGibbon et al. 1995). An early response transcription factor, krox-24 is induced in neurons following neurotransmitter or trophic factor activation of the mitogen-activated protein kinase (MAPK) signalling pathway, subsequently regulating the expression of a diverse range of neuronal proteins and thereby contributing to neuronal 123 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF plasticity (Knapska and Kaczmarek 2004). Basal expression of krox-24 in rodent striatal neurons may provide a more reliable visualisation of surviving neurons than late-response gene products. Visual assessment of the normal rat striatum showed the vast majority of striatal neurons expressing NeuN also co-express both krox-24 and DARPP-32 (Figure 5-18). A late addition to the pathological analysis to complement DARPP-32 based quantification of striatal neurons, I did not retrospectively quantify krox-24 maintenance in the initial high-titre AAV-BDNF and AAV-GDNF investigations. A NeuN DARPP-32 B NeuN krox-24 Figure 5-18 Double-immunofluorescent labelling of striatal neurons Co-labelling of striatal neurons in the normal striatum with NeuN (red) and either (A) DARPP-32 or (B) krox-24 (green), demonstrated the vast majority of neurons expressing the general neuronal marker NeuN also express DARPP-32 and krox-24. All expression of DARPP-32 and krox-24 was localised to the NeuN-positive neurons. Scale bar = 30µm. The distribution of krox-24 expression within the AAV-BDNF treated striatum appeared similar to DARPP-32 immunostaining, although as a nuclear protein krox-24 staining was more distinct and had less variability in DAB staining intensity (Figure 5-19). Quantification of krox-24 immunopositive cells was performed using the optical fractionator stereology probe on eight 124 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF equispaced sections through the striatum (Figure 5-20A). An estimated 1,060,000 ± 110,000 krox-24 positive cells remained within the analysed region of the striatum in the control AAV-Luciferase / PBS treated rats, and 1,420,000 ± 180,000 in the reduced-titre AAV-BDNF treated rats (Coefficient of Error ≤ 0.05). The intact contralateral striatal regions contained an estimated 2,090,000 ± 60,000 krox-24 positive cells (Coefficient of Error ≤ 0.03). Overall I found similar maintenance of the krox24 positive striatal cells to that quantified by DARPP-32 analysis (Section 5.7.1.3), however the survival of krox-24 expressing neurons following AAV-BDNF at 68 ± 8% did demonstrate a significant level of protection when compared with the 50 ± 6% krox-24 cell maintenance in AAVLuciferase / PBS control rats (t-test P < 0.05; Figure 5-20B). Correlation of the krox-24 expression with DARPP-32 cell maintenance demonstrated a parallel link between the maintenance of krox-24 expression and DARPP-32 expression following QA lesioning that was independent of the prior AAV-BDNF or control treatment (Figure 5-20C). krox-24 A D AAV-BDNF AAV-Luciferase DARPP-32 B C E F Figure 5-19 Krox-24 expression in the striatum Representative images displaying the expression of krox-24 in striatal neurons following QA lesioning in (A,B) AAV-BDNF treated rats and (D,E) AAV-Luciferase treated rats. (C,F) Adjacent sections immunostained for DARPP-32 (copied from Figure 5-14D and Figure 5-15D) show the positive distribution correlation between krox-24 and DARPP-32 positive cells in the striatum. Regions outlined by boxes are shown at higher magnification in figures immediately to the right. Scale bar = 2mm for whole sections (A,D) and 200µm for higher magnifications (B,C,E,F). 125 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF A B C Figure 5-20 Maintenance of krox-24 expressing striatal neurons Graphs displaying (A) actual and (B) relative numbers of striatal neurons expressing krox-24 as estimated by optical fractionator stereological cell counting in the treated (ipsilateral) and intact (contralateral) striatal hemispheres in the follow-up, reduced-titre AAV-BDNF investigation. Relative cell counts represent the number of krox-24 expressing cells within the lesioned striatum as a proportion of krox-24 positive cells present in the contralateral striatum. Unpaired t-test: * P < 0.05. (C) Plot showing the correlation between the maintenance of krox-24 expressing neurons and DARPP-32 positive neurons in the striatum. Deming linear correlation: slope = 1.02 ± 0.10; r2 = 0.87, P < 0.0001. R value represents the overall Pearson correlation coefficient. 126 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.7.3 Analysis of transduced striatal cells To assess the phenotype of surviving AAV vector transduced cells within the striatum at the conclusion of the neuroprotective studies I performed double-immunofluorescent staining for transgene protein expression – Luciferase or HA – and either NeuN, DARPP-32, krox-24 or GFAP. Co-labelling of Luciferase and NeuN demonstrated that the vast majority of transduced cells were striatal neurons ( Figure 5-21A) although a small amount of co-localisation was also observed with GFAP ( Figure 5-21D), suggesting that while the AAV vectors preferentially transduced neurons they were capable of also transducing glial cells. Despite the vast majority of AAV-Luciferase transduced striatal cells expressing the general neuronal marker NeuN, expression of specific markers of striatal neurons – DARPP-32 or krox-24 – by the AAV vector transduced cells varied with the different transgene being expressed. No DARPP-32 or krox-24 expression was observed within Luciferase expressing cells ( Figure 5-21B,C). While the majority of AAV-BDNF transduced cells positively labelled for krox-24, the intensity of DARPP-32 expression appeared to be inversely related to the level of BDNF expression ( Figure 5-22). A similar pattern was also observed for the GDNF expressing cells with very few maintaining expression of DARPP-32, and the majority of AAV-GDNF transduced cells also displaying a large reduction in detectable krox-24 expression (Figure 5-23). Figures on the following pages Figure 5-21 Immunofluorescent analysis of AAV-Luciferase transduced cells Representative single-slice confocal images from an AAV-Luciferase treated rat from the second investigation showing AAV-Luciferase transduced cells (red) within the lesioned striatum and immunofluorescent staining for (A) NeuN-positive cells, (B) DARPP-32 expression, (C) krox-24 expression and (D) GFAP expression (all displayed in green). Few Luciferase-positive cells actually survived QA lesioning. (A) Co-labelling with NeuN was evident for most Luciferase expressing cells. AAV-Luciferase transduced cells very rarely co-expressed (B) DARPP-32 or (C) krox-24. (D) A few Luciferase-expressing cells appeared to co-localise with GFAP expression (arrows). Scale bar: 30µm Figure 5-22 Immunofluorescent analysis of AAV-BDNF transduced cells in the striatum Representative single-slice confocal images from AAV-BDNF treated rats showing HA-positive BDNF expressing cells within the striatum (red) together with immunofluorescent staining for (A-C) DARPP-32 or (D) krox-24 expression (both displayed in green). (A) Co-labelling with HA and DARPP-32 was only evident for some BDNF expressing cells in the undiluted AAV-BDNF study with many morphologically identical cells not appearing to express DARPP-32 (arrows). A greater proportion of HA-positive cells co-labelled in (B) the reduced-titre AAV-BDNF investigation, although DARPP-32 staining was still weaker for the HA-positive transduced cells than nontransduced cells, especially in (C) the non-QA lesioned AAV-BDNF treated rats. (D) Co-labelling for HA and krox-24 was evident for transduced cells in the reduced-titre AAV-BDNF study although not all HA-positive cells displayed krox-24 expression (arrowheads). Scale bar: 30µm 127 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF A AAV-Luciferase Luciferase NeuN B AAV-Luciferase Luciferase DARPP-32 C AAV-Luciferase Luciferase krox-24 D AAV-Luciferase Luciferase GFAP Figure 5-21 Immunofluorescent analysis of AAV-Luciferase transduced cells 128 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF A High-titre AAV-BDNF HA DARPP-32 B Reduced-titre AAV-BDNF HA DARPP-32 C Reduced-titre AAV-BDNF No-QA HA DARPP-32 D Reduced-titre AAV-BDNF HA krox-24 Figure 5-22 Immunofluorescent analysis of AAV-BDNF transduced cells in the striatum 129 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF A AAV-GDNF HA DARPP-32 B AAV-GDNF HA DARPP-32 C AAV-GDNF HA krox-24 Figure 5-23 Immunofluorescent analysis of AAV-GDNF transduced cells in the striatum Representative single-slice confocal images from AAV-GDNF treated rats showing HA-positive GDNF expressing cells within the striatum (red) together with immunofluorescent staining for (A,B) DARPP-32 or (C) krox-24 expression (both displayed in green). (A) Co-labelling with HA and DARPP-32 was not evident in the QA-lesioned striatum despite overlapping distribution in some areas showing general maintenance of DARPP-32 positive neurons. (B) AAV-GDNF transduced cells were also found to survive within striatal areas largely devoid of DARPP-32 expressing cells. (C) Labelling of krox-24 revealed co-localisation with some HA-positive cells (arrows) although many of the AAV-GDNF transduced cells lacked krox-24 expression. Scale bar: 30µm 130 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.7.4 Dopaminergic innervations of the striatum Using tyrosine hydroxylase (TH) as a marker to visualise the maintenance of dopaminergic input into the striatum I assessed TH-positive immunostained fibres within the treated striatum relative to the contralateral hemisphere. A clear loss of TH-positive fibres was visible in the striatum of control treated rats proximal to the site of QA injection, with significant disruption to the structural arrangement of TH fibres in a larger portion of the lesioned striatum surrounding the central lesion core (Figure 5-24A). QA-induced disruption of TH-fibres was less evident in the reduced-titre AAV-Luciferase A Low-titre AAV-BDNF B i AAV-GDNF C v iii ii vi iv i iii v ii iv vi Figure 5-24 Tyrosine hydroxylase expression in the striatum Representative images displaying TH-immunoreactivity in the striatum following QA injection in (A) AAV-Luciferase, (B) reduced-titre AAV-BDNF, and (C) AAV-GDNF treated rats. The distribution of TH-positive fibres was greatly affected in the control treated striatum (A) with a loss of TH expression at the lesion centre (ii) and a condensing of the TH-positive matrix component of the striatum in the surrounding QA-lesioned striatum (i). AAV-BDNF treated rats displayed less disruption to the distribution of TH expression in the striatum (iii, iv), but AAV-GDNF delivery did not induce any obvious changes compared to controls (v, vi). Dashed lines indicate the extent of striatal disruption (middle images) and the TH-depleted core (bottom images). Scale bar = 1mm for top images and 200µm for higher magnification images. 131 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF AAV-BDNF treated rats with the injected striatum generally displaying greater maintenance of the normally dispersed arrangement of the TH-positive fibres within the striatal matrix, and less evident depletion of TH proximal to the site of injection (Figure 5-24B). TH-expression in AAV-GDNF treated rats from the initial investigation was also visually assessed given the potent trophic action of GDNF on dopaminergic neurons (Choi-Lundberg and Bohn 1995), however the localisation of striatal TH appeared to be disrupted by QA in a similar pattern and extent to that observed in the control treated rats (Figure 5-24C). To quantify the maintenance of dopaminergic striatal innervations imparted by AAV-BDNF delivery, integrated density measurements of TH-positive staining in the treated striatum relative to the contralateral striatum were performed. Control rats displayed an 80 ± 3% maintenance of TH immunostaining density which was not significantly altered by the reduced-titre AAV-BDNF treatment at 82 ± 3% TH maintenance (Figure 5-25A). The loss of striatal TH innervations showed a significant correlation with the loss of DARPP-32 positive striatal neurons (Pearson r = 0.66, P < 0.01; Figure 5-25B). With no alteration in the overall level of TH-staining in the AAV-BDNF treated rats despite extensive maintenance of TH distribution, and no apparent maintenance of DARPP-32 neurons by AAV-GDNF, I did not undertake density quantification of TH-staining in the AAV-GDNF treated striatum. A B Figure 5-25 Maintenance of tyrosine hydroxylase striatal innervations (A) Graph showing the relative density of TH-positive fibre staining in the treated striatum compared with TH-staining intensity within the contralateral striatum. Maintenance of striatal TH was determined by measuring the integrated density of DAB immunoreactivity in the striatum averaged over eight equispaced coronal sections. (B) Plot displaying the correlation between the maintenance of TH expression in the striatum and relative maintenance of DARPP-32 expressing striatal neurons. Linear regression analysis: slope = 0.32 ± 0.09, P < 0.01. R value represents overall Pearson correlation coefficient. 132 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.7.5 Cortical projection fibres in the striatum A AAV-Luciferase Contralateral AAV-BDNF H Ipsilateral Ipsilateral B B E E II C C FF JJ D D G G K K Figure 5-26 SMI32 expression in the cortex and striatum Representative images of SMI32 expression in the contralateral (left hemisphere) and ipsilateral (right hemisphere) cortex and striatum following QA injection in (A,E-G) AAV-Luciferase and (HK) reduced-titre AAV-BDNF treated rats. (B,E,I) Pyramidal neurons predominantly in cortical layers III and V overlaying the striatum appeared unaffected by QA striatal lesioning. SMI32 expression in (C,D) the intact contralateral striatum and (F,G,J,K) the AAV vector and QA treated striatum. Regions outlined in boxes are shown at higher magnification in the figures immediately below. Scale bar = 1mm for whole sections (A,H), 100µm for (B,E,I), 200µm for (C,F,J), 50µm for (D,G,K). 133 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Maintenance of cortical neuronal fibres within the striatum was visualised by immunostaining for SMI32 – a non-phosphorylated neurofilament protein – expressed by a subpopulation of pyramidal neurons (Campbell and Morrison 1989; Gerfen 1992), which are vulnerable to HD degeneration (Cudkowicz and Kowall 1990; Hedreen et al. 1991; Macdonald and Halliday 2002). The majority of SMI32 positive neurons are located in layers III and V with no apparent loss of neurons observed in the primary motor or sensory areas overlying the striatum following unilateral QA lesioning (Figure 5-26). Distribution of SMI32 expression in the striatum was however altered following QA lesioning. Within the intact striatum SMI32 appeared largely contained within fibres innervating the striosome-like patches which are irregularly dispersed throughout the striatum (Figure 5-26C,D). Following QA-lesioning of the control treated striatum, the striatal density of SMI32 staining increased with a greater proportion of the striatal area containing SMI32 consistent with a loss of striatal matrix and possible enlargement of the striosome compartments or fibre bundles (Figure 5-26F,G). This QA-induced alteration in striatal organisation is similar to the changes observed with TH staining (Section 5.7.4). The expression of SMI32 was also enhanced in AAV-BDNF treated rats in the striatum proximal to the site of QA injection with the fibres appearing larger than in the control rats, but was less extensively increased in the more distal surrounding striatum (Figure 5-26J,K). AAV-GDNF treated rats from the initial investigation were not assessed for SMI32 due to the lack of any apparent striatal neuroprotection. 5.7.6 Striatal projection nuclei To investigate whether enhanced BDNF expression prior to QA lesioning of the striatum had any bearing on neuronal maintenance in the striatal projection nuclei I performed immunocytochemistry to identify neurons within the substantia nigra and globus pallidus. Analysis was only performed on the control and reduced-titre AAV-BDNF treated rats from the follow-up investigation with no evidence of any striatal neuroprotection being imparted by AAV-GDNF delivery. 5.7.6.1 Substantia Nigra Dopaminergic neurons in the substantia nigra pars compacta region were visualised by TH immunocytochemical staining and showed no observable differences between brain hemispheres in either control (Figure 5-27) or AAV-BDNF treated rats (Figure 5-28). Similarly the parvalbuminpositive GABAergic neurons in the substantia nigra pars reticulata appeared to be equally unaffected by the striatal QA lesion with visually comparable expression in each hemisphere. With the nigral neurons not appearing to be influenced by the QA lesion I analysed the maintenance of striatonigral axonal innervations. The projection of DARPP-32 positive fibres, originating from the vulnerable 134 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF DARPP-32 expressing striatal projection neurons, into the substantia nigra appeared to be partially depleted in the ipsilateral nigra (Figure 5-27C and Figure 5-28C). To quantify the loss of striatonigral innervations I measured the density of DARPP-32 immunoreactivity in the ipsilateral substantia nigra relative to the contralateral side of the brain (Figure 5-29A). Control rats displayed a 29 ± 3% loss of DARPP-32 immunostaining that was significantly reduced to 10 ± 5 % in AAVBDNF rats. Comparison with the relative maintenance of DARPP-32 positive neuronal cell bodies in the striatum showed a direct correlation (Pearson r = 0.75, P < 0.001) between striatal cell bodies and DARPP-32 immunoreactivity in the substantia nigra (Figure 5-29B). A Contralateral Ipsilateral Tyrosine Hydroxylase B Parvalbumin C DARPP-32 Figure 5-27 Visual assessment of the substantia nigra in control AAV-Luciferase treated rats Representative images of the substantia nigra in an AAV-Luciferase treated brain displaying the comparison between the treated (ipsilateral) and un-treated (contralateral) hemispheres. Immunocytochemical staining of (A) TH-positive neurons in the pars compacta, (B) parvalbuminpositive neurons in the pars reticulata, and (C) DARPP-32 positive neurons and efferent fibres. Regions outlined by boxes are shown at higher magnification in figures to the right. Scale bar = 2mm for whole sections and 200µ m for higher magnification images. 135 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Contralateral Ipsilateral A Tyrosine Hydroxylase B Parvalbumin C DARPP-32 Figure 5-28 Visual assessment of the substantia nigra in reduced-titre AAV-BDNF treated rats Representative images of the substantia nigra in a reduced-titre AAV-BDNF treated brain displaying the comparison between the treated (ipsilateral) and un-treated (contralateral) hemispheres. Immunocytochemical staining of (A) TH-positive neurons in the pars compacta, (B) parvalbuminpositive neurons in the pars reticulata, and (C) DARPP-32 positive neurons and efferent fibres. Regions outlined by boxes are shown at higher magnification in figures to the right. Scale bar = 2mm for whole sections and 200µ m for higher magnification images. 136 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF A B Figure 5-29 DARPP-32 innervations of the substantia nigra (A) Graph showing the density of DARPP-32 immunoreactivity within the substantia nigra, ipsilateral to the treated striatum, as a proportion of DARPP-32 immunostaining in the contralateral substantia nigra. DARPP-32 density was measured on three coronal sections through the substantia nigra. (B) Plot displaying the correlation between DARPP-32 positive fibre innervations of the substantia nigra and maintenance of the DARPP-32 positive striatal neurons. * Un-paired t-test P < 0.01. Deming linear correlation: slope = 0.70 ± 0.15, P < 0.001. R value represents overall Pearson correlation coefficient. 5.7.6.2 Globus Pallidus Pallidal neurons were visualised by immunostaining for parvalbumin and NeuN with a clear loss of neurons seen within the ipsilateral globus pallidus of AAV-Luciferase treated control rats (Figure 5-30A,B). AAV-BDNF treated rats did not generally have any obvious loss of pallidal neurons (Figure 5-31A,B). Additional staining for DARPP-32 showed the mostly intact caudal region of the striatum adjacent to the globus pallidus remained mostly intact, but displayed reduced DARPP-32 immunoreactivity in the globus pallidus compared with the contralateral hemisphere for both control (Figure 5-30C) and AAV-BDNF treated rats (Figure 5-31C). With parvalbumin positive neurons in the globus pallidus clearly affected by the QA lesion in some rats, I performed stereological estimates over four equispaced coronal sections through the pallidus (Stereology coefficient of error ≤ 0.09). In contrast to all other analysis parvalbumin-positive neurons in the globus pallidus showed a significant difference between the AAV-Luciferase and PBS control groups (t-test P < 0.01; Figure 5-32A). AAV-Luciferase animals were massively depleted of parvalbumin neurons in the globus pallidus with only 15 ± 8 % maintained relative to the contralateral hemisphere while the PBS group had 87 ± 18 % maintenance. The parvalbuminpositive pallidal neurons were mostly maintained in the AAV-BDNF treated group (96 ± 10 %). There was no significant correlation between the survival of DARPP-32 positive striatal neurons and the parvalbumin expressing pallidal neurons (Pearson r = 0.42, P = 0.07; Figure 5-32B). 137 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Contralateral Ipsilateral Parvalbumin A NeuN B DARPP-32 C Figure 5-30 Visual assessment of the globus pallidus following AAV-Luciferase treatment Representative images of the globus pallidus in a control AAV-Luciferase treated brain displaying the comparison between the treated (ipsilateral) and un-treated (contralateral) hemispheres. Immunocytochemical staining of (A) parvalbumin-positive pallidal neurons, (B) NeuN-positive neurons, and (C) DARPP-32 immunoreactivity. Dashed lines indicate the boundary between the striatum and globus pallidus. Scale bar = 250µm. 138 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Contralateral Ipsilateral Parvalbumin A NeuN B DARPP-32 C Figure 5-31 Visual assessment of the globus pallidus following reduced-titre AAV-BDNF treatment Representative images of the globus pallidus in a reduced-titre AAV-BDNF treated brain displaying the comparison between the treated (ipsilateral) and un-treated (contralateral) hemispheres. Immunocytochemical staining of (A) parvalbumin-positive pallidal neurons, (B) NeuN-positive neurons, and (C) DARPP-32 immunoreactivity. Dashed lines indicate the boundary between the striatum and globus pallidus. Scale bar = 250µm. 139 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF A B Figure 5-32 Survival of parvalbumin-positive neurons in the globus pallidus (A) Graph displaying the relative proportions of parvalbumin-immunopositive neurons surviving in the globus pallidus as estimated by optical fractionator stereology on four coronal sections through the globus pallidus. (B) Plot comparing the survival of parvalbumin-positive neurons relative to the maintenance of DARPP-32 positive striatal neurons. * Un-paired t-test P < 0.01. R value represents overall Pearson correlation coefficient, P = 0.07. 5.7.7 Pathological correlations with functional behaviour impairments To assess whether there were significant correlations between the functional behavioural impairments and the loss of striatal neurons or pallidal neurons I analysed overall Pearson correlations and individual treatment group linear relationships for each of the behavioural tests undertaken in the follow-up, reduced-titre AAV-BDNF investigation. Due to the significant maintenance of parvalbumin-expressing pallidal neurons in PBS controls compared with the loss in AAV-Luciferase treated rats I have separately identified whether rats received PBS or AAVLuciferase in the correlations against pallidal neurons but not for striatal DARPP-32 correlations where there was no significant difference between the groups. The PBS and AAV-Luciferase treated rats were still combined as a single control group to assess for linear relationships as they did not perform differently as two distinct groups in any of the functional behavioural assessments. 5.7.7.1 Spontaneous Ipsilateral Forelimb Use Bias The proportion of exploratory behaviour undertaken by the rats with their ipsilateral forelimb following QA injection was found to be significantly correlated with the loss of DARPP-32 positive neurons in the striatum (Pearson r = -0.49, P < 0.05; Figure 5-33A). However, with the prior delivery of AAV-BDNF attenuating development of an ipsilateral forelimb use bias, independent 140 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF analysis for a linear trend between DARPP-32 maintenance and ipsilateral asymmetry score indicated that the extent of ipsilateral bias in AAV-BDNF treated rats was not greatly influenced by the loss of DARPP-32 positive striatal neurons. This was in contrast to the strong linear relationship between the loss of DARPP-32 striatal neurons and ipsilateral forelimb use in the control group (P < 0.001). Exploratory ipsilateral forelimb use in the AAV-BDNF treated rats did however appear to be strongly linked to the ipsilateral:contralateral ratio of parvalbumin-positive neurons in the globus pallidus (Linear regression P < 0.01) with a 1:1 ratio correlating with a non-biased 52 ± 2 % ipsilateral asymmetry score (Figure 5-33B). Overall though the maintenance of parvalbumin- positive pallidal neurons did not significantly correlate with the ipsilateral asymmetry score (Pearson r = -0.39, P = 0.10) with the control rats displaying preferential ipsilateral forelimb use independent from the relative number of parvalbumin positive neurons in the globus pallidus (P = 0.518). A B Figure 5-33 Preferential forelimb use correlation with striatum and globus pallidus cell loss Plots correlating the extent of ipsilateral forelimb use bias with the relative proportion of (A) DARPP-32 and (B) parvalbumin-positive neurons remaining within the striatum and globus pallidus respectively. Data points represent individual rats’ average score over the seven post-QA trials. R values represent overall Pearson correlation coefficients independent of treatment. Solid lines represent significant non-zero linear correlations (P < 0.05) and the dotted lines non-significant linear trends for individual treatment groups. Comparison with controls: (A) slope P = 0.14; elevation P < 0.001; (B) slope P = 0.001. 5.7.7.2 Contralateral Sensorimotor Neglect Preferential retrieval of sugar pellets from the side of the corridor adjacent to the rats treated brain hemisphere was significantly correlated with the anatomical state of the globus pallidus (Pearson r = -0.68, P < 0.01) but not the extent of DARPP-32 striatal cell loss (Pearson r = -0.41, P = 0.08; Figure 5-34A). Maintenance of parvalbumin-positive pallidal neurons appeared to contribute towards 141 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF attenuating development of a contralateral sensorimotor neglect following QA injection. Individual AAV-BDNF and control group assessment for linear relationships indicated that this sensorimotor neglect correlation with pallidal neurons was stronger for AAV-BDNF treated rats (Linear trend slope = -63 ± 14, P < 0.0001) than the control rats (slope = -19 ± 5, P < 0.001; Figure 5-34B). While there was not overall a significant correlation with the DARPP-32 striatal neurons, the percentage of ipsilateral retrievals made by control rats did display a significant linear relationship with the loss of DARPP-32-positive striatal neurons (slope = -77 ± 19, P < 0.01; Figure 5-34A). AAV-BDNF treated rats did not show any relationship between attenuation of preferential ipsilateral retrievals and the gross maintenance of DARPP-32-positive striatal neurons. A B Figure 5-34 Sensorimotor neglect correlation with striatal and pallidal neuronal cell loss Plots correlating the percentage of ipsilateral sugar pellet retrievals made in the “corridor” task with the relative proportion of (A) DARPP-32 and (B) parvalbumin-positive neurons maintained within the striatum and globus pallidus respectively. Data points represent individual rats’ average retrieval preference over the four post-QA trials. R values represent overall Pearson correlation coefficients independent of treatment. Solid lines represent significant non-zero linear correlations (P < 0.05) and the dotted lines non-significant linear trends for individual treatment groups. Comparison with controls: (A) slope P < 0.05; (B) slope P < 0.01. 5.7.7.3 Apomorphine-Induced Rotations Apomorphine-induced rotational behaviour did not display any overall correlation with DARPP-32positive striatal cell loss (Pearson r = -0.32, P = 0.19; Figure 5-35A). Separate linear trend analysis with the individual groups also failed to show any significant correlations for either the AAV-BDNF rats (P = 0.069) or control rats (P = 0.86). In contrast, comparison of apomorphine-induced rotations with parvalbumin-positive neurons in the globus pallidus did reveal a significant correlation with the maintenance of pallidal neurons (Pearson r = -0.66, P < 0.01; Figure 5-35B). This correlation was 142 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF very significant for the control treated rats (Linear trend P < 0.001), but not for the AAV-BDNF treated rats (P = 0.07) although both treatment groups showed similar trends. The direction of apomorphine-induced rotation also appeared to be strongly dependent on the integrity of the globus pallidus with a loss of pallidal neurons resulting in rotational behaviour towards the lesioned side, while apomorphine induced the rats to rotate contralaterally away from the lesioned brain hemisphere when the globus pallidus was fully maintained (Figure 5-35B). A B Figure 5-35 Correlation of apomorphine-induced rotational behaviour with striatal and pallidal neuronal cell loss Plots displaying the percentage of apomorphine-induced rotations in an ipsilateral direction in relation to the maintenance of (A) DARPP-32 and (B) parvalbumin-positive neurons within the striatum and globus pallidus respectively. Each data point represents the result of a single post-QA trial with three trials conducted per rat. Shaded boxes show the predominant contralateral rotational behaviour when the relative number of parvalbumin pallidal neurons is greater in the treated hemisphere and preferential ipsilateral rotations when the globus pallidus is compromised. R values represent overall Pearson correlation coefficients independent of treatment. Solid lines represent significant non-zero linear correlations (P < 0.05) and the dotted lines non-significant linear trends for individual treatment groups. AAV-BDNF treatment did not significantly alter the linear correlations observed in the control rats. 143 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.8 Discussion Through these preventative in vivo studies I investigated the pathological and functional impact of increasing the availability of neurotrophic factors using AAV vectors to establish continuous expression of either BDNF or GDNF within the striatum prior to challenging the striatal neurons with QA-administered excitotoxicity. While the protective effects of neurotrophic factors have been previously studied in models of Huntington’s disease with varying degrees of success (Section 1.6), the ongoing development and advances in gene transfer technologies make additional investigation necessary to enhance our understanding of both delivery vectors and potential gene-based therapeutics in an in vivo research setting. Stereotype-2 AAV vectors used in many early AAV gene transfer studies have significantly lower efficiency of neuronal transduction than the chimeric 1/2 serotype vectors used in these investigations. Using behaviour tests to measure functional motor deficits consequential of an interhemispherical imbalance in the integrity of motor and sensorimotor neuronal processing in unilaterally treated rats, I endeavoured to correlate the prevention of physical symptomatic impairments with the maintenance of basal ganglia anatomical organisation provided by the prior intrastriatal delivery of the AAV-BDNF or AAV-GDNF vectors. 5.8.1 AAV-mediated BDNF or GDNF expression prior to QA The AAV1/2 vectors I constructed – containing cDNA sequences for BDNF or GDNF under the control of a constitutive CBA promoter and WPRE sequence – provided an efficient mechanism for establishing high-level neurotrophic factor expression in the CNS. Spread of the AAV vector transduction throughout the rodent striatum following a single injection appeared reasonably extensive in the majority of cases although somewhat irregular in its penetration resulting in patches of intensive expression and other areas with few or no transduced cells. With the neurotrophic factors being secreted from transduced cells, the uneven spread of transduction was not immediately concerning as the non-transduced striatal neurons were still likely to be exposed to an increased level of BDNF or GDNF as the neurotrophic factors were released, diffusing away from the transduced cells. 5.8.1.1 Undiluted AAV-BDNF Following the development of seizures, hyperactivity and continuous weight loss in some of the AAV-BDNF treated rats, evident five weeks after AAV-BDNF vector delivery and forcing early termination of the BDNF portion of the study, I had to reassess the delivery procedure for this vector. 144 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF No welfare concerns were raised with any of the other AAV vectors, and with the delayed onset the deleterious effects were most likely due specifically to the BDNF expression. BDNF has previously been implicated in seizure severity, thought to be modulated via BDNF activity in the entorhinal cortex and hippocampus (Scharfman 1997; Croll et al. 1999). Additionally BDNF can act as an appetite suppressor causing weight loss following ventricular infusion (Pelleymounter et al. 1995), and positively regulates locomotor activity (Kernie et al. 2000) which is consistent with the effects I observed. With the massive escalation in striatal BDNF levels following undiluted AAV-BDNF delivery, and with observed anterograde transport of the AAV-BDNF vector to striatal projection nuclei, it is probable that the availability of BDNF was also enhanced in other regions of the rodent brain including the entorhinal cortex which projects efferent fibres to the striatum. Retrograde transport of AAV-vectors to the entorhinal cortex has also been previously shown to occur (Kaspar et al. 2002). Analysis of the transgenic BDNF expression in the striatum showed an inverse correlation with DARPP-32 expression suggesting the intensive BDNF expression was detrimentally impairing the functionality of transduced striatal neurons, but in direct contrast was supplying neuroprotective support to the neighbouring DARPP-32 positive neurons. While I did not conclusively determine whether this loss of DARPP-32 expression was due to a BDNF-induced down regulation of expression or an enhanced susceptibility of transduced neurons to QA-induced excitotoxicity, the persistence of HA-expressing cell bodies in the striatum and their co-labelling with krox-24 indicates survival – and presumed functional activity – of a population of BDNF expressing striatal neurons four weeks post-QA. Given the neuronal preference of the AAV1/2 vectors, and the normally high expression of DARPP-32 by striatal projection neurons (Ouimet et al. 1984; Ouimet et al. 1998), it is reasonable to assume that the vast majority of AAV-BDNF transduced striatal cells would have been expressing DARPP-32 at the time of transduction, prior to being challenged with QA. Striatal neurons transduced by AAV-GDNF or AAV-Luciferase also lacked DARPP-32 expression at the conclusion of the study. While some of the transduced neurons may well have been non-DARPP-32 expressing interneurons that were spared by QA-lesioning, the majority are likely to have been striatal projection neurons indicating a disruption of DARPP-32 expression, not specific to transgene expression, following AAV vector transduction. Given this apparent down-regulation of DARPP-32 expression in transduced striatal neurons, the use of DARPP-32 as a marker of striatal projection neurons to quantify BDNF and GDNF-derived support against QA-induced cell death will have lead to an underestimation of the actual number of surviving neurons. This may have contributed to the lack of a significant increase in the stereologically estimated striatal neurons maintained in AAVBDNF treated rats relative to the control rats despite visually distinguishable areas of protected 145 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF neurons and the complete preservation of striatal volume (Section 5.7.1). However, functional implications of DARPP-32 down-regulation will need serious consideration given the central role of DARPP-32 in mediating intracellular signalling of both dopamine and glutamate in the dopaminoceptive striatal projection neurons (Greengard et al. 1999). A reduction in DARPP-32 has been reported in presymptomatic HD mice and may be a contributing factor to HD pathogenesis (Bibb et al. 2000). Despite the non-significant maintenance of DARPP-32 positive neurons in the striatum, extensive BDNF expression in the initial investigation ameliorated the development of QA-induced ipsilateral forelimb use bias with the rats actually tending to favour their contralateral forelimb. The preservation / enhancement of forelimb motor control suggests a possible augmentation of brain function due to increased BDNF expression that was independent of actual neuronal cell maintenance in the striatum. Together these observations suggested that despite serious detrimental effects on the rats’ welfare and the potentially detrimental alterations to the function of transduced neurons, BDNF could still offer both neuroprotective support to striatal neurons against excitotoxic insult and prevent associated behavioural deficits. Provided BDNF delivery can be sufficiently regulated to limit the spread and control the level of expression it could well be of therapeutic benefit, and therefore I felt that AAV-mediated BDNF expression warranted further investigation. 5.8.1.2 AAV-GDNF The delivery of AAV-GDNF and subsequent rise in striatal GDNF availability from normally very low expression levels in the adult striatum (Schaar et al. 1993; Springer et al. 1994; Choi-Lundberg and Bohn 1995) imparted significant attenuation of the ipsilateral forelimb use bias that developed in control rats following QA lesioning. The contrasting development of a contralateral forelimb use bias as seen in the AAV-BDNF treated rats was not however recorded in analysis of AAV-GDNF treated rats, suggesting that the maintenance of forelimb motor control provided by AAV-GDNF was probably mediated via a different signalling pathway than the AAV-BDNF effects. Apomorphineand amphetamine-induced rotational behaviour did not appear greatly altered from the performance of control rats, with the AAV-GDNF treated rats preferentially displaying unidirectional rotations, usually towards the QA injected striatal hemisphere. Despite the maintenance of forelimb motor control, AAV-GDNF delivery prior to QA did not enhance the resistance of DARPP-32 positive striatal projection neurons, supporting the observation of the drug-induced rotational testing. This lack of neuroprotection was in contrast to my earlier findings of partial protection of striatal neurons following transduction with a lower expression level AAV2-GDNF vector (Kells et al. 2004) and other reports of AAV-mediated GDNF delivery in the 3146 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF NP mitochondrial toxin (McBride et al. 2003) and N171-82Q genetic models of HD (McBride et al. 2006), but in agreement with Popovic and colleagues (2005) who reported no attenuation of behavioural or neuropathological changes in R6/2 Huntington mice following LV vector-mediated GDNF expression. Given these previous results the lack of AAV-GDNF derived neuroprotection against QA-induced excitotoxicity was unexpected. I had hypothesised that the enhanced neuronal transduction efficiency of the new chimeric AAV1/2 vector, and higher CBA promoter driven GDNF expression, would increase GDNF-derived support of the striatal neurons. A common factor in the studies which have demonstrated GDNF expression-derived protection is the assessment of NeuN immunoreactivity (McBride et al. 2003; Kells et al. 2004; McBride et al. 2006), in contrast to the DARPP-32 expression used in both the LV-GDNF study (Popovic et al. 2005) and current analysis to quantify neuroprotection, raising the possibility that the neurons may survive but with altered phenotype / functionality. Alternatively the conflicting findings with GDNF may reflect the different processes of striatal cell loss between neurotoxins – excitotoxic QA versus mitochondrial inhibitor 3NP – or genetic models – R6/2 (Hdh exon-1 with 150 CAG repeats) versus N171-82Q (Hdh Nterminal with 82 CAG repeats) – of HD. The 3-NP and N171-82Q models display a more protracted development of behavioural impairments and pathological changes suggesting a less intensive insult that may be better counteracted by GDNF. A further explanation of these differential GDNF findings could be the extent of transduction and / or the production of GDNF with the more efficient AAV1/2 and LV vectors potentially generating GDNF that exceeds therapeutically beneficial levels. Direct comparisons of the quantity of GDNF expression between studies was unfortunately not possible due to differences in the quantification protocols and in the reporting of transgene expression, however in my earlier investigations with AAV2-GDNF (Kells et al. 2004) the transgenic expression of GDNF was considerably lower [Unpublished data]. A recent study by Eslamboli et al. (2005) reported extensive protection of dopaminergic nigral neurons and attenuation of behavioural deficits following low-level GDNF expression in the primate striatum (three- to four-fold increase) prior to 6-hydroxydopamine modelling of Parkinson’s disease. High levels of striatal GDNF were shown to bilaterally increase TH and dopamine turnover causing a reduction in striatal dopamine (Eslamboli et al. 2005), and have also been reported to increase the loss of striatal neurons following ischemic stress (Arvidsson et al. 2003). In reassessing the expression pattern of GDNF within the striatum it became apparent that large numbers of GDNF expressing cells were still present within the striatum following QA injection suggesting they had in fact received protection against QA-induced cell death. On correlating the location of these surviving AAV-GDNF transduced striatal cells to the area of QA-lesioning, defined by DARPP-32 immunoreactivity, it was evident that the majority of HA-positive cells resided in 147 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF areas largely devoid of DARPP-32 expression. Additional correlation with the general neuronal marker NeuN displayed a similar lack of immunopositive cells in the region of HA staining, although NeuN cells appeared more prevalent than DARPP-32 positive cells (Section 5.7.1.1). Furthermore, double-immunofluorescent labelling of the AAV-GDNF treated striatum revealed only minimal colabelling of HA and DARPP-32 positive cells displaying a down-regulation of DARPP-32 in the HA-positive GDNF expressing cells (Section 5.7.3). The loss of DARPP-32 is highly likely to affect neurotransmitter signalling (Section 5.8.1.1 and 7.1). It therefore appeared that while many AAVGDNF transduced neurons did actually survive QA-induced excitotoxicity, the expression of their usually dominant signalling proteins used as identification markers were severely compromised. How this alteration in protein expression influences the functionality of the striatal neurons was not specifically investigated but highlights the need for caution and assessment of host cells physiological functions following in vivo gene transfer to direct production of a bioactive therapeutic molecule. Stereological analysis of NeuN-immunopositive striatal neurons may possibly have better estimated the proportion of striatal neurons maintained, although from the lack of correlation in immunostaining it appeared that NeuN may also have been down-regulated in the AAV-GDNF transduced cells by the end-point of the investigation – 11-weeks post AAV-GDNF delivery. Although not quantified, TH staining in the striatum – indicative of dopamine innervations – following QA-lesioning did not appear to be altered by the enhanced GDNF expression. However the dopamine neurons in general are not affected by QA lesioning and therefore any TH-positive fibre sprouting induced by GDNF – as reported following striatal GDNF expression in animal models of Parkinson’s disease (Rosenblad et al. 2000; Kirik et al. 2000a; Georgievska et al. 2002) – may simply not be visibly discernable. Therefore, although indirect evidence suggested that AAV-GDNF delivery imparted neuroprotective support to transduced striatal neurons, continuous high-level GDNF expression appeared to cause alterations in neuronal processes. Despite the fact that I was not aware of any adverse alterations in the rats behaviour during the extended period of functional behavioural testing – actually observing maintenance of forelimb motor control – the changes in protein expression patterns are likely to ultimately result in disruption to neuronal function, potentially negating any benefits of initial AAVGDNF derived neuroprotective support. While better regulation of the GDNF expression may well circumvent some of these undesired effects, I opted against proceeding further with investigation of this AAV-GDNF vector in the QA lesion model of HD. However it is possible that a significantly lower level of GDNF expression maybe beneficial (Eslamboli et al. 2005), and that the extensive expression I induced exceeded tolerable quantities for a region that has a very low level of endogenous GDNF expression in adulthood (Springer et al. 1994; Choi-Lundberg and Bohn 1995). 148 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.8.2 Reduced-titre AAV-BDNF behavioural and pathological protection Continuing investigation with delivery of the AAV-BDNF vector, I elected to shift the striatal injections site away from the same stereotaxic coordinates as the QA injection and split the vector delivery over two stereotaxic sites flanking the QA injection site in an attempt to even out the striatal distribution of transduced cells. To minimise any toxicity associated with AAV-BDNF transduction I also diluted the vector ten-fold to reduce the multiplicity of transduction and thereby decrease the intensity of BDNF expression per transduced cell. This reduced the enhanced expression level of BDNF in the striatum from ~1000-times endogenous expression to a ~150-fold increase at the time of QA administration, and prevented the previously observed weight-loss and seizure development. Visual assessment of AAV-BDNF transduced cells showed extensive transduction of neurons around the site of injection, but significantly less spread than the undiluted vector injections. To more fully assess the AAV-BDNF delivery on functional behaviour – without the confounding impact of a QA lesion – I additionally assessed a smaller cohort of reduced-titre AAV-BDNF treated rats for 11weeks post-AAV vector delivery in parallel with the main neuroprotective investigation. 5.8.2.1 Behavioural Protection Prior to QA-lesioning rats that received reduced-titre AAV-BDNF showed an enhanced tendency to use their contralateral forelimb for initiating exploratory movement and the induction of ipsilateral rotational behaviour following apomorphine administration, compared with unbiased lateralised behaviour of the control rats. While this behaviour was similar to that observed in the initial hightitre AAV-BDNF study, the slight preferential use of the contralateral forelimb did not develop into a strong contralateral bias in either the QA-lesioned or non-lesioned rats with both groups remaining insignificantly altered from baseline analysis in contrast to the ipsilateral bias in control rats and contralateral bias in the initial BDNF study (Section 5.6.1). While not significantly different, the two AAV-BDNF treated groups were separated after the main group received a QA injection inducing a small shift towards greater use of their ipsilateral forelimb, while the non-QA injected rats maintained slightly greater contralateral forelimb use, although neither AAV-BDNF treated group could be considered to have a significant forelimb use bias. These results suggested that while the expression of BDNF may provide some enhancement of basal ganglia coordinated motor control, the lack of a significant contralateral usage in the absence of striatal cell death implies AAV-BDNF derived amelioration of QA induced ipsilateral forelimb bias is unlikely to be solely attributed to 149 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF BDNF activity compensating for the loss of striatal cell loss, but involve the maintenance of functional striatal projection neuron circuitry by BDNF. The transient display of apomorphine-induced ipsilateral rotational behaviour initially following AAV-BDNF delivery, before predominantly switching to induce contralateral rotations in both QA lesioned and non-QA lesioned rats (Section 5.6.2.1), suggests that BDNF expression causes opposing short and long-term actions. QA striatal lesioning of control rats caused a loss of dopaminoceptive projection neurons, generally resulting in ipsilateral rotations following either apomorphine or amphetamine administration. Therefore enhanced BDNF expression appeared to be acting long-term to enhance dopamine activity, but initially resulted in impairment to the striatal neurons ability to process dopamine signalling. With apomorphine directly stimulating both D1 and D2 dopamine receptors (Arnt and Hyttel 1985), rotational behaviour was most probably mediated through alterations in the expression of dopamine receptors by surviving striatal neurons, thereby making the results of drug-induced rotational testing difficult to conclusively assess in regards to any direct neuroprotective support provided by AAV-BDNF. However this does further highlight challenges facing the use of neurotrophic factors as therapeutic agents with widespread neurotrophic factor receptor expression and multiple signalling pathways creating a high potential for inducing nontargeted effects that clearly need consideration prior to any clinical investigation. BDNF has previously been shown to enhance dopamine activity by increasing both the striatal release and turnover of dopamine (Altar et al. 1992b; Altar et al. 1994), and induces behavioural changes indicative of increased dopamine activity (Martin-Iverson et al. 1994; Martin-Iverson and Altar 1996). More recently though, BDNF has been shown to positively-regulate the expression of post-synaptic D3 dopamine receptors in the striatum (Guillin et al. 2001), suggesting that the unilateral delivery of BDNF may have resulted in sensitisation of the surviving striatal neurons to dopamine agonists, thereby counteracting any QA-induced deficiency in motor effector output. Therefore the initial dysfunction following AAV-BDNF delivery may have been consequential of a rapid increase in striatal BDNF levels causing functional disruption or desentisation of the dopamine receptors. Dopamine has been proposed to actually contribute to the degeneration of striatal neurons such that a sudden increase in BDNF-induced dopamine release, without a corresponding increase in dopamine uptake, may initially cause neuronal dysfunction (Filloux and Townsend 1993; Hastings et al. 1996; Hattori et al. 1998). Dopamine toxicity may also have been a contributing factor to the deleterious effects in the initial high-expression AAV-BDNF investigation. Another contributing factor may be the involvement of TrkB receptors which are likely to have been down-regulated at some stage following continuous enhancement of BDNF expression (Frank et al. 1996). 150 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Additional evidence of AAV-BDNF delivery supplying neuroprotective support resulting in attenuation of functional impairments was observed in the “corridor” task assessment of sensorimotor neglect. The reduced-titre AAV-BDNF delivery completely prevented acquirement of the contralateral sensorimotor neglect observed following QA injection in control rats (Section 5.6.3). Although the non-lesioned and QA lesioned groups of AAV-BDNF treated rats did not significantly differ in their “corridor” task performance, the AAV-BDNF delivery alone did significantly change the non-lesioned rats’ preferential food retrieval from a slight baseline contralateral preference towards development of a slight contralateral neglect. This tended to suggest that AAV-BDNF delivery attenuated the acquirement of contralateral sensorimotor neglect through protection against QA-induced alterations and not via BDNF-induced enhancement of other compensatory mechanisms as suggested by the apomorphine-induced rotations. 5.8.2.2 Neuropathological Protection Despite significant attenuation of behavioural deficits, analysis of neuronal survival following AAVBDNF delivery revealed only limited enhancement of striatal maintenance, compared with the control treated rats. Maintenance of krox-24 expressing striatal neurons was significantly enhanced with a 36% increase in neuron survival following AAV-BDNF delivery; however the parallel analysis of DARPP-32 positive neurons did not quite reach significance despite a similar 38% average increase in the relative number of DARPP-32-positive cells. The lack of significance due to greater variation in the quantification of DARPP-32 neurons is likely to have arisen from the downregulation of DARPP-32 or the less distinct immunostaining of cytoplasmic DARPP-32, compared with the krox-24 nuclear staining, making the striatal neurons more difficult to visually discern against the positive staining of dendritic processes. Co-labelling of the BDNF expressing transduced neurons with DARPP-32 and krox-24 still displayed inverse staining intensities, although krox-24 was detectable in most HA-positive cells and the majority also appeared to display some DARPP-32 expression. While a small enhancement of cellular survival could potentially lead to an observable prevention of behavioural deficits in this QA-lesion model of HD, I felt that the quantified partial protection of striatal neurons supplied by AAV-BDNF was unlikely to have been solely responsible for the amelioration of functional impairments. Coupled with the maintenance of striatal projection neurons I observed that atrophy of the striatum was significantly reduced in the AAV-BDNF treated rats (Section 5.7.1.4), suggesting that the structural organisation of the striatum was protected against QA-induced excitotoxicity by BDNF, possibly to a greater extent than individual striatal neurons. Therefore I propose that the functionality of the striatum in regard to the assessed motor coordination 151 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF tasks is more reliant on structural organisation than the total number of striatal projection neurons, and that AAV-BDNF by enhancing the maintenance of this structural architecture, in addition to providing individual neurons with greater resistance against QA, was able to ameliorate the behavioural deficits. The striatum is topographically organised with the sensory and motor areas of the neocortex projecting to the striatum in a highly organised but overlapping longitudinal arrangement (McGeorge and Faull 1989). Therefore overall preservation of this structural arrangement, with a reduction in neuronal density but without areas of total projection neuron loss, may well attenuate the severity of behavioural deficits. Further analysis revealed that preservation of substantia nigra innervations by striatonigral DARPP32 positive fibres was significantly enhanced by AAV-BDNF, with the nigral neurons themselves not appearing to be greatly affected by the striatal lesion (Section 5.7.6.1). Maintenance of striatonigral projections was correlated with the maintenance of DARPP-32 positive neurons in the striatum. Despite no apparent loss of dopaminergic neurons in the substantia nigra pars compacta I assessed dopaminergic innervations of the striatum by measuring the density of TH-immunostaining to try and determine if maintenance of dopamine input may have been responsible for the reduced severity of behavioural impairments. While the integrated density measurements indicated equal loss of TH-positive fibres in the striatum averaging ~19% reduction relative to the contralateral striatum across all rats following QA lesioning, the control rats visually displayed intensification of the THpositive fibres from the dispersed matrix into defined tracts interwoven around TH-negative patches (Section 5.7.4). Prior AAV-BDNF delivery attenuated this apparent atrophy of the striatal matrix, with TH-staining remaining diffusely distributed throughout the striatum, indicating a general maintenance of the striatal architecture by BDNF. Further evidence for preservation of the striosome-matrix arrangement following AAV-BDNF delivery was provided by visual assessment of the neurofilament protein SMI32 expression in the striatum – presumed to be contained within afferent projections of the SMI32-positive pyramidal neurons in the motor and sensory cortex (Section 5.7.5). Localised to striosome-like patches, SMI32 appeared to be greatly enhanced following QA lesioning possibly due to fibre sprouting or structural rearrangement occurring within these afferent corticostriatal fibres following the death of their target striatal neurons. While SMI32 expression was greatly enhanced across a substantial proportion of the striatum in control rats, with an accompanying increase in the density of striosomes, AAV-BDNF treated rats generally only displayed an increased intensity of SMI32 directly proximal to the QA injection site. A smaller enhancement of SMI32 following BDNF expression is possibly indicative of less fibre sprouting due to a greater maintenance or re-establishment of synaptic connections in the striatum following QA lesioning. 152 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF Pallidal neurons, in contrast to those in the substantia nigra, were subjected to QA induced neurotoxicity although this was highly variable with the two control groups displaying significantly contrasting levels of parvalbumin-positive neuronal maintenance (Section 5.7.6.2). A reason for these opposing results was undetermined with no correlation to the extent of DARPP-32 loss in the striatum. While it is possible that chance variation in the positioning of the QA lesion could have contributed to the opposing observations given the small group sizes, the influence of AAVLuciferase transduction or Luciferase expression making these pallidal neurons more vulnerable to neurotoxic insult cannot be discounted. Negative impacts of marker proteins have previously been reported (Detrait et al. 2002; Krestel et al. 2004), possibly due to intensive expression of a foreign protein that may cause cellular stress through disruption of degradation mechanisms or accumulation to toxic levels – especially over extended periods of expression. An empty DNA expression cassette may provide a more valid control for AAV vector transduction. With only five rats in each control group and reasonably high amounts of variability with the QA lesion – demonstrated here with only one of the five PBS rats having extensive pallidal cell loss – I was unable to reliably draw any conclusions regarding the vulnerability of the pallidal neurons although it was important to consider their maintenance in respect to the striatal output via the globus pallidus and the impact that it has on motor control in the behavioural testing. 5.8.2.3 Pathological and Behavioural Correlations To investigate whether there was any association between the quantified maintenance of the basal ganglia nuclei and the observed amelioration of functional deficits I performed correlation analysis between the results of each behavioural test and the relative maintenance of DARPP-32 striatal neurons and parvalbumin-positive pallidal neurons (Section 5.7.7). The loss of DARPP-32 striatal neurons showed significant correlation with reductions in the striatal area, TH striatal density and DARPP-32 fibre innervations of the substantia nigra, although none of these were reduced to the same extent as striatal neuron loss. Therefore I choose to directly compare the functional behaviour with the relative DARPP-32 positive cell maintenance, with the assumption that any correlations would also be reflected in these related structures. Separate correlations with parvalbumin-positive neurons in the globus pallidus were analysed due to the lack of any apparent correlation with the maintenance of striatal neurons (Section 5.7.6.2). As the behavioural tests are designed to assess an imbalance in functional control between the brains two hemispheres I performed all correlations against the ratio of neurons remaining in the treated hemisphere compared to the contralateral untreated hemisphere despite the possibility that the contralateral hemisphere may not be entirely unaffected by the AAV-vector delivery or QA lesion. I did not however observe any indications of 153 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF the contralateral hemisphere being influenced in any of the immunocytochemical analysis; although transduction of cortical neurons may have caused a small enhancement in striatal BDNF due to the bilateral projections of cortical neurons (McGeorge and Faull 1989). Both the development of spontaneous ipsilateral forelimb use bias and contralateral sensorimotor neglect showed a positive correlation with the severity of striatal lesioning in the control rats as expected. However with only partial protection of the striatum, the reduced severity of behavioural deficits exhibited by the AAV-BDNF treated rats did not directly reflect maintenance of the striatum but instead correlated with the relative maintenance of parvalbumin expressing pallidal neurons. A correlation with the maintenance of the globus pallidus was also observed in the control rats development of sensorimotor neglect but not spontaneous ipsilateral forelimb use; possibly indicating the greater executive function required in processing the sensory information and making a left / right selection in the “corridor” task. AAV-BDNF treated rats that had a loss of parvalbuminexpressing pallidal neurons were observed to develop ipsilateral forelimb use equivalent to that seen in control rats with similar striatal cell loss, suggesting that the overall AAV-BDNF amelioration of the behavioural impairments was potentially the result of a dynamic interaction between maintenance of the striatal anatomical structure / organisation and striatal output via the globus pallidus. The inducement of rotational behaviour following apomorphine administration also reflected the integrity of the globus pallidus with rats that had no loss of parvalbumin-positive neurons preferentially rotating away from the lesioned striatum, while ipsilateral rotations were generally induced post-QA lesioning when the ipsilateral globus pallidus exhibited neuron loss. Although I was unable to determine whether AAV-BDNF delivery provided any direct neuroprotective support to the pallidal neurons in this investigation, the significant correlations between the maintenance of striatal neurons and attenuation of behavioural deficits was indicative of BDNF-derived trophic support mitigating the extent of disruption to functional behaviour. Given the evidential requirement for pallidal neurons to facilitate normal sensorimotor behaviour, it is probable that efferent striatal projections to the globus pallidus, predominantly arising from the striatal matrix (Gerfen 1985), are better maintained in the AAV-BDNF treated rats independent of pallidal neuron survival. Assessment of DARPP-32 staining in the substantia nigra demonstrated BDNF-enhanced maintenance of the striatonigral projections, arising from both matrix and striosome compartments (Gerfen 1985), however DARPP-32 staining of the globus pallidus was less defined and definitive assessment of the striatopallidal projection survival should be undertaken with tracer studies. 154 Neurotrophic Factor Delivery: AAV-BDNF and AAV-GDNF 5.8.3 Conclusion Despite the vital role that neurotrophic factors, and specifically BDNF, play in maintaining the plasticity and functionality of the striatum, and the disruption of BDNF production / transportation in the HD brain, there has only been limited success to date in alleviating cell loss in the striatum through the administration of BDNF. While a low level of neuroprotection has been previously found (Bemelmans et al. 1999; Kells et al. 2004), these studies together with the current investigations show the significant impact that BDNF can impart on the coordination of functional behaviour and the ultimate need to be able to finely control the extent and localisation of enhanced BDNF expression. To my knowledge, all of the reported BDNF studies to date have used acute administration of neurotoxic chemicals to induce selective cell death in the vulnerable striatal projection neurons in which the cells are probably exposed to more intense insult than the actual neurons are in HD patients in which there is a slow progressive loss. Given the ability to provide protection in these studies, it is therefore conceivable that the continuous expression of neurotrophic factors would actually provide a greater level of support potentially with lower levels of exogenous expression being required; which will additionally limit any adverse effects of the transgene expression. Therefore the delivery of neurotrophic factors to transgenic models in which there is a progression of symptom onset and neuropathological changes, as has been reported for GDNF, may provide a truer reflection of the actual potential of neurotrophic factor based therapy for HD. Ultimately the results of these neurotrophic factor investigations highlight the need to apply caution in the enhancement of endogenously expressed trophic factors, with excessive uncontrolled production potentially deleterious, while tightly controlled expression could be of significant therapeutic benefit. This is of particular importance given that protective treatments are likely to be of greatest efficacy when applied early in the degenerative disease process – potentially even prior to the onset of physical symptoms in HD. Consideration will also need to be given to areas of HD neuropathology beyond the caudate-putamen and whether neurotrophic factor delivery to the striatum is sufficient to attenuate HD symptoms, or if other areas of primary or secondary neurodegeneration are also suited to targeting with neurotrophic factor gene delivery. 155 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP Chapter 6 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6.1 Overview The proposal that apoptotic cell death processes are implicated in Huntington’s disease neurodegeneration (Section 1.7) raises the question of whether enhancing the expression of antiapoptotic factors in the vulnerable striatal projection neurons can reduced their susceptibility to neurotoxic processes occurring in the Huntington’s disease brain. In this investigation I constructed AAV vectors encoding either Bcl-xL or XIAP, using them to transduce striatal neurons prior to an intrastriatal injection of the glutamate analogue QA. Treated rats were observed in behavioural tests undertaken to assess whether the anti-apoptotic factor expression could provide any amelioration of motor impairments following unilateral QA-induced striatal lesioning. While there was some lessening of behavioural deficits in the AAV-Bcl-xL and AAV-XIAP treated rats, I found no quantified reduction of QA-induced pathology in assessed neuronal populations of the basal ganglia. Overall these investigations showed the ability to deliver anti-apoptotic factors to the central nervous system without causing any immediately apparent welfare concerns, however suggest that the enhancement of an isolated pro-survival protein is not sufficient to counteract acute excitotoxic insult of the striatal neurons. 6.2 Procedures Chimeric AAV1/2 vectors were constructed containing cDNA encoding Bcl-xL or XIAP and were assessed with in vitro assays to ensure they directed the expression of biologically functional antiapoptotic factors (Chapter 2; Kells et al. 2006), prior to verifying in vivo transduction and expression in the rodent striatum. Following the AAV vector testing I undertook two in vivo studies, conducted as described (Chapter 4) to assess the influence of Bcl-xL and XIAP expression on QA induced striatal excitotoxicity and associated functional behavioural deficits. In the initial neuroprotective investigation I delivered 4.0µL AAV-Bcl-xL (n = 10*) to a single stereotaxic site within the rodent * n values here represent the final number of rats included in the analysis of the investigations (Section 4.3). 156 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP striatum prior to QA injection at the same stereotaxic coordinates. To further enhance the number of striatal neurons transduced, and their distribution within the striatum, I conducted a second investigation delivering 8.0µL of AAV-Bcl-xL (n = 11) or AAV-XIAP (n = 9) split between two stereotaxic sites in the striatum flanking the QA injection coordinates. These two neuroprotective investigations were undertaken in parallel with the previously described neurotrophic factors studies (Chapter 5) utilising the same groups of control rats which received either AAV-Luciferase (First study: n = 8; Second study: n = 5) or sterile PBS (First study: n = 10; Second study: n = 5) prior to QA lesioning as described (Table 4-2, Table 4-3). 6.3 In Vivo AAV Vector Testing A B C D Figure 6-1 Single delivery site for AAV-Bcl-xL transduction and subsequent Bcl-xL expression (A) Representative coronal sections of a rat brain showing the main regions of HA-tagged Bcl-xL expression three weeks after a single 4.0µL striatal injection of the AAV-Bcl-xL vector. Cells expressing HA-tagged Bcl-xL were largely contained within the injected striatum where a large population of the striatal neurons appeared to be transduced. Higher power images of AAV-Bcl-xL transduced neurons in (B) the striatum, (C) globus pallidus, and (D) substantia nigra. Scale bar = 2mm for whole sections (A); 200µm for (B); 100µm for (C,D). 157 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP To assess the spread of in vivo AAV-Bcl-xL and AAV-XIAP transduction and transgene expression prior to investigating the preventative therapeutic efficacy of over-expressing anti-apoptotic factors in the striatum, I injected naïve rats using the same stereotaxic surgical protocols to be undertaken on the main cohorts of rats in the neuroprotective investigations. The initial single site delivery protocol was determined from the earlier AAV-Luciferase delivery testing (Section 4.4.2) which I then modified for the second two-site delivery study designed to increase the number of transduced cells and better distribute vector transduction around the QA-injection site. Three weeks post-AAV delivery the enhanced Bcl-xL (Single site: Figure 6-1; Two-site: Figure 6-2) and XIAP (Two-site: Figure 6-3) expression was visualised by immunocytochemical staining against the HA-epitope attached to the C-terminal of the anti-apoptotic factors. Both AAV-Bcl-xL and AAV-XIAP transduced cells were observed in an extensive but often irregularly dispersed distribution A B C D Figure 6-2 AAV-Bcl-xL transduction and Bcl-xL expression following two-site striatal delivery (A) Representative coronal sections of a rat brain showing the main regions of HA-tagged Bcl-xL expression three weeks after two 4.0µL striatal injections of the AAV-Bcl-xL vector. Cells expressing HA-tagged Bcl-xL were largely contained within the injected striatum where a large population of the striatal neurons appeared to be transduced. Higher power images of AAV-Bcl-xL transduced neurons in (B) the striatum, (C) globus pallidus, and (D) substantia nigra. Scale bar = 2mm for whole sections (A); 100µ m for higher magnification images (B-D). 158 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP throughout the rostral-caudal extent of the striatum. Transgenic Bcl-xL protein appeared to be distributed throughout the cytosol of transduced neurons (Figure 6-2B) making identification of individual transduced neurons less evident compared to the nuclear localisation of XIAP expression (Figure 6-3B). A population of cells within the globus pallidus and substantia nigra pars compacta ipsilateral to the injected striatum were also HA-positive indicating transportation of the AAVvectors and transduction of cells within the striatal target nuclei. The ipsilateral substantia nigra pars reticulata also displayed HA-immunoreactivity although this was mainly restricted to the striatonigral axonal fibres with very few identifiable HA-positive cell bodies. I also detected the presence of HAtagged proteins throughout fibres in the cerebral cortex ipsilateral to the two-site intrastriatal injection of AAV-Bcl-xL, suggesting either low level transduction of these cortical neurons – a potential effect of the larger volume of vector delivered – or retrograde transport of Bcl-xL produced in the striatum. A B C D Figure 6-3 Spread of AAV-XIAP transduction and subsequent XIAP expression (A) Representative coronal sections of a rat brain showing the main regions of HA-tagged XIAP expression three weeks after two 4.0µL striatal injections of the AAV-XIAP vector (A). Cells expressing HA-tagged XIAP were largely contained within the injected striatum where a large population of the striatal neurons appeared to be transduced. Higher power images of AAV-XIAP transduced neurons in (B) the striatum, (C) globus pallidus, and (D) substantia nigra. Scale bar = 2mm for whole sections (A); 100µ m for higher magnification images (B-D). 159 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6.4 Bcl-xL and XIAP Expression Level To determine the quantity of Bcl-xL and XIAP expression within the striatum following AAVmediated gene delivery I injected additional cohorts of rats with the AAV-Bcl-xL or AAV-XIAP vectors using the same surgical protocol employed for the neuroprotective investigation. The rats were euthanised three weeks post-surgery and their striatal tissue separately isolated for ELISAbased quantification of the Bcl-xL or XIAP content (Section 4.6). Expression was assessed three weeks post-AAV delivery to provide a measure of the increased anti-apoptotic factor expression equivalent to the expression levels in the main neuroprotective study groups at the time of QA injection. Bcl-xL expression in the striatum following the initial single site AAV-Bcl-xL injection was enhanced ~20-fold to 1200 ± 300 ng / mg total protein compared with the 49 ± 2 ng / mg total protein in the control AAV-Luciferase injected striatum (Mann-Whitney P < 0.01; Figure 6-4A). The contralateral striatum displayed a small increase in Bcl-xL expression at 66 ± 4 ng / mg total protein indicating a small amount of expression crossing over into the non-injected striatal hemisphere. The control AAV-Luciferase vector injections did not induce a significant alteration in Bcl-xL expression with the injected striatum having 49 ± 2 ng Bcl-xL / mg total protein and the non-injected striatum 39 ± 7 ng / mg total protein. Significantly higher Bcl-xL expression was generated following the 8.0µL twosite AAV-Bcl-xL injections with a ~2.5-fold increase over the single site injected striatum to 3100 ± 700 ng / mg total protein (~60-fold increase over controls; Mann-Whitney P < 0.05). A significant amount of transgenic Bcl-xL (120 ± 10 ng / mg total protein) was again detected in the non-injected contralateral striatum (Mann-Whitney P < 0.01). AAV-XIAP delivery resulted in a much lower quantity of transgenic protein expression than the AAV-Bcl-xL delivery with the injected striatum containing 150 ± 20 ng XIAP / mg total protein, although this represented a comparable ~70-fold enhancement over the endogenous XIAP expression detected in the AAV-Luciferase injected control striatum (Mann-Whitney P < 0.01; Figure 6-4B). A small amount of hemispherical crossover of transgenic protein expression was again detected in the contralateral non-treated striatum with 4.3 ± 0.3 ng XIAP / mg total protein detected compared with the 1.7 ± 0.7 ng / mg total protein in the control rats (Mann-Whitney P < 0.05). 160 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP A B Figure 6-4 ELISA quantification of Bcl-xL and XIAP expression in the striatum The level of (A) Bcl-xL or (B) XIAP protein expression in striatal tissue isolated three weeks after infusion of AAV-Bcl-xL, AAV-XIAP or AAV-Luciferase was quantified by ELISA. Total protein in the samples was measured by BCA assay to normalise the individual samples. Mann-Whitney tests: comparison with AAV-Luciferase * P < 0.05, ** P < 0.01; comparison with two-site AAV-Bcl-xL ‡ P < 0.01. 6.5 Functional Behaviour Assessment Throughout the neuroprotective studies I assessed the development of functional deficits by analysing the performance of each rat in behavioural tests designed to show hemispherical imbalances in brain function arising as a result of a unilateral lesion in the basal ganglia. The functional behavioural tests undertaken as previously described (Section 4.5) involved spontaneous forelimb use analysis, drug-induced rotational behaviour following either apomorphine or amphetamine dosing, and preferential left / right side food selection. All the rats were assessed with baseline measurements taken both before and after AAV delivery and for 5-7 weeks post-QA injection (Table 4-2, Table 4-3). 6.5.1 Spontaneous forelimb use Assessment of forelimb usage during spontaneous exploratory activity was undertaken by observing the rats rearing behaviour when placed into a clear cylinder (Section 4.5.1). Baseline assessment of the naïve rats prior to any treatment generally showed non-biased usage of both left and right forelimbs. AAV-Bcl-xL delivery in the initial single site delivery investigation did not induce any alterations in forelimb use following vector delivery (Figure 6-5A). Although initially appearing to attenuate development of the ipsilateral forelimb use bias seen in control rats, the AAV-Bcl-xL treated rats progressively acquired a strong preferential use of their ipsilateral forelimb while the 161 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP A B Figure 6-5 Spontaneous ipsilateral forelimb use asymmetry scores Plots displaying changes in group ipsilateral asymmetry scores as quantified during spontaneous exploration of the forelimb use assessment cylinder in (A) the first AAV-Bcl-xL and (B) second AAV-Bcl-xL / AAV-XIAP investigations. Ipsilateral asymmetry scores represent the combined use of the ipsilateral forelimb, as a percentage of the total left and right forelimb usage, for rearing, initial contact of the cylinder wall during an exploratory rear and landing. * Un-paired t-test between treatment groups: P < 0.05. Two-way RM ANOVA relative to the control group: † Bcl-xL P = 0.053, XIAP P < 0.001. 162 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP control rats appeared to undergo spontaneous recovery three-weeks post QA such that AAV-Bcl-xL provided no overall attenuation of the contralateral forelimb motor control deficit. In the second two-site vector delivery investigation the control rats developed and maintained a strong ipsilateral forelimb bias that tended to be restricted by the prior delivery of AAV-Bcl-xL but was not quite statistically significant (Two-way RM ANOVA P = 0.053; Figure 6-5B). Delivery of AAV-XIAP in the second investigation induced a small but significant shift towards greater usage of the treated rats’ contralateral forelimb (44 ± 4% ipsilateral asymmetry) prior to QA lesioning, compared with the unbiased 53 ± 3% ipsilateral forelimb use in control rats (Unpaired ttest P < 0.05; Figure 6-5B). Following QA injection the AAV-XIAP treated rats showed an immediate shift back to non-biased forelimb usage with the over-expression of XIAP resulting in the complete attenuation of the ipsilateral forelimb use bias exhibited by the control treated rats (Twoway RM ANOVA P < 0.001; Figure 6-5B) Comparison of total activity level between the treatment groups showed that while all rats have a reduction in the number of exploratory rears from initial baseline testing, the rats with enhanced antiapoptotic factor expression overall showed higher levels of exploratory activity. With AAV-Bcl-xL treated rats in the first study and AAV-XIAP treated rats in the second study displaying significantly greater rearing behaviour than the control rats (Two-way RM ANOVA P < 0.05; Figure 6-6). A B Figure 6-6 Spontaneous exploratory rearing activity Plots displaying the number of exploratory rears in the cylinder during the five minute assessment of spontaneous forelimb use in (A) the first AAV-Bcl-xL and (B) second AAV-Bcl-xL / AAV-XIAP investigations. No statistics were performed on the pre-QA baseline data. Two-way RM ANOVA relative to the control group: † P < 0.05; ‡ Bcl-xL P = 0.11, XIAP P < 0.05. 163 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6.5.2 Drug-induced rotational behaviour Apomorphine- and amphetamine-induced rotational motor activity was analysed to assess alterations in striatal dopamine signalling following unilateral QA injection and the subsequent loss of dopamine receptive striatal neurons (Section 4.5.3). Analysis of the rotation data showed inconsistencies in rotational activity and direction between individual rats within the treatment A B Figure 6-7 Apomorphine-induced rotational behaviour Plots displaying each rat’s rotational behaviour towards the treated side of their brain as a percentage of total rotations following subcutaneous administration of 1mg/kg of apomorphine in (A) the first AAV-Bcl-xL and (B) second AAV-Bcl-xL / AAV-XIAP investigations. Rats that had less than 0.5 rotations per minute were excluded from this analysis. Lines represent the median result for each group at each time point. 164 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP groups – possibly resultant of minor variations in the actual positioning of the AAV-vector or QA injections – complicating the assessment of dopamine related functional motor impairment. Therefore statistical analysis was restricted to the assessment of total rotation rates post QA, independent from the direction of induced rotation. 6.5.2.1 Apomorphine As expected from the earlier QA lesion testing (Section 3.3.4.1), the administration of apomorphine to rats with a unilateral striatal lesion tended to induce rotational motor behaviour towards the lesioned hemisphere (Figure 6-7), in contrast to a lower level of unbiased motor activity induced prior to QA lesioning (Figure 6-8). The extent of apomorphine-induced rotations varied significantly between individual rats in the two anti-apoptotic factor investigations with the rats showing up to 20 rotations per minute irrespective of their treatment, and with no significant overall differences in rotation rates between the groups in either investigation (Figure 6-8A,B). For reliable assessment of rotational behaviour I excluded trial data for rats that displayed less than 0.5 rotations per minute following apomorphine administration. Baseline and post-AAV trials showed a wide spread in the extent and direction of rotational behaviour which was skewed towards a contralateral dominance by the selective treatment of the individual rats more dominant hemisphere (Figure 6-7A,B). The AAV vector delivery and transgene expression did not appear to be having any direct influence on the rotational behaviour prior to QA. Following QA injection the control rats predominantly had a strongly ipsilateral rotational bias while A B Figure 6-8 Total apomorphine-induced rotations Plots displaying the mean number of apomorphine-induced rotations for each group in (A) the first AAV-Bcl-xL and (B) second AAV-Bcl-xL / AAV-XIAP investigations following subcutaneous administration of 1mg/kg apomorphine. No significance was found between treatment groups and controls post-QA. 165 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP a number of anti-apoptotic factor expressing rats showed less unidirectional behaviour with a slight majority of AAV-Bcl-xL and AAV-XIAP treated rats in the second study actually displaying a greater preference towards contralateral rotations (Figure 6-7B). Although not statistically analysed, the observed changes to rotational behaviour in the second investigation were indicative of Bcl-xL and XIAP derived alterations to the QA-induced neuropathological disruptions. 6.5.2.2 Amphetamine Rotational behaviour induced by amphetamine administration was only assessed in the initial AAVBcl-xL study where greater rotational behaviour was induced in AAV-Bcl-xL treated rats following A B Figure 6-9 Amphetamine-induced rotational behaviour Plots displaying the rotational behaviour induced following an i.p. injection of 5mg/kg of amphetamine in the first AAV-Bcl-xL investigation. (A) Individual rats’ rotational behaviour towards the treated (ipsilateral) side of their brain as a percentage of total rotations and (B) the total rotation rate for each group. Rats that had less than 0.1 rotations per minute were excluded from this analysis. Lines in (A) represent the median result for each group at each time point. No significance was found in rotation rates between AAV-Bcl-xL treatment rats and controls post-QA. 166 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP QA-injection than in the AAV-Luciferase / PBS treated control rats (Figure 6-9B). However the extent of this behaviour varied extensively between individual rats, with many not displaying any persistent rotational behaviour following amphetamine dosing and therefore being excluded from the assessment of rotation direction. Analysis of the number of ipsilateral rotations as a percentage of total rotational behaviour found that post-QA the majority of rats in both the AAV-Bcl-xL and control treated groups predominantly or solely performed unidirectional rotations, with the majority rotating towards the lesioned hemisphere (Figure 6-9A). The slight contralateral rotation preference observed in the pre-QA baseline trials was due to the selection of the dominant brain hemisphere for treatment. No statistical analysis was performed on the directional rotation data due to the lack of consistency between similarly treated rats in their extent or direction of rotation. 6.5.3 Sensorimotor “corridor” task Inclusion of the “corridor” task in the second study provided an assessment of the impact that overexpressing anti-apoptotic factors had on the development of sensorimotor neglect following QAlesioning. During baseline and post-AAV testing all treatment groups displayed a fairly even distribution of sugar pellet retrievals from both the left and right sides of the corridor with only the AAV-XIAP treated rats displaying an increase in the proportion of retrievals from the ipsilateral side Figure 6-10 Preferential food selection in the sensorimotor “corridor” task Plot displaying group changes in the number of sugar pellet retrievals from the same side as the treated striatum as a percentage of the 20 retrievals per trial. No significance was found between treatment groups and controls post-QA. 167 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP prior to QA injection (paired t-test P < 0.05; Figure 6-10). The slight preference for contralateral retrievals in all groups during baseline testing is resultant of the intentional selection of each rat’s dominant brain hemisphere for treatment. Post-QA the development of contralateral sensorimotor neglect, resulting in preferential ipsilateral sugar pellet retrieval, was strongest in the control rats (77 ± 2%) but also observed in both the AAV-XIAP (73 ± 1%) and AAV-Bcl-xL (65 ± 1%) treated groups with no significant alterations between the different treatments (Two-way RM ANOVA postQA P > 0.05). 6.6 Immunocytochemical Analysis Immunocytochemical staining was undertaken to determine if AAV-vector mediated expression of the anti-apoptotic factors Bcl-xL or XIAP supplied any neuroprotective support against QA-induced excitotoxic stress in the striatum of adult rats. Following the completion of behavioural testing I processed the rat brains for immunocytochemical analysis, sectioning coronally through striatum, globus pallidus and substantia nigra for immunostaining against various neuronal makers. 6.6.1 AAV-mediated transgene expression and QA lesioning of the striatum Initial assessment of the surviving AAV-mediated transgene expression was undertaken by visualising the HA-tagged anti-apoptotic factors or control Luciferase expression within the striatum. The extent of the QA-induced striatal lesioning was visually assessed by both DARPP-32 and NeuN immunostaining with stereological quantification to determine the maintenance of the vulnerable DARPP-32 positive striatal projection neurons. 6.6.1.1 Study 1 – AAV-Bcl-xL Delivery In the first study AAV-Bcl-xL was delivered to the striatum at a single stereotaxic injection site prior to QA injection at the same coordinates. Using adjacent striatal sections individually immunostained for HA, DARPP-32 and NeuN to show the distribution of enhanced Bcl-xL expression and neuronal maintenance (Figure 6-11) I observed a generally positive correlation between immunoreactivity, although HA staining was less extensive than the neuronal markers. Intense HA staining of cellular processes in the striatum, suggesting intracellular accumulation of transgenic Bcl-xL, made identification of transduced neurons less distinct (Figure 6-11C). Despite a scattering of DARPP-32 and NeuN-positive cells within the area of striatal lesioning – particularly in the more distal “transition zone” – the extent of neuronal sparing varied considerably between individual AAV-Bcl168 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP xL treated rats (Figure 6-11D-I) and was also observed in AAV-Luciferase / PBS treated control rats (Figure 5-13D-I). Overall there was no visually apparent evidence that prior AAV-Bcl-xL delivery reduced striatal cell loss. A HA D G DARPP-32 NeuN B E H C F I Figure 6-11 Single injection site AAV-Bcl-xL transduction and striatal cell maintenance Representative images from adjacent striatal sections showing AAV-mediated Bcl-xL expression and neuronal cell loss seven weeks post-QA. (A-C) HA-immunostaining displayed the distribution of Bcl-xL expression within the striatum resultant of AAV-Bcl-xL transduction prior to QA injection. Maintenance of striatal neurons was detected by (D-F) DARPP-32 and (G-I) NeuN immunostaining. Scale bar = 2mm for (A,D,G); 200µ m for (B,E,H); 50µm for (C,F,I). 169 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6.6.1.2 Study 2 – AAV-Bcl-xL and AAV-XIAP Delivery The second investigation of anti-apoptotic factors in which AAV vectors encoding either Bcl-xL or XIAP were injected into two striatal sites flanking the QA injection coordinates showed some maintenance of transgene expression within the striatum. As with the previous AAV-Bcl-xL study, the striatal transgene expression was still only present in areas that also contained DARPP-32 and NeuN expression for both AAV-Bcl-xL (Figure 6-12) and AAV-XIAP (Figure 6-13) treated rats. With reasonably defined areas of striatal lesioning visualised with DARPP-32 or NeuN staining, the brains expressing enhanced levels of anti-apoptotic factors were not visually discernable from AAVLuciferase / PBS treated control brains (Figure 5-15). A HA D G DARPP-32 NeuN B E H C F I Figure 6-12 Dual injection site AAV-Bcl-xL transduction and striatal cell maintenance Representative images from adjacent striatal sections showing AAV-mediated Bcl-xL expression and neuronal cell loss eight weeks post-QA. (A-C) HA-immunostaining displayed the distribution of Bcl-xL expression within the striatum resultant of AAV-Bcl-xL transduction prior to QA injection. Maintenance of striatal neurons was detected by (D-F) DARPP-32 and (G-I) NeuN immunostaining. Scale bar = 2mm for (A,D,G); 200µ m for (B,E,H); 50µm for (C,F,I). 170 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP A D HA G DARPP-32 NeuN B E H C F I Figure 6-13 Dual injection site AAV-XIAP transduction and striatal cell maintenance Representative images from adjacent striatal sections showing AAV-mediated XIAP expression and neuronal cell loss eight weeks post-QA. (A-C) HA-immunostaining displayed the distribution of Bcl-xL expression within the striatum resultant of AAV-XIAP transduction prior to QA injection. Maintenance of striatal neurons was detected by (D-F) DARPP-32 and (G-I) NeuN immunostaining. Scale bar = 2mm for (A,D,G); 200µ m for (B,E,H); 50µm for (C,F,I). 6.6.1.3 Stereological Analysis – DARPP-32 Quantification of the DARPP-32 immunopositive neurons using optical fractionator stereology probes in the first AAV-Bcl-xL study estimated the number of DARPP-32 positive neurons remaining within the sampled region of the treated caudate-putamen at 1,300,000 ± 100,000 in the AAV-Bcl-xL treated rats compared with 1,150,000 ± 120,000 in the control AAV-Luciferase / PBS treated rats (Figure 6-14A; Coefficient of Error ≤ 0.04). The adjacent intact contralateral striatum contained an overall average across all rats of 2,200,000 ± 60,000 (Coefficient of Error ≤ 0.02). In the second investigation I limited the stereological analysis of DARPP-32 positive neurons to a smaller portion of the striatum centred around the QA lesion containing an estimated 820,000 ± 90,000 DARPP-32 positive neurons in AAV-Bcl-xL treated rats, 890,000 ± 100,000 in AAV-XIAP 171 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP treated rats and 760,000 ± 60,000 in control AAV-Luciferase / PBS treated rats (Figure 6-14C; Coefficient of Error ≤ 0.06). The corresponding contralateral striatal region was estimated to contain an overall average across all treated rats of 1,530,000 ± 30,000 DARPP-32 positive neurons (Coefficient of Error ≤ 0.03). First AAV-Bcl-xL Investigation A B Second AAV-Bcl-xL / AAV-XIAP Investigation C D Figure 6-14 Maintenance of DARPP-32 striatal projection neurons Graphs displaying the actual and relative numbers of DARPP-32 positive neurons in the striatum as estimated by optical fractionator stereological analysis of the treated (ipsilateral) and intact (contralateral) striatal hemispheres in (A,B) the first AAV-Bcl-xL and (C,D) second AAV-Bcl-xL / AAV-XIAP neuroprotective studies. Population estimates were determined from 12 sections spanning 3.56mm of the striatum for the first study and 7 sections spanning 1.96mm of the anterior striatum for the second study. Relative cell counts represent the number of DARPP-32 positive neurons within the lesioned striatum as a proportion of DARPP-32 expressing neurons in the contralateral intact striatum estimated independently for each rat. Statistical analysis revealed no significance differences between treatment groups at the 5% level (P > 0.05). 172 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP Analysis of the stereological estimates to assess the relative maintenance of DARPP-32 positive striatal neurons showed the control AAV-Luciferase / PBS treated rats to have 52 ± 4% maintenance in the initial study and 47 ± 4% in the second investigation. Over-expression of an anti-apoptotic factor – Bcl-xL or XIAP – in the striatum prior to QA injection did not significantly enhance the maintenance of DARPP-32 neurons. AAV-Bcl-xL treated rats had 57 ± 5% maintenance of DARPP-32 neurons in the first study (Figure 6-14B) and 52 ± 4% in the second (Figure 6-14D). Delivery of AAV-XIAP was estimated to have enhanced the maintenance of DARPP-32 positive neurons by ~23% to 58 ± 7% relative survival, although this failed to be statistically significance (P = 0.20; Figure 6-14D). 6.6.1.4 Striatal Atrophy First Single Injection AAV-Bcl-xL Study Second Dual Injection AAV-Bcl-xL / AAV-XIAP Study A C B D Figure 6-15 Atrophy of the lesioned striatum Graphs displaying the size of the lesioned striatum as a proportion of the intact contralateral striatal area, and plots correlating striatal atrophy with DARPP-32 positive cell loss in the striatum. (A,B) First AAV-Bcl-xL study and (C,D) second AAV-Bcl-xL / AAV-XIAP neuroprotective investigation. Cross-sectional striatal area was measured on 12 coronal sections for the initial study and 7 sections for the second study. Combined correlation analysis between the striatal atrophy and the loss of DARPP-32 positive neurons displayed Deming linear regression correlations for both the first (B; slope = 0.51 ± 0.06, P < 0.001) and second (D; slope = 0.53 ± 0.08, P < 0.001) anti-apoptotic factor investigations. R values represent Pearson correlation coefficient independent of treatment. 173 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP As an alternative measure of the overall striatal maintenance I analysed the volume of the striatum which was observed to have significantly atrophied in most cases, with an associated enlargement of the adjacent lateral ventricle. By using contours drawn to perform stereological cell estimates, I calculated the striatal volume relative to the contralateral hemisphere for each rat. The AAVmediated delivery of anti-apoptotic factors did not alter the extent of striatal atrophy following QA lesioning in either study. In the initial investigation the AAV-Bcl-xL injected striatum contracted to 79 ± 2% of the contralateral volume compared with 73 ± 2% in control AAV-Luciferase / PBS treated rats (Figure 6-15A). Similarly, in the second study the striatum was reduced to 77 ± 2% of the contralateral striatal volume in AAV-Bcl-xL treated rats, 76 ± 4% in AAV-XIAP treated rats and 75 ± 3% in the control group of rats (Figure 6-15C). Analysis of the correlation between the relative striatal area and the maintenance of DARPP-32 positive striatal neurons showed a direct relationship with the loss of striatal neurons following QA injection ultimately resulting in a reduction in the overall striatal volume (Figure 6-15B, D). Neither AAV-Bcl-xL nor AAV-XIAP delivery prior to the QA injection significantly altered this correlation. 6.6.2 Krox-24 expression Stereological analysis of krox-24 expressing neurons was performed in the second investigation to further confirm the loss of striatal neurons using an alternative marker for the striatal neurons. The distribution of krox-24 positive neurons matched with the DARPP-32 immunostaining although being a transcription factor the krox-24 immunostaining was restricted to the nucleus resulting in more distinct staining (Figure 6-16). Quantification of the krox-24 positive neurons was performed using optical fractionator probes across eight equispaced striatal sections with the number of neurons estimated at 1,060,000 ± 100,000 in AAV-Bcl-xL treated rats and 1,280,000 ± 150,000 following AAV-XIAP treatment compared with the 1,060,000 ± 110,000 krox-24 positive cells in AAVLuciferase / PBS control rats (Figure 6-17A; Coefficient of Error ≤ 0.05). The corresponding contralateral striatal region contained an estimated average of 2,160,000 ± 40,000 krox-24 positive cells (Coefficient of Error ≤ 0.03). Overall the quantified maintenance of krox-24 positive neurons reflected the DARPP-32 analysis with 50 ± 5% relative maintenance of krox-24 neurons in the control rats that was not significantly enhanced by the prior delivery of either AAV-Bcl-xL (50 ± 5%) or AAV-XIAP (58 ± 7%; Figure 6-17B). Correlation of the krox-24 expression with DARPP-32 cell maintenance demonstrated a parallel link between the maintenance of krox-24 expression and DARPP-32 expression following QA lesioning 174 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP that was independent of the prior AAV-mediated anti-apoptotic factor expression or control treatment (Figure 6-17C). krox-24 A D G AAV-Bcl-xL AAV-XIAP AAV-Luciferase DARPP-32 B C E F H I Figure 6-16 Krox-24 expression in the striatum Representative images displaying the expression of krox-24 in striatal neurons following QA lesioning in (A,B) AAV-Bcl-xL, (D,E) AAV-XIAP, and (G,H) AAV-Luciferase treated rats. (C,F,I) Adjacent sections (copied from Figure 6-12E, Figure 6-13E and Figure 5-15D) immunostained for DARPP-32 show the distribution correlation between krox-24 and DARPP-32 positive cells in the striatum. Scale bar = 2mm for whole sections and 200µm for higher magnification images. 175 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP A B C Figure 6-17 Maintenance of krox-24 expressing striatal neurons Graphs displaying the (A) actual and (B) relative number of striatal neurons expressing krox-24 as estimated by optical fractionator stereological cell counting in the treated (ipsilateral) and intact (contralateral) striatal hemisphere in the second anti-apoptotic factor investigation. Relative cell counts represent the number of krox-24 expressing cells within the lesioned striatum as a proportion of krox-24 positive cells present in the contralateral striatum. (C) Plot showing the correlation between the relative maintenance of krox-24 positive neurons and DARPP-32 expressing neurons in the striatum. Trend line shows the overall Deming linear regression correlation across all treatment groups: slope = 1.05 ± 0.07, P < 0.001. R value represents overall Pearson correlation coefficient independent of treatment. 6.6.3 Analysis of transduced striatal cells To further assess the state of AAV-vector transduced cells in the striatum at the conclusion of the investigations, I performed double-label immunofluorescent staining of the transgene expression with HA and either DARPP-32 or krox-24. Previous assessment of AAV-Luciferase transduced cells in the striatum showed co-expression of Luciferase with NeuN, but not with DARPP-32 or krox-24 ( Figure 5-21). In contrast, for both AAV-Bcl-xL and AAV-XIAP treated rats I observed HA-positive cells co-labelling with DARPP-32 and krox-24 in the striatum (AAV-Bcl-xL: Figure 176 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6-18; AAV-XIAP: Figure 6-19). While the majority of HA-positive neurons in both AAV-Bcl-xL and AAV-XIAP treated rats co-expressed DARPP-32, krox-24 expression appeared highly variable with not all of the transduced HA-positive cells co-labelling. However this irregularity in krox-24 labelling intensity was also seen in non-transduced striatal neurons, so may not be related to the enhanced expression of the anti-apoptotic factors. A AAV-Bcl-xL HA DARPP-32 B AAV-Bcl-xL HA krox-24 Figure 6-18 Immunofluorescent double-labelling of AAV-Bcl-xL transduced striatal neurons Representative single-slice confocal images from an AAV-Bcl-xL treated rat from the second investigation showing AAV-Bcl-xL transduced cells (red) within the QA-injected striatum and immunofluorescent staining for (A) DARPP-32 expression and (B) krox-24 expression (both displayed in green). Co-labelling of the HA-positive cells with DARPP-32 or krox-24 was found in the striatum, although many of the AAV-Bcl-xL transduced cells only displayed weak krox-24 labelling (arrowheads). Scale bar: 30µm 177 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP A AAV-XIAP DARPP-32 HA B AAV-XIAP HA krox-24 Figure 6-19 Immunofluorescent double-labelling of AAV-XIAP transduced striatal neurons Representative single-slice confocal images from an AAV-XIAP treated rat from the second investigation showing AAV-XIAP transduced cells (red) within the QA-injected striatum and immunofluorescent staining for (A) DARPP-32 expression and (B) krox-24 expression (both displayed in green). Co-labelling with DARPP-32 or krox-24 was evident for most HA-positive cells. However, a few HA-positive cells completely lacked DARPP-32 labelling (arrows) and krox24 expression appeared reduced in a large proportion of AAV-XIAP transduced cells (arrowheads). Scale bar: 30µm 6.6.4 Dopaminergic innervations of the striatum To visually assess the maintenance of dopaminergic input into the striatum I performed immunostaining for TH expression. The extent of TH-immunoreactivity appeared to be altered in the striatum of all treatment groups with a clear loss of TH proximal to the site of QA injection (Figure 6-20). Additionally, the structural organisation of TH-positive fibres appeared to be disrupted in a larger portion of the striatum surrounding the lesion centre with the normally diffuse TH labelling of the striatal matrix becoming condensed into tighter bundles surrounding what would appear to be an enlargement of striosome patches. Neither AAV-Bcl-xL nor AAV-XIAP delivery to the striatum prior to QA lesioning provided any attenuation of the disruption to the structural organisation of dopaminergic innervations. 178 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP AAV-Luciferase A AAV-Bcl-xL B AAV-XIAP C iii i v iv vi ii i iii v ii iv vi Figure 6-20 Tyrosine hydroxylase expression in the striatum Representative images displaying TH-immunoreactivity in the striatum following QA injection in (A) AAV-Luciferase, (B) AAV-Bcl-xL, and (C) AAV-XIAP treated rats. The distribution of THimmunopositive fibres was affected in the striatum of all treatment groups with a reduction in THexpression at the lesion core (bottom images), and a condensing of the TH-positive matrix component of the striatum in the surrounding QA-lesioned striatum (middle images). Dashed lines provide an approximate outline of the lesion core largely devoid of TH (bottom images), and the limits of the striatal alterations induced by QA (middle images). Scale bar = 1mm for top images and 200µ m for higher magnification images. 6.6.5 Cortical projection fibres in the striatum Corticostriatal projections were assessed by SMI32 expression in the striatum. Following QA lesioning there was an apparent increase in the staining of SMI32-positive fibres in the striatum relative to the intact contralateral striatum (Figure 6-21). SMI32 immunostaining was most intensely enhanced in the striatum proximal to the site of QA injection. Additionally the normally dispersed patches of SMI32 staining became a dominant feature filling the striatum and often appearing to be arranged into disjointed bands in the less severely damaged area surrounding the centre of the QA 179 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP lesion (Figure 6-21D). This alteration in cortical projections following QA lesioning did not appear to be affected by the prior enhancement of Bcl-xL (Figure 6-21F-H) or XIAP (Figure 6-21I-K) expression in the striatal neurons. B C D E G H J K A AAV-Luciferase F AAV-Bcl-xL I AAV-XIAP Figure 6-21 SMI32 expression in the striatum Representative images displaying SMI32-immunoreactivity in the contralateral intact striatum (left hemispheres and (B,C)) and following QA injection in (A,D,E) AAV-Luciferase, (F-H) AAV-Bcl-xL and (I-K) AAV-XIAP treated rats. The arrangement of SMI32-immunopositive fibres was affected in the striatum of all treatment groups with an increase in the density of SMI32 positive patches, particularly around the QA-lesion core. Scale bar = 1mm for whole sections; 200µ m for middle column of images; 50µ m for high magnification images on the right. 180 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6.6.6 Striatal projection nuclei With the AAV-vector transduction extending to cells in the target nuclei of the striatal axonal projections (Figure 6-2, Figure 6-3), I used immunocytochemical staining to assess whether the limited enhancement of Bcl-xL or XIAP expression in the nigral or pallidal neurons had any bearing on their survival following QA lesioning of the striatum. 6.6.6.1 Substantia Nigra Assessment of the substantia nigra was performed by immunocytochemical staining of the dopaminergic neurons in the pars compacta with TH, a population of GABA-receptive neurons in the pars reticulata using parvalbumin, and the maintenance of GABAergic innervations by striatonigral projection neurons by measuring the density of DARPP-32 immunostaining. I did not observe any apparent changes to the number of TH-positive neurons in either the controls (Figure 5-27), or antiapoptotic factor treated rats (AAV-Bcl-xL: Figure 6-23A; AAV-XIAP: Figure 6-24A) with similar TH expression in each hemisphere. Similarly with the GABAergic neurons in the pars reticulata there was no apparent reduction in parvalbumin expressing neurons following QA striatal lesioning (Figure 6-23B, Figure 6-24B). While the nigral neurons appeared largely unaffected, the loss of GABAergic striatal projection neurons did reduce the intensity of DARPP-32 fibres innervating the substantia nigra (Figure 6-23C, Figure 6-24C). The reduced GABAergic innervations were quantitatively assessed by measuring the density of immunostaining in the ipsilateral substantia nigra A B Figure 6-22 DARPP-32 innervations of the substantia nigra (A) Graph showing the density of DARPP-32 immunoreactivity within the substantia nigra, ipsilateral to the treated striatum, as a proportion of DARPP-32 immunostaining in the contralateral substantia nigra. DARRP-32 density was measured on three coronal sections through the substantia nigra. (B) Plot displaying the correlation between DARPP-32 positive fibre innervations of the substantia nigra and maintenance of the DARPP-32 positive neurons. Deming linear regression correlation across all treatment groups: slope = 0.67 ± 0.15, P < 0.001. R value represents overall Pearson correlation coefficient independent of treatment. 181 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP relative to the contralateral hemisphere in the same coronal plane. The control rats displayed a 29 ± 3% reduction in the density of DARPP-32 fibre innervations that was not significantly altered by either AAV-Bcl-xL delivery (20 ± 3% reduction, P = 0.08) or AAV-XIAP treatment (20 ± 5% reduction, P = 0.15; Figure 6-22A). When compared with the maintenance of DARPP-32-positive striatal neurons, the reduction in DARPP-32 fibre immunostaining within the substantia nigra showed a strong, direct correlation with the loss of the striatal neurons (Pearson r = 0.65; Figure 6-22B). Contralateral Ipsilateral A Tyrosine Hydroxylase B Parvalbumin C DARPP-32 Figure 6-23 Visual assessment of the substantia nigra in AAV-Bcl-xL treated rats Representative images of the substantia nigra from an AAV-Bcl-xL treated brain displaying the comparison between the treated (ipsilateral) and un-treated (contralateral) hemispheres. Immunocytochemical staining of (A) TH-positive neurons in the pars compacta, (B) parvalbuminpositive neurons in the pars reticulata, and (C) DARPP-32 positive neurons and efferent fibres. Scale bar = 2mm for whole sections and 200µ m for higher magnification images. 182 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP Contralateral Ipsilateral A Tyrosine Hydroxylase B Parvalbumin C DARPP-32 Figure 6-24 Visual assessment of the substantia nigra in AAV-XIAP treated rats Representative images of the substantia nigra from an AAV-XIAP treated brain displaying the comparison between the treated (ipsilateral) and un-treated (contralateral) hemispheres. Immunocytochemical staining of (A) TH-positive neurons in the pars compacta, (B) parvalbuminpositive neurons in the pars reticulata, and (C) DARPP-32 positive neurons and efferent fibres. Scale bar = 2mm for whole sections and 200µ m for higher magnification images. 183 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6.6.6.2 Globus Pallidus Assessment of the globus pallidus was undertaken by immunostaining for parvalbumin and NeuN which displayed a loss of pallidal neurons in the ipsilateral hemisphere for AAV-Luciferase (refer previous chapter Figure 5-30A,B), AAV-Bcl-xL (Figure 6-26A,B) and AAV-XIAP (Figure 6-27A,B). Immunostaining for DARPP-32 generally showed a reduced density of DARPP-32 positive fibres within the globus pallidus consistent with the loss of striatopallidal projections postQA for all treatment groups despite the maintenance of DARPP-32-positive cells in the striatum directly adjacent to the globus pallidus (AAV-Luciferase, Figure 5-30C; AAV-Bcl-xL, Figure 6-26C; AAV-XIAP, Figure 6-27C). Neuronal maintenance in the globus pallidus was determined by quantifying the parvalbuminpositive neurons using stereological estimates of the ipsilateral and contralateral pallidal nuclei assessed over four equispaced coronal sections. As previously mentioned (Section 5.7.6.2), I found a significant split between the PBS vehicle and AAV-Luciferase control groups with no apparent alteration to the number of parvalbumin-positive pallidal neurons in the majority of PBS treated rats, in contrast to the extensive loss of parvalbumin expressing pallidal neurons in AAV-Luciferase treated rats. Maintenance of pallidal neurons containing parvalbumin in the AAV-Bcl-xL and AAVXIAP treated rats varied extensively with the AAV-Bcl-xL group having 72 ± 10 % maintenance and A B Figure 6-25 Survival of parvalbumin-positive neurons in the globus pallidus (A) Graph displaying the proportions of parvalbumin-immunopositive neurons surviving in the globus pallidus as estimated by optical fractionator stereology on four coronal sections through the globus pallidus relative to the contralateral hemisphere. (B) Plot comparing the survival of parvalbumin-positive neurons relative to the maintenance of DARPP-32 positive striatal neurons. R value represents overall Pearson correlation coefficient independent of treatment (P < 0.05). Solid line represents significant non-zero linear correlation (P < 0.05) and the dotted lines non-significant linear trends for individual treatment groups. No statistically significant differences were found between treatment groups. 184 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP AAV-XIAP rats showing only 55 ± 11% maintenance (Figure 6-25A). Overall there was a correlation between the loss of parvalbumin-positive pallidal neurons and the striatal projection neurons (Pearson r = 0.43, P < 0.05) although this correlation was only clearly seen with the AAVXIAP treated group of rats (Figure 6-25B). Contralateral Ipsilateral Parvalbumin A NeuN B DARPP-32 C Figure 6-26 Visual assessment of the globus pallidus following AAV-Bcl-xL treatment Representative images of the globus pallidus of an AAV-Bcl-xL treated brain displaying the comparison between the treated (ipsilateral) and un-treated (contralateral) hemispheres. Immunocytochemical staining of (A) parvalbumin-positive pallidal neurons, (B) NeuN-positive neurons, and (C) DARPP-32 immunostaining. Scale bar = 250µm. 185 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP Contralateral Ipsilateral Parvalbumin A NeuN B DARPP-32 C Figure 6-27 Visual assessment of the globus pallidus following AAV-XIAP treatment Representative images of the globus pallidus of an AAV-XIAP treated brain displaying the comparison between the treated (ipsilateral) and un-treated (contralateral) hemispheres. Immunocytochemical staining of (A) parvalbumin-positive pallidal neurons, (B) NeuN-positive neurons, and (C) DARPP-32 immunostaining. Scale bar = 250µm. 186 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6.6.7 Pathological correlations with functional behaviour impairments Without any clear indications from the pathological analysis of significant AAV-Bcl-xL or AAVXIAP supplied neuroprotection I analysed data collected from individual rats to directly assess correlations between the quantified basal ganglia pathology and the extent of functional motor impairment for each of the behaviour tests in the second investigation. Due to the significant maintenance of parvalbumin-expressing pallidal neurons in PBS controls compared with the large loss in AAV-Luciferase treated rats I have separately identified whether rats received PBS or AAVLuciferase in the correlations against pallidal neurons but not for striatal correlations where there was no significant difference between the groups. The PBS and AAV-Luciferase treated rats were however combined as a single control group to assess for any linear relationships as they did not perform differently as two distinct groups in any of the functional behavioural assessments. 6.6.7.1 Spontaneous Forelimb Use The proportion of exploratory behaviour undertaken by the rats with their ipsilateral forelimb showed a direct correlation with the loss of DARPP-32 positive striatal neurons, with greater cell loss following the unilateral lesioning causing increased use of the rats ipsilateral forelimb (Pearson A B Figure 6-28 Preferential forelimb usage correlation with neuron loss in the striatum and globus pallidus Plots correlating the extent of ipsilateral forelimb use bias with the relative proportion of (A) DARPP-32 and (B) parvalbumin-positive neurons remaining within the striatum and globus pallidus respectively. Data points represent individual rats’ average score over the seven post-QA trials. R values represent overall Pearson correlation coefficient for combined data from all rats. Solid lines represent significant non-zero linear correlations (P < 0.05) and the dotted lines non-significant linear trends for individual treatment groups. Comparison with controls: (A) Bcl-xL slope P = 0.26, elevation P < 0.01; XIAP slope P = 0.08, elevation P < 0.001; (B) Bcl-xL slope P < 0.05; XIAP slope P = 0.72, elevation P < 0.001. 187 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP r = -0.43, P < 0.05). However the analysis of individual linear trends for the AAV-Bcl-xL, AAVXIAP and control treated groups found that only the AAV-Luciferase / PBS treated control rats actually displayed significant correlation between ipsilateral forelimb use and quantified striatal cell loss (P < 0.05, Figure 6-28A). Preferential usage of the rats ipsilateral forelimb did not show any overall correlation with the loss of pallidal neurons (Pearson r = -0.05, P = 0.79), with only AAVBcl-xL treatment showing a weak correlation (Figure 6-28B). 6.6.7.2 Sensorimotor Neglect Development of sensorimotor neglect on the contralateral side following QA-lesioning resulted in a greater proportion of ipsilateral sugar pellet retrievals in the “corridor” task, and was significantly correlated with both DARPP-32-positive striatal cell loss (Pearson r = -0.39, P < 0.05) and the loss of parvalbumin-positive pallidal neurons (Pearson r = -0.68, P < 0.001; Figure 6-29). Individual assessment of linear trends for each treatment group showed a strong correlation for the AAV-XIAP and AAV-Luciferase / PBS control treated rats between the loss of DARPP-32 neurons and the extent of sensorimotor neglect, but not for the AAV-Bcl-xL treated rats which displayed a wide spread of preferential sugar pellet retrievals that were seemingly unrelated to the level of striatal lesioning. Separate linear comparisons with the pathological state of the globus pallidus revealed A B Figure 6-29 Sensorimotor neglect correlation with striatal and pallidal neuronal cell loss Plots correlating the percentage of ipsilateral sugar pellet retrievals made in the “corridor” task with the relative proportion of (A) DARPP-32 and (B) parvalbumin-positive neurons remaining within the striatum and globus pallidus respectively. Data points represent individual rats’ average retrieval preference over the four post-QA trials. R values represent overall Pearson correlation coefficient for combined data from all rats. Solid lines represent significant non-zero linear correlations (P < 0.05) and the dotted lines non-significant linear trends for individual treatment groups. Comparison with controls: (A) Bcl-xL slope P = 0.10, elevation P < 0.01; XIAP not-significant; (B) Bcl-xL slope P < 0.05; XIAP slope P = 0.001. 188 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP significant non-zero linear trends for each treatment group, although the correlations were weakened for the control and AAV-Bcl-xL groups by a few outliers that displayed strong ipsilateral bias despite no disruption of pallidal neurons. Significantly though, all rats that had extensive loss of pallidal neurons also displayed strong contralateral sensorimotor neglect. 6.6.7.3 Apomorphine-Induced Rotations Comparisons between the proportion of apomorphine-induced ipsilateral rotations and the extent of DARPP-32 neuronal cell loss showed no overall correlation (Pearson r = -0.22, P = 0.24; Figure 6-30A). Individual treatment group analysis however demonstrated that the AAV-XIAP treated group did show a trend towards a greater proportion of contralateral rotations when the striatal lesion is smaller, but ipsilateral rotations when there is high loss of DARPP-32 expressing striatal cells. In contrast the proportion of rats preferentially rotating in a contralateral direction showed strong correlation with the maintenance of the globus pallidus independent of treatment (Pearson r = -0.75, P < 0.001; Figure 6-30B). A B Figure 6-30 Correlation of apomorphine-induced rotational behaviour with striatal and pallidal neuronal cell loss Plots displaying the percentage of rotations in an ipsilateral direction in relation to the maintenance of (A) DARPP-32 and (B) parvalbumin-positive neurons within the striatum and globus pallidus respectively. Each data point represents the result of a single post-QA trial with three trials conducted per rat. Shaded boxes show the predominant contralateral rotational behaviour when the relative number of parvalbumin pallidal neurons is greater in the treated hemisphere and preferential ipsilateral rotations when the globus pallidus is compromised. R values represent overall Pearson correlation coefficient for combined data from all rats. Solid lines represent significant non-zero linear correlations (P < 0.05) and the dotted lines non-significant linear trends for individual treatment groups. Neither AAV-Bcl-xL nor AAV-XIAP treatment significantly altered the linear correlations observed in the control rats. 189 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP 6.7 Discussion Through the delivery of AAV vectors encoding anti-apoptotic factors to the rodent striatum I investigated the potential for increasing the expression of Bcl-xL or XIAP within the striatal neurons with a view to enhancing their resistance against excitotoxic cell death processes that contribute to HD neurodegenerative disease processes. While apoptotic processes are likely to be major contributing factors in HD neurodegeneration, and other neurodegenerative disorders, until recently there has been little published literature surrounding the use of anti-apoptotic factors as therapeutic agents (Section 1.7). A major limiting factor for direct therapeutic administration of endogenous anti-apoptotic factors is their intracellular localisation and site-of-action requiring efficient and extensive targeting of the vulnerable neurons. The AAV1/2 vectors I investigated provide an avenue to direct the continuous expression of a therapeutic protein within transduced neurons. By injecting either AAV-Bcl-xL or AAV-XIAP into the rodent striatum – inducing over-expression of Bcl-xL and XIAP respectively – and using behavioural tests to measure motor activity before and after an intrastriatal injection of QA I assessed whether these anti-apoptotic factors could provide any symptomatic or neuropathological relief from the striatal excitotoxic insult. 6.7.1 AAV-mediated Bcl-xL or XIAP expression prior to QA The chimeric AAV1/2 vectors constructed to enhance Bcl-xL or XIAP expression prior to QA administration appeared to readily transduce the striatal neurons with significant enhancement of anti-apoptotic factor expression following a single AAV-Bcl-xL injection in the first study, and the dual injections of either AAV-Bcl-xL or AAV-XIAP in the second investigation (Section 6.3). While the AAV vectors appeared capable of travelling through the striatal parenchyma transducing neurons a reasonable distance from the injection coordinates, the actual distribution of transduced striatal cells was not consistent. This irregular distribution of cells with enhanced anti-apoptotic factor expression – some parts of the striatum appearing to have the majority of neurons transduced but adjacent areas showing no visible HA-positive cells – did not appear to follow any structural pattern in the striatum, although many rats showed numerous transduced cells adjacent to white matter tracts suggesting the vectors can more readily diffuse or are transported along these tracts. Assuming the transgenic protein expression remained intracellular, any enhanced resistance against the primary excitotoxic insult would have been restricted to the transduced cells, making extensive transduction of the targeted striatal neurons of vital importance. However the presence of HA-tagged transgenic protein throughout the cerebral cortex following AAV-Bcl-xL delivery suggested that the Bcl-xL 190 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP construct lacking the C-terminal transmembrane domain may possibly have been transported away from the transduced neurons. While transportation to the cortex may possibly be advantageous to cortical neurons subjected to apoptotic cell death, the Bcl-xL protein did not appear to be transported to other striatal neurons. Bcl-xL accumulated to a much greater extent within the striatum than XIAP despite the same regulatory elements driving expression; however both Bcl-xL and XIAP were enhanced 60-70 times above detected endogenous expression levels suggesting the different concentrations maybe related to normal processing of these anti-apoptotic factors. Transduction of nigral and pallidal neurons indicated the ability for these AAV vectors to be anterogradely and retrogradely transported to transduce neurons in the target nuclei in agreement with previous reports (Kaspar et al. 2002). 6.7.1.1 Neuropathological Changes In the first anti-apoptotic factor investigation I injected the AAV-Bcl-xL vector into the striatum at a single stereotaxic site prior to delivering QA to the same site three-weeks later. While the most extensive transduction of the striatal neurons were therefore likely to be found in close proximity to the QA injection site, the level of excitotoxic stress that these neurons were exposed to would also have been significantly greater than that in the more distal reaches of the QA-lesion. As a model of excitotoxic cell death that potentially replicates pathological mechanisms which contribute to HD neurodegeneration, I felt that the less intensive excitotoxic insult as the QA diffuses away from the injection site is possibly more representative of the challenges facing striatal neurons in HD patients than the lesion core, despite still being a very acute model. With striatal cells proximal to the site of QA injection facing very severe excitotoxicity, multiple forms of cell death pathways are likely to have been initiated including necrotic mechanisms (Portera-Cailliau et al. 1995; Bordelon et al. 1999), such that a single factor is unlikely to have any discernable impact. Therefore with this hypothesis of QA-induced cell loss and the lack of any neuroprotection in the initial AAV-Bcl-xL investigation I continued investigation of anti-apoptotic factors but increased AAV-vector delivery to two-sites within the striatum slightly removed from the site of QA injection. This enhanced the proportion of neurons transduced in the “transition zone” of the striatum surrounding the central core of the QA lesion where there is also greater survival of the less vulnerable striatal interneurons (Section 3.3.4.2). The independent enhancement of Bcl-xL or XIAP expression in the striatum by AAV-mediated gene transfer was not found to confer any statistically significant increase in the resistance of transduced striatal neurons against QA-induced excitotoxic cell death, although AAV-XIAP treatment did result in a slightly higher average survival of striatal neurons (Section 6.6.1.3). The lesioned area within 191 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP the treated striatum could be clearly delineated, despite the delivery of AAV-Bcl-xL or AAV-XIAP prior to QA administration, with no quantified maintenance of striatal neurons, and no apparent evidence of transduced neurons surviving within the confines of QA lesioning. Co-labelling of transduced neurons within the unlesioned portion of the striatum with HA and DARPP-32 or krox-24 indicated that expression of transgenic Bcl-xL or XIAP was not disrupting normal cellular processes to the extent seen with the continuous Luciferase expression in which the transduced neurons lost all expression of these marker proteins (Section 6.6.3). Although there was no evidence of Bcl-xL or XIAP supplied maintenance of the striatal neurons, I assessed other neuropathological changes in the basal ganglia to allow correlations with the changes in functional behaviour. While no significant differences were observed in the substantia nigra, parvalbumin-positive neurons in the globus pallidus showed remarkably different levels of maintenance between individual rats irrespective of treatment group (Section 6.6.6). Only AAV-XIAP rats exhibited a positive correlation with the extent of striatal lesioning, suggesting pallidal cell death is not solely consequential of the loss of afferent striatal projections, but may have involved direct QA exposure or be modulated by the AAV vector transduction. Contrasting results between AAV-Luciferase and PBS injected control rats prevented assessment regarding any potential neuroprotective action of the anti-apoptotic factors on maintenance of the globus pallidus, with Luciferase potentially enhancing degeneration of pallidal neurons as discussed previously (Section 5.8.2.2). Afferent innervations of the striatum by THpositive dopaminergic neurons (Section 6.6.4) and SMI32-positive corticostriatal projections (Section 6.6.5) demonstrated the disruption of the striosome-matrical architecture of the striatum following QA lesioning, but neither AAV-Bcl-xL nor AAV-XIAP delivery provided any visibly discernable protection against the disruption of striatal structural arrangement evident eight-weeks following acute excitotoxic insult. 6.7.1.2 Behavioural Protection Despite the lack of a significant reduction in any of the investigated pathological changes induced in the striatum by QA lesioning, I did observe some attenuation of functional behaviour impairments when compared with the AAV-Luciferase / PBS treated control rats. AAV-XIAP treated rats, while tending towards having enhanced striatal maintenance, displayed complete amelioration of an ipsilateral forelimb use bias in the spontaneous exploratory forelimb use assessment. AAV-Bcl-xL treated rats in the two-site delivery investigation also had a trend towards a reduction in the preferential ipsilateral forelimb use that was not quite significant and a similar trend towards reduced contralateral sensorimotor neglect. Across all treatment groups the extent of ipsilateral forelimb use bias was correlated with the loss of striatal neurons, while the severity of sensorimotor neglect 192 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP appeared to be more dependent on the maintenance of pallidal neurons. Therefore assuming the observed trends in functional behaviour for each treatment group reflected real effects of treatment, AAV-XIAP and AAV-Bcl-xL mediated prevention of a strong ipsilateral forelimb use bias would tend to indicate enhanced maintenance of striatal neuronal function, while reduced contralateral sensorimotor neglect in AAV-Bcl-xL treated rats is suggestive of greater globus pallidus functionality. Although this is circumstantial evidence, it is possible that the anti-apoptotic factors could have helped maintain the function of surviving neurons without significantly increasing overall survival of striatal neurons. Greater variation in the apomorphine-induced rotation direction for both the AAV-Bcl-xL and AAVXIAP treated rats, compared to the predominantly ipsilateral rotations in control rats, tended to also suggest a reduced severity of striatal lesioning or compensatory changes in the dopamine system specifically modulated by the enhanced anti-apoptotic factor expression; although I am not aware of any cellular pathway through which this could have occurred. In contrast, no attenuation of apomorphine- or amphetamine-induced rotational behaviour was seen in the initial single-site AAVBcl-xL delivery investigation suggesting a significant difference in the integrity of dopamine signalling or striatal efferent connections between AAV-Bcl-xL treated rats in the two studies. Although I only conducted limited overlapping neuropathological analysis of the two AAV-Bcl-xL studies I did not see any structural changes that would provide an explanation of these differential performances in apomorphine-induced behaviour. This therefore raises the question of other differences in the design of the two investigations. Besides vector delivery the only major design change was the dietary restriction of the rats in the second investigation required for the food retrieval based “corridor” task. Dietary restriction has been shown to induce an increase in endogenous BDNF expression (Mattson 2000; Duan et al. 2003) which could potentially have had an influence on the severity of QA lesioning and also more specifically on dopamine receptor expression. Without an evident reduction in the gross QA-induced striatal pathology in the dietary restricted study, and similar inducement of ipsilateral rotations in both groups of control rats, the prevention of ipsilateral rotational behaviour appeared to be a direct effect of the enhanced Bcl-xL expression – possibly in conjunction with a small increase in endogenous neurotrophic factor support. Alternatively, with the apomorphine-induced rotation direction correlating strongly with the maintenance of the globus pallidus, it could be that the pallidal neurons were better maintained in the second investigation – either by the greater Bcl-xL expression or chance positioning of the QA lesion causing differential loss of pallidal neurons between treatment groups. Overall though the sensorimotor neglect “corridor” task – which appeared to give the most sensitive measure of 193 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP functional behaviour in this QA lesion model – was not significantly attenuated by the prior delivery of AAV-Bcl-xL or AAV-XIAP. 6.7.2 Comparison with previous studies The lack of any significant protection of striatal projection neurons by the viral vector delivery of Bcl-xL or XIAP prior to QA-induced excitotoxicity was in contrast to a number of previously reported situations in which pro-survival proteins have protected specific populations of neurons from various apoptosis promoting insults (Section 1.7.1). XIAP has also recently been reported to protect against the excitotoxic kanic acid insult of CA3 neurons following in vivo delivery of a XIAP-PTD (protein-transducing-domain) fusion protein (Li et al. 2006) and also glutamate-induced death of embryonic motor neurons and dorsal root ganglion cultures in vitro (Garrity-Moses et al. 2006). While these anti-apoptotic factors appear capable of attenuating cell death following apoptotic-inducing excitotoxic signals, a recent study of transgenic mice over-expressing Bcl-2 failed to display any reduction in QA-induced striatal cell loss (Maciel et al. 2003). These findings suggest that QA-induced cell death is not likely to be solely dependant on a single apoptotic mechanism, but may involve multiple mechanisms of cell death; including non-Bcl protein regulated mitochondrial permeability transition – commonly induced by high intracellular Ca2+ accumulation following overexcitation (Bordelon et al. 1998) – and neuronal necrosis. With conflicting data surrounding the actual process of QA-induced cell death, it is possible that QA can activate multiple pathways leading to cell death depending on experimental conditions. Therefore, while Bcl-2 / Bcl-xL may protect striatal neurons against mitochondrial dependent apoptotic mechanisms, these pathways may simply be bypassed through activation of alternative cell death mechanisms including non-caspase dependent processes. The failure of XIAP to provide any significant protection is strongly indicative of caspase-independent apoptosis or necrotic cell death pathways. While I verified that both AAVvectors produced functionally active anti-apoptotic proteins capable of preventing the induction of apoptosis by staurosporine (Chapter 2, (Kells et al. 2006)), I did not specifically test for the attenuation of QA toxicity in vitro given uncertainties over the induction of apoptotic mechanisms. 6.7.3 Conclusion The selective degeneration of specific neuronal populations in HD, despite ubiquitous expression of mutant huntingtin in the CNS, indicates the induction of cell death through mechanisms specifically related to physiological properties of vulnerable neurons. Excitotoxicity is proposed as a leading initiator of HD neurodegeneration given intensive glutamate input to the striatum, selective 194 Anti-Apoptotic Factor Delivery: AAV-Bcl-xL and AAV-XIAP vulnerability of striatal projection neurons to chemical excitotoxins, and direct potentiation of glutamate receptor signalling by mutant huntingtin. With the developing and juvenile CNS appearing remarkably resilient to the presence of mutant huntingtin, it is conceivable that sensitive apoptotic mechanisms maybe initiated in older neurons that have reduced capacity to handle potentially destructive cellular stress. Therefore while apoptotic mechanisms initiated via excessive NMDA receptor signalling may be a significant contributor to the slow progressive degeneration in the HD brain, the failure of either enhanced Bcl-xL or XIAP expression to significantly attenuate QA-induced neuronal death in the current investigations may well have been due to the acute, intensive insult simultaneously initiating multiple cell death pathways such that single factor intervention is insufficient to ultimately block neurodegeneration. Whether the same process occurs in the HD brain with cell death occurring via multiple apoptotic and necrotic mechanisms is at this stage unknown, although apoptotic hallmarks are present in post-mortem HD brains (Section 1.7). The in vivo gene delivery and overexpression of Bcl-xL or XIAP did not appear to disrupt neuronal function, but conversely showed a potential preservation of neuronal signalling given the tendency towards a reduction in the degree of behavioural impairments exhibited by the AAV-Bcl-xL and AAV-XIAP rats despite no overall enhancement of neuronal maintenance. Therefore these results suggest that while anti-apoptotic factor production may not be effective as a stand alone therapy, in conjunction with other more efficacious neuroprotective molecules anti-apoptotic factors may provide critical preservation of neuronal function in a complimentary fashion. Further investigation in a more subtle / progressive model of striatal degeneration may also reveal whether the enhancement of endogenously expressed pro-survival proteins can directly attenuate or slow the degeneration of striatal neurons. 195 General Discussion Chapter 7 General Discussion The prospect that neuroprotective agents which halt the relentless progression of neurodegenerative processes occurring in the CNS of HD patients, and other neurological disorders will one day be clinical reality was the ultimate goal driving this thesis research and other similar bodies of current neuroscience investigation. 7.1 AAV-mediated Gene Delivery Utilising AAV vectors to mediate the transfer of genetic constructs to the rodent brain, I directed the enhanced expression of a neurotrophic factor – BDNF or GDNF – or anti-apoptotic factor – Bcl-xL or XIAP – within vulnerable striatal projection neurons. While AAV vectors themselves have been shown to be non-toxic, and are currently used extensively in both pre-clinical research and clinical trials, the spread of vector particles and expression of the encoded therapeutic molecules needs to be closely analysed on a use-by-use basis. Following intrastriatal injection of the AAV1/2 vectors I observed extensive transduction of striatal neurons as well as numerous neurons in the globus pallidus, substantia nigra pars compacta and pars reticulata, and cerebral cortex (Section 4.4.2). This spread of transduction to cells residing in nuclei connected to the striatum via afferent and efferent projections indicated the ability for the AAV1/2 vector to be retrogradely, anterogradely and possibly postsynaptically transported in agreement with previous assessment of AAV vector transportation (Burger et al. 2004). While this may be potentially advantageous for many applications including neuroprotection, through the production of a therapeutic agent at both the targeted cell body and its terminal projection fields, it may also result in undesired deleterious actions. The extensive overexpression of BDNF following high-titre AAV vector delivery to the striatum in the initial investigation resulted in seizure development, aggression, weight-loss and death (Section 5.3). These deleterious effects were likely to have been induced by enhanced BDNF activity outside the striatum due to transportation of the AAV-BDNF vector particles, or alternatively the direct transportation / diffusion of the overexpressed BDNF protein, presumably to the entorhinal cortex or hippocampus (Pelleymounter et al. 1995; Naert et al. 2006). Although beyond the scope of this thesis research, future consideration should be given to assessing the impact of enhanced expression of endogenous proteins – particularly secreted neurotrophic 196 General Discussion factors – on processes of neurogenesis that occur in the subependymal layer bordering the caudateputamen and appear to be modified in both the HD brain (Curtis et al. 2003; Curtis et al. 2005), and following QA lesioning (Tattersfield et al. 2004; Gordon et al. 2007). The generation of new neurons has been shown to occur in the striatum, however the normal physiological importance of this phenomenon is not clear. Significant changes in the striatal expression of neurotrophic factors vital for directing brain development are likely to effect the migration, maturation, and / or survival of subependyma-derived neuroblasts. Although the enhancement of neurotrophic factors prior to excitotoxic insult was undertaken to primarily investigate protection against neurodegenerative processes and subsequent functional deficits, without direct assessment of proliferative cell markers the possible restoration of the striatum via neurotrophic factor mediated neurogenesis / migration cannot be discounted. With the mammalian brain potentially containing a regenerative capacity, gene delivery may provide an avenue through which to promote both neuronal restoration and enhance long-term survival of new neurons in the HD brain despite the persistence of pathogenic genetic pressures. The level of expression required to provide therapeutic intervention following gene transfer is also an area that needs to be assessed in vivo. The expression cassette I used for all of the AAV vectors was designed to generate high-level expression that was sustained throughout the entire investigation. The hybrid CBA promoter is not down-regulated with expression maintained indefinitely following AAV vector transduction (Klein et al. 2002). While the need for continuous long-term expression is presumed to be a necessity to counteract slow neurodegenerative processes, the required level of expression will be specific to the biotherapeutic molecule. Following the welfare issues with AAVBDNF treatment I reduced the expression level by diluting the vector stock prior to delivery; however this did result in a discernable reduction in the number of neurons that were transduced. While this was not of great concern for secreted neurotrophic factors, this approach is clearly not suitable for non-secreted therapeutics in which extensive transduction is paramount. A potentially better approach would be the incorporation of lower expression or regulatable promoters allowing extensive transduction of targeted cells but control of the transgene expression levels (Eslamboli et al. 2005). Surprisingly, the AAV-GDNF transduced neurons in the striatum, while being observed to have survived QA-induced cell death, displayed complete loss of striatal projection neuron markers likely to indicate severely compromised function due to the high expression of GDNF protein (Section 5.7.3). This further highlights the concern with continuous expression of a transgene product in which the host cells function maybe compromised either by the physical production of the transgenic protein or via continuous exposure. While a similar loss of neuronal proteins was observed in the AAV-Luciferase transduced neurons, the high-expression of anti-apoptotic factors 197 General Discussion did not induce the same disruption to normal protein expression, suggesting the alterations are specific to gene expression (Section 6.6.3). Significant changes to dopaminergic mechanisms in the striatum following enhanced GDNF expression have been previously quantified including increased dopamine synthesis and turnover (Hudson et al. 1995; Kirik et al. 2000b), with the indication that enhanced dopamine exacerbates neuronal cell death in the striatum (Cyr et al. 2003). While neurochemical alterations were not assessed in the current investigations, the reduction in DARPP-32 neuron expression following high expression of BDNF or GDNF indicate that biochemical signalling pathways would have been compromised. DARPP-32 has been referred to as a “molecular switch” modulating the integration of dopaminergic and glutamatergic signalling cascades through its potent inhibition of protein phosphatase-1 (Gould and Manji 2005); with the phosphorylation state of all physiological effectors in striatal dopaminoceptive neurons regulated by the DARPP-32 / protein phosphatase-1 cascade (Greengard et al. 1999). DARPP-32 knockout mice display significant reductions in their molecular, physiological and functional behaviour changes normally induced by dopamine and dopamine-mediated drugs of abuse (Greengard et al. 1999; Valjent et al. 2005). The use of regulatable promoters allowing the on / off switching of transgene expression would also provide the ability for pulsatile application of the therapeutic molecule (Jiang et al. 2004; Goverdhana et al. 2005), which could be more therapeutically beneficial than straight continuous expression that may lead to down-regulation of receptors and effector proteins. 7.2 Preventative Therapy for Huntington’s Disease To investigate whether the continuous delivery of these neurotrophic factors or anti-apoptotic proteins could enhance the resistance of striatal projection neurons, which are selectively vulnerable to cell death processes in HD, I locally injected QA into the striatum as a model of HD pathological changes (Chapter 3). QA has been extensively used to model the selective loss of striatal neurons seen in HD through over excitation of NMDA receptors expressed by striatal projection neurons. While the actual mechanism of neurodegeneration that occurs in HD has not been fully elucidated, there is reasonable evidence to suggest that cell death pathways stimulated by excitotoxic insults are involved with expanded huntingtin causing potentiation of the NR2B subunit expressed by striatal neurons (Chen et al. 1999; Zeron et al. 2001). It is also conceivable that QA itself may contribute towards the neurodegeneration (Chiarugi et al. 2001), with an increase in QA being reported in the HD brain (Guidetti et al. 2004). Therefore while QA tends to replicate the striatal pathology of HD, the direct intrastriatal injection causes an acute, intense neurotoxic insult localised to the tissue surrounding the site of injection not representative of the slow progressive HD neurodegeneration. 198 General Discussion With complete non-selective cell loss directly at the site of QA injection, neuroprotection is more likely to occur in the surrounding penumbra region where the lower QA concentrations have spared the less vulnerable interneurons. Given that the QA stimulation of excitotoxic mechanisms greatly exceeds any similar activation in the actual HD brain, I therefore conversely expect that any protection seen against QA-induced pathology would be considerably greater in the HD brain. Of the four proteins assessed as potential therapeutic agents, only enhanced BDNF expression resulted in a significant quantified protection of striatal neurons against QA-induced excitotoxicity, displaying gross maintenance of the striatal cytoarchitecture in addition to the survival of individual striatal projection neurons (Section 5.7.1). While AAV-GDNF transduced cells survived within the penumbra of the lesioned striatum, they lacked DARPP-32 and krox-24 expression suggesting a loss of neuron functionality. Neither Bcl-xL nor XIAP overexpression provided any significant protection to the transduced striatal cells – visually apparent by the absence of HA-positive cells within the lesioned striatum. However given the spread of the QA lesion – covering approximately half of the striatum – and substantial variation in the extent of neuronal cell death, it is possible that small levels of neuroprotective support may have occurred in the peripheral reaches of the lesion but were below the resolving power of these in vivo studies. Downregulation of marker proteins raises additional questions over the actual extent of neuronal cell death following QA, as against the loss of phenotypic markers. A phenomena that has previously been reported in other neurological models, such as the loss of TH expression following 6hydroxydopamine lesioning despite the persistence of dopaminergic cell bodies and nigrostriatal projections (Reis et al. 1978; Shirao et al. 1992; Sauer and Oertel 1994). The use of chemical tracers or histological markers, not biochemically linked to the GABAergic phenotype of the striatal projection neurons, may have provided more definitive assessment of the striatal neurons fate and any maintenance of axonal projections following QA. However the loss of Luciferase-expressing cells within the lesioned striatum of AAV-Luciferase treated control rats suggest that in the absence of enhanced trophic support the striatal neurons do ultimately die in this 50nmol QA lesion model, although no time-course of neurodegeneration was determined. The functional state of the transduced neurons expressing high-levels of BDNF or GDNF but with significantly down-regulated DARPP-32 and krox-24 expression remains to be determined, with the possibility that attenuation of behavioural impairments is resultant of neurotrophic factor activity on other non-transduced striatal neurons or actions outside of the striatum The very limited protection of striatal neurons solely by BDNF, despite reports of enhanced neuronal survival with GDNF and anti-apoptotic factors, brings into question the suitability / relevance of the 199 General Discussion QA lesion model for conducting neuroprotective investigations of the striatum. GDNF has been previously shown to protect striatal neurons against cell death induced by the mitochondrial toxin 3NP (McBride et al. 2003) and also in a transgenic mice model of HD (McBride et al. 2006). The failure of both Bcl-xL and XIAP to provide protection against QA neurotoxicity suggests the inducement of neurotic cell death or possibly the activation of multiple apoptotic mechanisms such that a single factor is unable to ultimately prevent degeneration. Despite the apparent severity of QA-induced striatal degeneration, promising reductions in the extent of QA lesioning have been reported by transplantation of encapsulated choroid plexus cells secreting multiple neurotrophic factors (Borlongan et al. 2004; Emerich et al. 2006), and following the combined delivery of pharmacological agents pyruvate and minocycline (Ryu et al. 2006), suggesting that delivery of multiple factors can overcome the QA-induced excitotoxic insult. With BDNF gene transfer alone providing what appeared to be significant maintenance of the gross striatal architecture but minimal enhancement of neuron survival, it would possibly have been more advantageous to have investigated co-delivery of the anti-apoptotic factor vectors looking for any additional complementary physical and functional enhancement of the BDNF-mediated protection. It is conceivable that the overexpression of BDNF may have enhanced the striatal neurons capacity to handle an increase in NMDA receptor activity, but not to the extent of blocking the activation of apoptotic mechanisms. While QA has provided a readily available model to study excitotoxicity in the striatum, and more specifically the degeneration of the vulnerable projection neurons; the severity and acuteness with which QA induces neuronal cell death maybe preventing the true realisation of therapeutic potential when used to investigate possible neuroprotective agents for HD. With the development of new transgenic HD models showing progressive degeneration of striatal projection neurons reminiscent of HD, the reliance on neurotoxic chemical based modelling of HD neurodegeneration has effectively been eliminated. Additionally, the realisations that pathological changes in the HD brain are not restricted to the caudate-putamen, but are evident in numerous regions of the brain such as the neocortex, globus pallidus, substantia nigra and thalamus (Vonsattel and DiFiglia 1998) make it important that future research into preventative therapies for HD do not neglect to consider the impact of these regions. The confirmation of BDNF as a potential biotherapeutic molecule to prevent or restrict the degeneration of striatal projection neurons following AAV vector mediated gene delivery to the striatum, and the significant amelioration of functional behavioural defects displayed in these investigations further highlights the importance of BDNF in the functional maintenance of the striatum. With the necessity of BDNF for the functional maintenance of the striatal GABAergic neurons (Nakao et al. 1995; Ventimiglia et al. 1995), the well documented reduction of BDNF levels 200 General Discussion in HD patients (Ferrer et al. 2000; Zuccato et al. 2001) and transgenic mice (Zuccato et al. 2005), and the impaired transport of BDNF by mutant huntingtin (Zuccato et al. 2001; Zuccato et al. 2003), an enhancement of striatal BDNF appears promising as a therapeutic approach. The disruption of intracellular transport by expanded huntingtin may limit the transportation of BDNF to the striatal target nuclei following the transduction of striatal neurons that I observed in these investigations; however this may also limit the spread to undesired CNS regions reducing the potential for deleterious side effects. Finally the attenuation of behavioural deficits was found to correlate with the survival of both striatal and pallidal neurons, although the loss of parvalbumin-positive GABAergic neurons in the globus pallidus showed considerable variation within each treatment group that was largely independent from the extent of striatal degeneration. A specific cause for the degeneration of pallidal neurons following intrastriatal QA injection was undetermined, but potentially involved direct QA exposure, secondary neurodegeneration, or an adverse consequence of AAV vector transduction. With considerable variation in the extent of pallidal degeneration any maintenance following overexpression of BDNF, Bcl-xL or XIAP was indeterminable. It was however interesting to observe gross alterations in functional behaviour correlations with the integrity of both the striatum and globus pallidus following striatal gene delivery. While the gross level of locomotor activity did not appear to display large changes following striatal lesioning, the unilateral QA lesion did induce lateralised imbalances in motor control with an apparent reduction in sensorimotor activity contralateral to the lesioned striatum. Attenuation of contralateral forelimb use deficits by AAVmediated therapeutic expression, without maintenance of DARPP-32 striatal neurons, resulted in a weaker correlation between preferential forelimb use and striatal integrity, with the AAV-BDNF treated rats specifically acquiring a strong correlation to the extent of pallidal cell survival, which was not exhibited by the control rats (Section 5.7.7.1). A similar pattern was observed for the amelioration of sensorimotor neglect following AAV-BDNF delivery, with a reduced correlation to the relative survival of striatal neurons, but enhanced correlation with pallidal neuronal maintenance. The degree of contralateral neglect in the “corridor” task was found to correlate with the integrity of the globus pallidus for all rats irrespective of treatment – presumably reflecting the enhanced sensory complexity of this lateralised selection task over the spontaneous forelimb use analysis. In addition to the reduced contralateral motor activity observed in these two non-drug induced behavioural analyses, the administration of apomorphine following the unilateral striatal lesioning induced rotational behaviour that was generally towards the lesioned striatum. However correlation analysis with the relative survival of pallidal neurons showed a strong relationship between the integrity of the globus pallidus and the direction of induced rotation following striatal lesioning – ipsilateral 201 General Discussion rotations induced when the pallidus was disrupted but contralateral rotations if the pallidus was fully maintained. AAV-BDNF delivery weakened the directional correlation with end-point pallidal pathology, possibly due to direct BDNF-induced modification of dopamine processing / signalling (Section 5.8.2.1) and / or enhanced maintenance of striatal projection neurons that resulted in apomorphine-induced ipsilateral rotations in initial tests following QA injections but contralateral rotations in later trials. Variation in the extent of striatal lesioning alone appeared to have no discernable influence on the direction of the apomorphine-induced rotations in these neuroprotective investigations despite previous reports of the lesion size and position influencing drug-induced rotations (Dunnett et al. 1988; Norman et al. 1992; Fricker et al. 1996; Nakao et al. 1996; Nakao and Brundin 1997). These behavioural deficit correlations with neuropathology changes highlight the importance of the globus pallidus as a major striatal output nucleus for facilitating motor control, with BDNF overexpression specifically appearing to have a much greater functional impact when the pallidal neurons are maintained. Assessment of the globus pallidus has not previously been reported in neuroprotective investigations following striatal lesioning, however lesioning of the GPe (equivalent to the rodent globus pallidus) has been proposed as a potential treatment to correct HD symptoms (Joel et al. 1998; Ayalon et al. 2004; Reiner 2004; Temel et al. 2006). The basic direct pathway / indirect pathway model of basal ganglia circuitry describes two major striatal output circuits; stimulation of the direct pathway leading to disinhibition of the thalamus via the GPi and SNr, and the indirect striatopallidal projections increasing thalamic inhibition via the GPe, subthalamic nucleus and GPi (Penney and Young 1986; Crossman 1987; Albin et al. 1995; Joel 2001). Although the direct GPi / SNr pathway is generally considered to promote intended movements by enhancing thalamocortical activity, and the indirect GPe pathway suppressing movements, they are presumed to have complimentary roles in controlling desired locomotion (Penney and Young 1986; DeLong 1990). Ayalon et al. (2004) reported that bilateral QA lesioning of the rat globus pallidus resulted in attenuation of behavioural impairments acquired following QA lesioning of the striatum and that pallidal lesions alone did not generally induce severe impairments in motor and cognitive tasks; however a slight reduction in activity was observed in some of their functional assessments as predicted by the basal ganglia circuitry model following a lesion of the GPe. The current findings that contralateral hypokinesia and sensorimotor neglect are exhibited following striatal lesioning, and are potentially compounded by a loss of pallidal neurons, appear contradictory to the earlier studies but may be resultant of different behavioural assessments or the use of unilateral lesioning compared with the earlier bilateral lesions. Lesioning of the globus pallidus is suggested as a treatment strategy for attenuating the hyperkinetic chorea symptoms of HD generally exhibited in the earlier stages of 202 General Discussion disease progression; however the implications raised in these investigations – that a loss of pallidal neurons following striatal degeneration will enhance hypokinesia – as predicted by basal ganglia circuitry – highlights the concern that pallidal lesioning may exuberate the late-stage HD akinesia / bradykinesia symptoms (Reiner 2004). With the correlations between non-drug induced behavioural impairments and the integrity of the globus pallidus being significantly enhanced in the AAV-BDNF treated rats, it is possible that the observed BDNF-mediated amelioration of motor deficits were resultant of BDNF attenuating or compensating for the loss of striatal neurons but having no direct effect on any pathological changes in the globus pallidus. Although not investigated in the current studies, the differential loss / maintenance of striatopallidal (direct pathway) and striatoentopenducular (indirect pathway) neurons, and the integrity of the entopenducular nucleus should also be considered to further elucidate the pathological basis of the behavioural deficits I observed. Lesioning of the GPi (equivalent to the rodent entopenducular nucleus; pallidotomy) is known to alleviate hypokinetic motor dysfunction in Parkinson’s disease patients through eliminating the over-inhibition of the thalamus and thereby allowing easier initiation of movement (Bergman and Deuschl 2002). Byler et al. (2006) recently demonstrated that unilateral electrolytic lesioning of the entopenducular nucleus in rats induced hyperlocomotion with a tendency for ipsilateral circling behaviour, but they also reported the development of sensorimotor asymmetries and contralateral forelimb motor deficits which they proposed were due to non-specific damage to fibres of passage within the internal capsule (Byler et al. 2006). This would suggest that hyperactive motor impairments normally induced by specific entopenducular nucleus lesioning can be modified by pathological disruption to other basal ganglia circuitry, and that pathological disruption of pallidal neurons in the direct pathway may not be evident from behavioural testing. With the primary pathology in the current investigations localised to the striatum, there exists the possibility that behavioural impairments could partially be attributed to an unequal loss / protection of striatopallidal neurons differentially projecting to the internal and external segments of the globus pallidus. However with the exception of AAV-BDNF treated rats that did not develop contralateral deficits, the lesioned striatum displayed fairly extensive loss of DARPP-32 neurons and provided no indication of differential vulnerabilities between the GABAergic projections as have been previously reported to occur following QA lesioning (Figueredo-Cardenas et al. 1994) and in early stage HD (Reiner et al. 1988; Sapp et al. 1995). A more tenable explanation for the behavioural deficit correlations with neuropathology changes maybe that the striatal lesioning disrupts processing of sensory inputs from the cortex, thereby resulting in reduced ability to initiate contralateral motor activity response to sensory input, with disruption to the globus pallidus acting to exaggerate any impairment of striatal output signalling in comparison to the intact contralateral basal ganglia. 203 General Discussion 7.3 Conclusion Overall this thesis highlights the vital importance to assess putative biotherapeutic agents using in vivo model systems and the need to consider adverse complications that can arise from nonregulatable control of transgene expression following gene delivery. Utilisation of AAV1/2 vectors to mediate gene transfer to the striatum led to efficient transduction of striatal neurons, directing highlevel expression of biologically functional proteins within the striatum prior to QA-induced lesioning. Additional transduction of neurons in CNS regions directly connected to the striatum also occurred to a lesser extent, which could potentially be of therapeutic benefit for striatal protection though enhanced trophic support within the terminal-fields, but may also induce unintended consequences such as the observed induction of seizures by BDNF. Maintenance of the striatum by BDNF, accompanied by an attenuation of the behavioural impairments exhibited following acute excitotoxic lesioning of the striatum, provided further evidence of the promising therapeutic potential that BDNF holds for preventing the progression of HD. Further investigation is required however to determine an optimal level of BDNF expression and fully elucidate the impact of continuous long-term overexpression. Enhancement of striatal GDNF did not replicate the previously reported neuroprotective support of striatal neurons, with substantial disruption to the striatal architecture and an apparent downregulation of both the intracellular dopamine and glutamate signal modulating protein DARPP-32, and the early response neuronal transcription factor krox-24. Similar to the enhanced BDNF expression, the disruption to normal physiological protein expression suggested large increases in the striatal expression of GDNF is detrimental to the functionality of dopaminoceptive striatal neurons, negating any trophic assistance GDNF may potentially provide against excitotoxic stress when available to striatal neurons at lower concentrations. Together these BDNF and GDNF investigations demonstrated the prospect neurotrophic factor based therapy holds for reducing the susceptibility of striatal neurons in neurodegenerative disease, although the imperative need for greater control over the location and quantity of endogenously expressed proteins is clearly manifested. To my knowledge this is the first study to demonstrate downregulation of DARPP-32 and krox-24 in striatal projection neurons following the overexpression of neurotrophic factors and QA lesioning. Enhanced expression of a single anti-apoptotic protein by striatal projection neurons did not convey any reduction in their susceptibility to acute excitotoxic-induced neurodegeneration. However the overexpression of either Bcl-xL or XIAP in the striatum provided attenuation against the severity of behavioural impairments, suggesting enhanced functionality of the maintained neurons with no 204 General Discussion apparent transgene induced disruption of normal host cell signalling pathways. As intracellular therapeutic agents, the high efficiency of transduction provided by the AAV1/2 vectors, without any adverse effects in these studies, is vital to ensure extensive expression of the vulnerable striatal neurons. Therefore while these investigations suggest anti-apoptotic factors in isolation may not prevent excitotoxic-induced neurodegeneration, they may well be of significant therapeutic benefit in a combined treatment strategy to prevent unintended inducement of apoptotic death and enhance the maintenance of neuronal function. This thesis investigation also demonstrates the critical involvement of basal ganglia neurons beyond the striatum in modulating sensorimotor behaviour following acute lesioning of the striatum, and the importance for preventative HD treatment strategies to also consider the impact of other primary or secondary areas of neurodegeneration on HD symptoms. Maintenance of the globus pallidus proved vital in attenuating the degree of sensorimotor deficits and reversing drug-induced effects on locomotor activity following QA lesioning. As more accurate transgenic models of HD become available and inducible promoters of transgene expression are incorporated into the gene expression cassettes, future investigation targeting in vivo delivery of neurotrophic factors and other endogenously expressed pro-survival factors to areas of mutant huntingtin induced neurodegeneration may well display considerable enhancement of the pathological and behavioural protection I observed in these investigative therapeutic studies. Ultimately the safe and efficient demonstration of chimeric AAV-vectors to deliver biotherapeutics to the striatal neurons and specifically the maintenance of striatal structure and amelioration of functional impairments following the enhancement of BDNF, known to be depleted in the HD brain, confirms the promising potential for in vivo gene therapy strategies to prevent the onset or slow progression of neurodegeneration. 205 Appendix A: Gene Sequences and DNA Plasmids Appendix A: Gene Sequences and DNA Plasmids A.1 cDNA Gene Sequences Combined sequences from pGEM-T Easy cloning vectors and reverse sequencing of AAV-backbone vectors across the C-terminal HA-tag. Initial ATG (methionine) start codon and the TGA TAG TAA stop codons are highlighted. Restriction sites flanking the cDNA sequences are underlined. HA-tag sequence: TAT CCG TAT GAT GTT CCT GAT TAT GCT A.1.1 Brain Derived Neurotrophic Factor BDNF cDNA sequence PCR cloned out of pAM/CRE-BDNF-WPRE-bGHpA (M. During and D. Young, Department of Molecular Medicine and Pathology, The University of Auckland) BLAST alignment with: Homo sapiens brain-derived neurotrophic factor (BDNF), transcript variant 1, mRNA. Accession Number: NM_170735.4 Complete match with bases 563..1303 Signal peptide 563..616 Pro-protein 617..1303 Mature peptide 947..1303 Protein Identity with Rattus norvegicus: 96% (positives 97%) CTC GAG CAC CAG GTG AGA AGA GTG ATG ACC ATC CTT TTC CTT ACT ATG GTT ATT TCA TAC TTT GGT TGC ATG AAG GCT GCC CCC ATG AAA GAA GCA AAC ATC CGA GGA CAA GGT GGC TTG GCC TAC CCA GGT GTG CGG ACC CAT GGG ACT CTG GAG AGC GTG AAT GGG CCC AAG GCA GGT TCA AGA GGC TTG ACA TCA TTG GCT GAC ACT TTC GAA CAC GTG ATA GAA GAG CTG TTG GAT GAG GAC CAG AAA GTT CGG CCC AAT GAA GAA AAC AAT AAG GAC GCA GAC TTG TAC ACG TCC AGG GTG ATG CTC AGT AGT CAA GTG CCT TTG GAG CCT CCT CTT CTC TTT CTG CTG GAG GAA TAC AAA AAT TAC CTA GAT GCT GCA AAC ATG TCC ATG AGG GTC CGG CGC CAC TCT GAC CCT GCC CGC CGA GGG GAG CTG AGC GTG TGT GAC AGT ATT AGT GAG TGG GTA ACG GCG GCA GAC AAA AAG ACT GCA GTG GAC ATG TCG GGC GGG ACG GTC ACA GTC CTT GAA AAG GTC CCT GTA TCA AAA GGC CAA CTG AAG CAA TAC TTC TAC GAG ACC AAG TGC AAT CCC ATG GGT TAC ACA AAA GAA GGC TGC 206 Appendix A: Gene Sequences and DNA Plasmids AGG GGC ATA GAC AAA AGG CAT TGG AAC TCC CAG TGC CGA ACT ACC CAG TCG TAC GTG CGG GCC CTT ACC ATG GAT AGC AAA AAG AGA ATT GGC TGG CGA TTC ATA AGG ATA GAC ACT TCT TGT GTA TGT ACA TTG ACC ATT AAA AGG GGA AGA AGA TCT TAT CCG TAT GAT GTT CCT GAT TAT GCT TGA TAG TAA A.1.2 Glial cell-line Derived Neurotrophic Factor GDNF cDNA sequence PCR cloned out of AAV/NSE-GDNF-WPRE-bGHpA (M. During and D. Young, Department of Molecular Medicine and Pathology, The University of Auckland) BLAST alignment with: Rattus norvegicus glial cell-line derived neurotrophic factor (GDNF), mRNA. Accession Number: NM_019139.1 Complete match with bases 50..682 Signal peptide 50..106 Mature peptide 344..682 Protein Identity with Homo sapiens: 92% (positives 95%) C TCG AGC GGA CGG GAC TCT AAG ATG AAG TTA TGG GAT GTC GTG GCT GTC TGC CTG GTG TTG CTC CAC ACC GCG TCT GCC TTC CCG CTG CCC GCC GGT AAG AGG CTT CTC GAA GCG CCC GCC GAA GAC CAC TCC CTC GGC CAC CGC CGC GTG CCC TTC GCG CTG ACC AGT GAC TCC AAT ATG CCC GAA GAT TAT CCT GAC CAG TTT GAT GAC GTC ATG GAT TTT ATT CAA GCC ACC ATC AAA AGA CTG AAA AGG TCA CCA GAT AAA CAA GCG GCG GCA CTT CCT CGA AGA GAG AGG AAC CGG CAA GCT GCA GCT GCC AGC CCA GAG AAT TCC AGA GGG AAA GGT CGC AGA GGC CAG AGG GGC AAA AAT CGG GGG TGC GTC TTA ACT GCA ATA CAC TTA AAT GTC ACT GAC TTG GGT TTG GGC TAC GAA ACC AAG GAG GAA CTG ATC TTT CGA TAT TGT AGC GGT TCC TGT GAA GCG GCC GAG ACA ATG TAC GAC AAA ATA CTA AAA AAT CTG TCT CGA AGT AGA AGG CTA ACA AGT GAC AAG GTA GGC CAG GCA TGT TGC AGG CCG GTC GCC TTC GAC GAC GAC CTG TCG TTT TTA GAC GAC AGC CTG GTT TAC CAT ATC CTA AGA AAG CAT TCC GCT AAA CGG TGT GGA TGT ATC AGA TCT TAT CCG TAT GAT GTT CCT GAT TAT GCT TGA TAG TAA 207 Appendix A: Gene Sequences and DNA Plasmids A.1.3 Bcl-xL Bcl-xL cDNA PCR cloned out of pcDNA3-HA-Bcl-xL (Science Reagents) BLAST alignment with: Homo sapiens BCL2-like 1 (BCL2L1), nuclear gene encoding mitochondrial protein, transcript variant 1, mRNA. Accession Number: NM_138578.1 Complete match with bases 367..1002 Gene 367-1068 Protein Identity with Rattus norvegicus: 97% (positives 98%) CT CGA GTT ATA AAA ATG TCT CAG AGC AAC CGG GAG CTG GTG GTT GAC TTT CTC TCC TAC AAG CTT TCC CAG AAA GGA TAC AGC TGG AGT CAG TTT AGT GAT GTG GAA GAG AAC AGG ACT GAG GCC CCA GAA GGG ACT GAA TCG GAG ATG GAG ACC CCC AGT GCC ATC AAT GGC AAC CCA TCC TGG CAC CTG GCA GAC AGC CCC GCG GTG AAT GGA GCC ACT GGC CAC AGC AGC AGT TTG GAT GCC CGG GAG GTG ATC CCC ATG GCA GCA GTA AAG CAA GCG CTG AGG GAG GCA GGC GAC GAG TTT GAA CTG CGG TAC CGG CGG GCA TTC AGT GAC CTG ACA TCC CAG CTC CAC ATC ACC CCA GGG ACA GCA TAT CAG AGC TTT GAA CAG GTA GTG AAT GAA CTC TTC CGG GAT GGG GTA AAC TGG GGT CGC ATT GTG GCC TTT TTC TCC TTC GGC GGG GCA CTG TGC GTG GAA AGC GTA GAC AAG GAG ATG CAG GTA TTG GTG AGT CGG ATC GCA GCT TGG ATG GCC ACT TAC CTG AAT GAC CAC CTA GAG CCT TGG ATC CAG GAG AAC GGC GGC TGG GAT ACT TTT GTG GAA CTC TAT GGG AAC AAT GCA GCA GCC GAG AGC CGA AAG GGC CAG GAA CGC TTC AAC CGC AGA TCT TAT CCG TAT GAT GTT CCT GAT TAT GCT TGA TAG TAA A.1.4 X-linked Inhibitor of Apoptosis XIAP cDNA PCR cloned out of pCI-neo-Flag-XIAP (Science Reagents) Homo sapiens Baculoviral IAP repeat-containing 4 (BIRC4), mRNA. Accession Number: NM_001167 Complete match with bases 129..1619 Gene 129..1619 Protein Identity with Rattus norvegicus: 89% (positives 95%) C TCG AGA ATG ACT TTT AAC AGT TTT GAA GGA TCT AAA ACT TGT GTA CCT GCA GAC ATC AAT AAG GAA GAA GAA TTT GTA GAA GAG TTT AAT AGA TTA AAA 208 Appendix A: Gene Sequences and DNA Plasmids ACT TTT GCT AAT TTT CCA AGT GGT AGT CCT GTT TCA GCA TCA ACA CTG GCA CGA GCA GGG TTT CTT TAT ACT GGT GAA GGA GAT ACC GTG CGG TGC TTT AGT TGT CAT GCA GCT GTA GAT AGG TGG CAA TAT GGA GAC TCA GCA GTT GGA AGA CAC AGG AAA GTA TCC CCA AAT TGC AGA TTT ATC AAC GGC TTT TAT CTT GAA AAT AGT GCC ACG CAG TCT ACA AAT TCT GGT ATC CAG AAT GGT CAG TAC AAA GTT GAA AAC TAT CTG GGA AGC AGA GAT CAT TTT GCC TTA GAC AGG CCA TCT GAG ACA CAT GCA GAC TAT CTT TTG AGA ACT GGG CAG GTT GTA GAT ATA TCA GAC ACC ATA TAC CCG AGG AAC CCT GCC ATG TAT AGT GAA GAA GCT AGA TTA AAG TCC TTT CAG AAC TGG CCA GAC TAT GCT CAC CTA ACC CCA AGA GAG TTA GCA AGT GCT GGA CTC TAC TAC ACA GGT ATT GGT GAC CAA GTG CAG TGC TTT TGT TGT GGT GGA AAA CTG AAA AAT TGG GAA CCT TGT GAT CGT GCC TGG TCA GAA CAC AGG CGA CAC TTT CCT AAT TGC TTC TTT GTT TTG GGC CGG AAT CTT AAT ATT CGA AGT GAA TCT GAT GCT GTG AGT TCT GAT AGG AAT TTC CCA AAT TCA ACA AAT CTT CCA AGA AAT CCA TCC ATG GCA GAT TAT GAA GCA CGG ATC TTT ACT TTT GGG ACA TGG ATA TAC TCA GTT AAC AAG GAG CAG CTT GCA AGA GCT GGA TTT TAT GCT TTA GGT GAA GGT GAT AAA GTA AAG TGC TTT CAC TGT GGA GGA GGG CTA ACT GAT TGG AAG CCC AGT GAA GAC CCT TGG GAA CAA CAT GCT AAA TGG TAT CCA GGG TGC AAA TAT CTG TTA GAA CAG AAG GGA CAA GAA TAT ATA AAC AAT ATT CAT TTA ACT CAT TCA CTT GAG GAG TGT CTG GTA AGA ACT ACT GAG AAA ACA CCA TCA CTA ACT AGA AGA ATT GAT GAT ACC ATC TTC CAA AAT CCT ATG GTA CAA GAA GCT ATA CGA ATG GGG TTC AGT TTC AAG GAC ATT AAG AAA ATA ATG GAG GAA AAA ATT CAG ATA TCT GGG AGC AAC TAT AAA TCA CTT GAG GTT CTG GTT GCA GAT CTA GTG AAT GCT CAG AAA GAC AGT ATG CAA GAT GAG TCA AGT CAG ACT TCA TTA CAG AAA GAG ATT AGT ACT GAA GAG CAG CTA AGG CGC CTG CAA GAG GAG AAG CTT TGC AAA ATC TGT ATG GAT AGA AAT ATT GCT ATC GTT TTT GTT CCT TGT GGA CAT CTA GTC ACT TGT AAA CAA TGT GCT GAA GCA GTT GAC AAG TGT CCC ATG TGC TAC ACA GTC ATT ACT TTC AAG CAA AAA ATT TTT ATG TCT CGC GAA TTC GGA AGA TCT TAT CCG TAT GAT GTT CCT GAT TAT GCT TGA TAG TAA 209 Appendix A: Gene Sequences and DNA Plasmids A.2 DNA Plasmid Maps A.2.1 AAV Backbone Plasmid ITR Nco I (434) CAG promoter XbaI (1026) BamHI (1127) Xho I (1134) Amp pAM/CAG-C-termHA-WPRE-bGHpA 5411 bp Eco RI (1163) BglII (1172) HA HindIII (1212) ClaI (1219) WPRE ClaI (1814) pUC19 ori bGH pA ITR Nco I (2393) SV40 ori Replacement of the insert DNA stop-codon with either a EcoRI or BglII restriction site allows inframe C-terminal fusion with the nine amino acid HA epitope tag. HA-tag sequence with 5’ EcoRI (GAATTC) and BglII (AGATCT) restriction sites and triplicate stop codons (TGA TAG TAA): GAA TTC GGA AGA TCT TAT CCG TAT GAT GTT CCT GAT TAT GCT TGA TAG TAA 210 Appendix A: Gene Sequences and DNA Plasmids A.2.2 AAV-Luciferase ITR NcoI (600) CBA promoter XbaI (1186) Amp BamHI (1293) XhoI (1298) EcoRI (1303) pAM/CBA-Luc-WPRE-bGHpA HindIII (1324) 7336 bp NcoI (1356) pUC19 ori Luciferase XbaI (3023) SV40 ori NcoI (4253) NotI (3042) HindIII (3049) ITR bGH pA WPRE AAV-expression cassette containing a Firefly Luciferase cDNA sequence supplied by M. During and D. Young 211 Appendix A: Gene Sequences and DNA Plasmids A.2.3 A.2.3.1 AAV Helper Plasmids AAV rep and cap genes XbaI (1) AAV-2 rep Restriction digest with XbaI 2 bands: - 7505bp - 3867bp pRV1 11372 bp AAV-2 cap XbaI (7505) pRV1: Diagrammatic representation only – restriction sites are only representative of fragment lengths when digested with XbaI, actual location within plasmid was unknown. Sac I (38) Sac I (578) Restriction digest with SacI 3 bands: - 3953bp - 2837bp - 540bp pH21 7320 bp Sac I (4531) AAV-1 cap 212 AAV-2 rep Appendix A: Gene Sequences and DNA Plasmids A.2.3.2 Adenoviral packaging genes HindIII (519) HindIII (14420) EcoRI (550) E4 EcoRI (2888) HindIII (2900) pF_ 6 pF∆6 HindIII (11483) VA 15423 bp Fiber pVIII E2A DNA binding protein EcoRI (9475) 33K HindIII (8472) 100kD hexon assembly protein Restriction digest with HindIII 5 bands: - 5572bp - 3011bp - 2937bp - 2381bp - 1522bp Or with EcoRI 3 bands: - 6587bp - 6490bp - 2338bp 213 Appendix B: General Materials Appendix B: General Materials & Protocols B.1 Molecular Biology Luria-Bertani broth (LB) 20 g/L LB broth, ready-made powder (USB) containing casein peptone, yeast extract and NaCl. LB amp Agar plates 20 g/L LB broth, ready-made powder (USB) 25 g/L Agar (USB) Autoclave sterilised Added 50 mg/L ampicillin poured 25 mL/plate Ampicillin (amp) 50 mg/mL Ampicillin, sodium salt (Sigma-Aldrich) in sterile dH2O added to LB or LB agar at 1:1000 (50 µg/mL) X-gal 20 mg/mL X-gal (Sigma-Aldrich) in dimethysulfoxide (DMSO; Sigma-Aldrich) spread 40µL onto pre-poured LB amp agar plates and dried. 1% Agarose Gel Electrophoresis 250 mg SeaKem®LE Agarose (BioWhittaker Molecular Applications) was added to 25 mL 1× TAE buffer and microwaved to melt agarose. 1µL ethidium bromide (Invitrogen) added to agarose and poured into a gel casting tray to set. DNA samples with 1× gel loading dye were transferred to wells in the gel alongside 5-10 µL 1kb+ DNA ladder (0.1µg/µL; Invitrogen) and gel electrophoresis run at 100 volts. DNA bands were visualised under short wave UV light using a Gel Documentation System. TAE buffer (50× stock) Tris base EDTA Glacial Acetic acid pH adjusted to 8.5 2M 100 mM 1M Tris EDTA (TE) Buffer Tris-HCl Na2EDTA autoclaved sterilised (pH 7.5) (pH 8.0) Gel Loading Dye (10×) Bromophenol Blue Xylene Cyanol Sucrose 10 mM 1 mM 0.25% w/v 0.25% w/v 40.0% w/v 214 Appendix B: General Materials B.2 Cell Culture B.2.1 Tissue culture media Culture Media / Reagent Supplier Cat. No. Dulbecco’s Modified Eagle Media (DMEM) GIBCO® 12100 Iscove’s Modified Dulbecco’s Media (IMDM) GIBCO® 12200 Leibovitz’s L-15 GIBCO® 11415 Neurobasal™ Medium GIBCO® 21103 100× L-glutamine GIBCO® 25030 100× Sodium pyruvate GIBCO® 11360 100× Penicillin-Streptomycin GIBCO® 15140 100× Non-essential amino acids GIBCO® 11140 B27 GIBCO® 17504 Fetal Bovine Serum (FBS) GIBCO® 10091 Dulbecco’s Modified Eagle Media (DMEM) 3.7g Sodium hydrogen carbonate pH 7.1 with 10N NaOH Added: Non-essential amino acids, Sodium pyruvate, Penicillin-Streptomycin Iscove’s Modified Dulbecco’s Media (IMDM) 3.0g Sodium hydrogen carbonate Leibovitz’s L15 Added: Penicillin-Streptomycin, D-glucose (3 g/L) Neurobasal™ Medium Added: L-glutamine, Penicillin-Streptomycin, D-glucose (3 g/L) B.2.2 Cell counting Hemocytometer Cell Counting 85µL serum-free media 10µL Trypan blue 5µL Cell suspension The trypan blue / cell mixture was transferred to a hemocytometer and viable cells in the four corner squares counted under a Nikon TE200 inverted scope. cell count 4 × 104 × 20 = Cells / mL 215 Appendix B: General Materials B.3 AAV Vector Genomic Titering Plasmid Standard Calculation 50µg/mL DNA of 1kb has 4.74 x 1013 molecules per mL 4.74 × 1013 × plasmid size (kb) plasmid conc. ( µg /mL) 5 × 10 4 Log template copies = Ct value − = DNA copies / µL y intercept slope Genome copies / mL = 10 Log template copies × dilution factor × B.4 Immunocytochemistry Buffers 10× Phosphate Buffered Saline (PBS) KH2PO4 18 mM 82 mM Na2HPO4 NaCl 1.4 M KCl 27 mM PBS-Triton 1× PBS plus 0.2% Triton X-100 0.4M PB NaH2PO4 Na2HPO4 pH adjusted to 7.2 76 mM 324 mM 0.1M Phosphate Buffer NaH2PO4 Na2HPO4 pH adjusted to 7.2 28 mM 72 mM Cyroprotectant Solution Phosphate buffer Sucrose Ethylene glycol 0.5 M 30 % w/v 30 % v/v 4% Paraformaldehyde Phosphate buffer 0.1 M Paraformaldehyde 4 % w/v heated to 60°C to dissolve pH adjusted to 7.4 216 1000 × 2 template volume used ( µL) Appendix B: General Materials DAB 3,3’-diaminobenzidine tetrahydrochloride (DAB) 0.5 mg/mL Phosphate buffer 0.1 M Hydrogen peroxide 0.01 % v/v Nickel sulphide 0.04 % w/v (optional) B.5 Antibodies Primary Antibody Host species Dilution Company Cat # NeuN mouse 1:1000 Chemicon MAB 377 Calbindin mouse 1:5000 Swant 300 DARPP-32 rabbit 1:750 Chemicon AB1656 Parvalbumin mouse 1:5000 Swant 235 NOS sheep 1:10000 Piers Emson - ChAT goat 1:500 Chemicon AB144P Calretinin rabbit 1:5000 Swant 7699/4 HA.11 mouse 1:1000 Covance MMS-101R Luciferase goat 1:10000 Chemicon AB3256 Tyrosine Hydroxylase rabbit 1:500 Chemicon AB152 Krox-24 (Egr-1 (C-19)) rabbit 1:1000 Santa Cruz Sc-189 GFAP rabbit 1:500 Dako Z0334 SMI32 mouse 1:10000 Sternberger Monoclonals SMI-32R Biotinlyated Secondary Antibody Host species Blocking Serum Company Cat # Anti-mouse Goat Goat Sigma-Aldrich B7264 Anti-rabbit Goat Goat Sigma-Aldrich B7389 Anti-sheep Donkey Goat Sigma-Aldrich B7390 Anti-goat Rabbit - Sigma-Aldrich B7014 Alexa Fluor® Secondary Antibody Host species Blocking Serum Company Cat # 488 anti-goat Donkey Donkey Invitrogen A11055 488 anti-rabbit Goat Donkey Invitrogen A11034 594 anti-mouse Goat Donkey Invitrogen A11032 594 anti-goat Donkey Donkey Invitrogen A11058 217 Literature References Literature References Abrous, D.N., Torres, E.M. and Dunnett, S.B. (1993). Dopaminergic grafts implanted into the neonatal or adult striatum: comparative effects on rotation and paw reaching deficits induced by subsequent unilateral nigrostriatal lesions in adulthood. Neuroscience 54(3): 657-68. Adams, J.M. (2003). Ways of dying: multiple pathways to apoptosis. Genes & Development 17(20): 2481-95. Alberch, J., Perez-Navarro, E. and Canals, J.M. (2004). Neurotrophic factors in Huntington's disease. Progress in Brain Research 146: 195-229. Albin, R.L., Young, A.B. and Penney, J.B. (1995). The functional anatomy of disorders of the basal ganglia. Trends in Neurosciences 18(2): 63-4. Alexander, G.E., DeLong, M.R. and Strick, P.L. (1986). Parallel organization of functionally segregated circuits linking basal ganglia and cortex. Annual Review of Neuroscience 9: 35781. Alexi, T., Venero, J.L. and Hefti, F. (1997). Protective effects of neurotrophin-4/5 and transforming growth factor-alpha on striatal neuronal phenotypic degeneration after excitotoxic lesioning with quinolinic acid. Neuroscience 78(1): 73-86. Alexi, T., Hughes, P.E., Faull, R.L. and Williams, C.E. (1998). 3-Nitropropionic acid's lethal triplet: cooperative pathways of neurodegeneration. Neuroreport 9(11): R57-64. Alexi, T., Hughes, P.E., van Roon-Mom, W.M., Faull, R.L., Williams, C.E., Clark, R.G. and Gluckman, P.D. (1999). The IGF-I amino-terminal tripeptide glycine-proline-glutamate (GPE) is neuroprotective to striatum in the quinolinic acid lesion animal model of Huntington's disease. Experimental Neurology 159(1): 84-97. Alexi, T., Borlongan, C.V., Faull, R.L., Williams, C.E., Clark, R.G., Gluckman, P.D. and Hughes, P.E. (2000). Neuroprotective strategies for basal ganglia degeneration: Parkinson's and Huntington's diseases. Progress in Neurobiology 60(5): 409-70. Allendoerfer, K.L., Cabelli, R.J., Escandon, E., Kaplan, D.R., Nikolics, K. and Shatz, C.J. (1994). Regulation of neurotrophin receptors during the maturation of the mammalian visual system. Journal of Neuroscience 14(3 Pt 2): 1795-811. Almeida, R.D., Manadas, B.J., Melo, C.V., Gomes, J.R., Mendes, C.S., Graos, M.M., Carvalho, R.F., Carvalho, A.P. and Duarte, C.B. (2005). Neuroprotection by BDNF against glutamate-induced apoptotic cell death is mediated by ERK and PI3-kinase pathways. Cell Death and Differentiation 12(10): 1329-43. Altar, C.A., Armanini, M., Dugich-Djordjevic, M., Bennett, G.L., Williams, R., Feinglass, S., Anicetti, V., Sinicropi, D. and Bakhit, C. (1992a). Recovery of cholinergic phenotype in the injured rat neostriatum: roles for endogenous and exogenous nerve growth factor. Journal of Neurochemistry 59(6): 2167-77. Altar, C.A., Boylan, C.B., Jackson, C., Hershenson, S., Miller, J., Wiegand, S.J., Lindsay, R.M. and Hyman, C. (1992b). Brain-derived neurotrophic factor augments rotational behavior and nigrostriatal dopamine turnover in vivo. Proceedings of the National Academy of Sciences of the United States of America 89(23): 11347-51. Altar, C.A., Boylan, C.B., Fritsche, M., Jackson, C., Hyman, C. and Lindsay, R.M. (1994). The neurotrophins NT-4/5 and BDNF augment serotonin, dopamine, and GABAergic systems 218 Literature References during behaviorally effective infusions to the substantia nigra. Experimental Neurology 130(1): 31-40. Altar, C.A., Cai, N., Bliven, T., Juhasz, M., Conner, J.M., Acheson, A.L., Lindsay, R.M. and Wiegand, S.J. (1997). Anterograde transport of brain-derived neurotrophic factor and its role in the brain. Nature 389(6653): 856-60. Altar, C.A. (1999). Neurotrophins and depression. Trends in Pharmacological Sciences 20(2): 5961. Altman, J. and Bayer, S.A. (1981). Development of the brain stem in the rat. V. Thymidineradiographic study of the time of origin of neurons in the midbrain tegmentum. Journal of Comparative Neurology 198(4): 677-716. Anderson, K.D., Panayotatos, N., Corcoran, T.L., Lindsay, R.M. and Wiegand, S.J. (1996). Ciliary neurotrophic factor protects striatal output neurons in an animal model of Huntington's disease. Proceedings of the National Academy of Sciences of the United States of America 93: 7346-7351. Aoki, C., Wu, K., Elste, A., Len, G., Lin, S., McAuliffe, G. and Black, I.B. (2000). Localization of brain-derived neurotrophic factor and TrkB receptors to postsynaptic densities of adult rat cerebral cortex. Journal of Neuroscience Research 59(3): 454-63. Araujo, D.M. and Hilt, D.C. (1997). Glial cell line-derived neurotrophic factor attenuates the excitotoxin-induced behavioral and neurochemical deficits in a rodent model of Huntington's disease. Neuroscience 81(4): 1099-110. Araujo, D.M. and Hilt, D.C. (1998). Glial cell line-derived neurotrophic factor attenuates the locomotor hypofunction and striatonigral neurochemical deficits induced by chronic systemic administration of the mitochondrial toxin 3-nitropropionic acid. Neuroscience 82(1): 117-27. Arenas, E., Perez-Navarro, E., Alberch, J. and Marsal, J. (1993). Selective resistance of tachykininresponsive cholinergic neurons in the quinolinic acid lesioned neostriatum. Brain Research 603(2): 317-20. Armanini, M.P., McMahon, S.B., Sutherland, J., Shelton, D.L. and Phillips, H.S. (1995). Truncated and catalytic isoforms of trkB are co-expressed in neurons of rat and mouse CNS. European Journal of Neuroscience 7(6): 1403-9. Arnt, J. and Hyttel, J. (1985). Differential involvement of dopamine D-1 and D-2 receptors in the circling behaviour induced by apomorphine, SK & F 38393, pergolide and LY 171555 in 6hydroxydopamine-lesioned rats. Psychopharmacology 85(3): 346-52. Arvidsson, A., Kirik, D., Lundberg, C., Mandel, R.J., Andsberg, G., Kokaia, Z. and Lindvall, O. (2003). Elevated GDNF levels following viral vector-mediated gene transfer can increase neuronal death after stroke in rats. Neurobiology of Disease 14(3): 542-56. Ashkenazi, A. and Dixit, V.M. (1998). Death receptors: signaling and modulation. Science 281(5381): 1305-8. Ayalon, L., Doron, R., Weiner, I. and Joel, D. (2004). Amelioration of behavioral deficits in a rat model of Huntington's disease by an excitotoxic lesion to the globus pallidus. Experimental Neurology 186(1): 46-58. Azzouz, M., Hottinger, A., Paterna, J.C., Zurn, A.D., Aebischer, P. and Bueler, H. (2000). Increased motoneuron survival and improved neuromuscular function in transgenic ALS mice after intraspinal injection of an adeno-associated virus encoding Bcl-2. Human Molecular Genetics 9(5): 803-11. 219 Literature References Bamji, S.X., Majdan, M., Pozniak, C.D., Belliveau, D.J., Aloyz, R., Kohn, J., Causing, C.G. and Miller, F.D. (1998). The p75 neurotrophin receptor mediates neuronal apoptosis and is essential for naturally occurring sympathetic neuron death. Journal of Cell Biology 140(4): 911-23. Baquet, Z.C., Gorski, J.A. and Jones, K.R. (2004). Early striatal dendrite deficits followed by neuron loss with advanced age in the absence of anterograde cortical brain-derived neurotrophic factor. Journal of Neuroscience 24(17): 4250-8. Barbacid, M. (1994). The Trk family of neurotrophin receptors. Journal of Neurobiology 25(11): 1386-403. Barde, Y.A. (1989). Trophic factors and neuronal survival. Neuron 2(6): 1525-34. Barinaga, M. (1994). Neurotrophic factors enter the clinic. Science 264(5160): 772-4. Barker, R.A. and Johnson, A. (1995). Nigral and Striatal Neurons. Neural Cell Culture, A Practical Approach. J. Cohen and G. P. Wilkin: 25-39. Bates, G.P. and Hockly, E. (2003). Experimental therapeutics in Huntington's disease: are models useful for therapeutic trials? Current Opinion in Neurology 16(4): 465-70. Bazzett, T.J., Becker, J.B., Kaatz, K.W. and Albin, R.L. (1993). Chronic intrastriatal dialytic administration of quinolinic acid produces selective neural degeneration. Experimental Neurology 120(2): 177-85. Bazzett, T.J., Becker, J.B., Falik, R.C. and Albin, R.L. (1994). Chronic intrastriatal quinolinic acid produces reversible changes in perikaryal calbindin and parvalbumin immunoreactivity. Neuroscience 60(4): 837-41. Beal, M.F., Kowall, N.W., Ellison, D.W., Mazurek, M.F., Swartz, K.J. and Martin, J.B. (1986). Replication of the neurochemical characteristics of Huntington's disease by quinolinic acid. Nature 321(6066): 168-71. Beal, M.F., Kowall, N.W., Swartz, K.J., Ferrante, R.J. and Martin, J.B. (1989). Differential sparing of somatostatin-neuroprptide Y and cholinergic neurons following striatal excitotoxin lesions. Synapse 3: 38-47. Beal, M.F., Ferrante, R.J., Swartz, K.J. and Kowall, N.W. (1991). Chronic quinolinic acid lesions in rats closely resemble Huntington's disease. Journal of Neuroscience 11(6): 1649-59. Beal, M.F. (1992a). Does impairment of energy metabolism result in excitotoxic neuronal death in neurodegenerative illnesses? Annals of Neurology 31(2): 119-30. Beal, M.F. (1992b). Role of excitotoxicity in human neurological disease. Current Opinion in Neurobiology 2(5): 657-62. Bemelmans, A.P., Horellou, P., Pradier, L., Brunet, I., Colin, P. and Mallet, J. (1999). Brain-derived neurotrophic factor-mediated protection of striatal neurons in an excitotoxic rat model of Huntington's disease, as demonstrated by adenoviral gene transfer. Human Gene Therapy 10(18): 2987-97. Benraiss, A., Chmielnicki, E., Lerner, K., Roh, D. and Goldman, S.A. (2001). Adenoviral brainderived neurotrophic factor induces both neostriatal and olfactory neuronal recruitment from endogenous progenitor cells in the adult forebrain. Journal of Neuroscience 21(17): 6718-31. Bergman, H. and Deuschl, G. (2002). Pathophysiology of Parkinson's disease: from clinical neurology to basic neuroscience and back. Movement Disorders 17 Suppl 3: S28-40. 220 Literature References Bibb, J.A., Yan, Z., Svenningsson, P., Snyder, G.L., Pieribone, V.A., Horiuchi, A., Nairn, A.C., Messer, A. and Greengard, P. (2000). Severe deficiencies in dopamine signaling in presymptomatic Huntington's disease mice. Proceedings of the National Academy of Sciences of the United States of America 97(12): 6809-14. Biffo, S., Offenhauser, N., Carter, B.D. and Barde, Y.A. (1995). Selective binding and internalisation by truncated receptors restrict the availability of BDNF during development. Development - Supplement 121(8): 2461-70. Bjorklund, A., Kirik, D., Rosenblad, C., Georgievska, B., Lundberg, C. and Mandel, R.J. (2000). Towards a neuroprotective gene therapy for Parkinson's disease: use of adenovirus, AAV and lentivirus vectors for gene transfer of GDNF to the nigrostriatal system in the rat Parkinson model. Brain Research 886(1-2): 82-98. Blomer, U., Kafri, T., Randolph-Moore, L., Verma, I.M. and Gage, F.H. (1998). Bcl-xL protects adult septal cholinergic neurons from axotomized cell death. Proceedings of the National Academy of Sciences of the United States of America 95(5): 2603-8. Bohn, M.C. (1999). A commentary on glial cell line-derived neurotrophic factor (GDNF). From a glial secreted molecule to gene therapy. Biochemical Pharmacology 57(2): 135-42. Bohn, M.C., Kozlowski, D.A. and Connor, B. (2000a). Glial cell line-derived neurotrophic factor (GDNF) as a defensive molecule for neurodegenerative disease: a tribute to the studies of antonia vernadakis on neuronal-glial interactions. International Journal of Developmental Neuroscience 18(7): 679-84. Bohn, M.C., Connor, B., Kozlowski, D.A. and Mohajeri, M.H. (2000b). Gene transfer for neuroprotection in animal models of Parkinson's disease and amyotrophic lateral sclerosis. Novartis Foundation Symposium 231: 70-89; discussion 89-93. Bolton, M.M., Lo, D.C. and Sherwood, N.T. (2000). Long-term regulation of excitatory and inhibitory synaptic transmission in hippocampal cultures by brain-derived neurotrophic factor. Progress in Brain Research 128: 203-18. Bonelli, R.M., Wenning, G.K. and Kapfhammer, H.P. (2004). Huntington's disease: present treatments and future therapeutic modalities. International Clinical Psychopharmacology 19(2): 51-62. Bordelon, Y.M., Chesselet, M.F., Erecinska, M. and Silver, I.A. (1998). Effects of intrastriatal injection of quinolinic acid on electrical activity and extracellular ion concentrations in rat striatum in vivo. Neuroscience 83(2): 459-69. Bordelon, Y.M., Mackenzie, L. and Chesselet, M.F. (1999). Morphology and compartmental location of cells exhibiting DNA damage after quinolinic acid injections into rat striatum. Journal of Comparative Neurology 412(1): 38-50. Borlongan, C.V., Koutouzis, T.K., Freeman, T.B., Hauser, R.A., Cahill, D.W. and Sanberg, P.R. (1997). Hyperactivity and hypoactivity in a rat model of Huntington's disease: the systemic 3nitropropionic acid model. Brain Research. Brain Research Protocols 1(3): 253-7. Borlongan, C.V., Skinner, S.J., Geaney, M., Vasconcellos, A.V., Elliott, R.B. and Emerich, D.F. (2004). Neuroprotection by encapsulated choroid plexus in a rodent model of Huntington's disease. Neuroreport 15(16): 2521-5. Borrell-Pages, M., Zala, D., Humbert, S. and Saudou, F. (2006). Huntington's disease: from huntingtin function and dysfunction to therapeutic strategies. Cellular & Molecular Life Sciences 63(22): 2642-60. 221 Literature References Boyd, J.G. and Gordon, T. (2002). A dose-dependent facilitation and inhibition of peripheral nerve regeneration by brain-derived neurotrophic factor. European Journal of Neuroscience 15(4): 613-26. Bramham, C.R. and Messaoudi, E. (2005). BDNF function in adult synaptic plasticity: the synaptic consolidation hypothesis. Progress in Neurobiology 76(2): 99-125. Brickell, K.L., Nicholson, L.F., Waldvogel, H.J. and Faull, R.L. (1999). Chemical and anatomical changes in the striatum and substantia nigra following quinolinic acid lesions in the striatum of the rat: a detailed time course of the cellular and GABA(A) receptor changes. Journal of Chemical Neuroanatomy 17(2): 75-97. Brown, L.L., Schneider, J.S. and Lidsky, T.I. (1997). Sensory and cognitive functions of the basal ganglia. Current Opinion in Neurobiology 7(2): 157-63. Bruyn, R.P. and Stoof, J.C. (1990). The quinolinic acid hypothesis in Huntington's chorea. Journal of Neurological Sciences 95(1): 29-38. Burger, C., Gorbatyuk, O.S., Velardo, M.J., Peden, C.S., Williams, P., Zolotukhin, S., Reier, P.J., Mandel, R.J. and Muzyczka, N. (2004). Recombinant AAV viral vectors pseudotyped with viral capsids from serotypes 1, 2, and 5 display differential efficiency and cell tropism after delivery to different regions of the central nervous system. Molecular Therapy 10(2): 302-17. Burger, C., Nguyen, F.N., Deng, J. and Mandel, R.J. (2005). Systemic mannitol-induced hyperosmolality amplifies rAAV2-mediated striatal transduction to a greater extent than local co-infusion. Molecular Therapy 11(2): 327-31. Burke, R.E. (2006). GDNF as a candidate striatal target-derived neurotrophic factor for the development of substantia nigra dopamine neurons. Journal of Neural Transmission. Supplementum(70): 41-5. Butterworth, N.J., Williams, L., Bullock, J.Y., Love, D.R., Faull, R.L. and Dragunow, M. (1998). Trinucleotide (CAG) repeat length is positively correlated with the degree of DNA fragmentation in Huntington's disease striatum. Neuroscience 87(1): 49-53. Byler, S.L., Shaffer, M.C. and Barth, T.M. (2006). Unilateral pallidotomy produces motor deficits and excesses in rats. Restorative Neurology & Neuroscience 24(3): 133-45. Campbell, M.J. and Morrison, J.H. (1989). Monoclonal antibody to neurofilament protein (SMI-32) labels a subpopulation of pyramidal neurons in the human and monkey neocortex. Journal of Comparative Neurology 282(2): 191-205. Canals, J.M., Marco, S., Checa, N., Michels, A., Perez-Navarro, E., Arenas, E. and Alberch, J. (1998). Differential regulation of the expression of nerve growth factor, brain-derived neurotrophic factor, and neurotrophin-3 after excitotoxicity in a rat model of Huntington's disease. Neurobiology of Disease 5(5): 357-64. Canals, J.M., Checa, N., Marco, S., Akerud, P., Michels, A., Perez-Navarro, E., Tolosa, E., Arenas, E. and Alberch, J. (2001). Expression of brain-derived neurotrophic factor in cortical neurons is regulated by striatal target area. Journal of Neuroscience 21(1): 117-24. Carter, B.J. (2005). Adeno-associated virus vectors in clinical trials. Human Gene Therapy 16(5): 541-50. Cattaneo, E. and McKay, R. (1990). Proliferation and differentiation of neuronal stem cells regulated by nerve growth factor. Nature 347(6295): 762-5. Cattaneo, E., Zuccato, C. and Tartari, M. (2005). Normal huntingtin function: an alternative approach to Huntington's disease. Nature Reviews Neuroscience 6(12): 919-30. 222 Literature References Cepeda, C., Ariano, M.A., Calvert, C.R., Flores-Hernandez, J., Chandler, S.H., Leavitt, B.R., Hayden, M.R. and Levine, M.S. (2001). NMDA receptor function in mouse models of Huntington disease. Journal of Neuroscience Research 66(4): 525-39. Chao, M.V. (1994). The p75 neurotrophin receptor. Journal of Neurobiology 25(11): 1373-85. Chao, M.V. and Hempstead, B.L. (1995). p75 and Trk: a two-receptor system. Trends in Neurosciences 18(7): 321-6. Charriaut-Marlangue, C. and Ben-Ari, Y. (1995). A cautionary note on the use of the TUNEL stain to determine apoptosis. Neuroreport 7(1): 61-4. Checa, N., Canals, J.M., Gratacos, E. and Alberch, J. (2001). TrkB and TrkC are differentially regulated by excitotoxicity during development of the basal ganglia. Experimental Neurology 172(2): 282-92. Chen, K., Henry, R.A., Hughes, S.M. and Connor, B. (2007). Creating a neurogenic environment: The role of BDNF and FGF2. Molecular and Cellular Neurosciences. Chen, N., Luo, T., Wellington, C., Metzler, M., McCutcheon, K., Hayden, M.R. and Raymond, L.A. (1999). Subtype-specific enhancement of NMDA receptor currents by mutant huntingtin. Journal of Neurochemistry 72(5): 1890-8. Chen, Z.Y., Patel, P.D., Sant, G., Meng, C.X., Teng, K.K., Hempstead, B.L. and Lee, F.S. (2004). Variant brain-derived neurotrophic factor (BDNF) (Met66) alters the intracellular trafficking and activity-dependent secretion of wild-type BDNF in neurosecretory cells and cortical neurons. Journal of Neuroscience 24(18): 4401-11. Chiarugi, A., Meli, E. and Moroni, F. (2001). Similarities and differences in the neuronal death processes activated by 3OH-kynurenine and quinolinic acid. Journal of Neurochemistry 77(5): 1310-8. Chittka, A., Arevalo, J.C., Rodriguez-Guzman, M., Perez, P., Chao, M.V. and Sendtner, M. (2004). The p75NTR-interacting protein SC1 inhibits cell cycle progression by transcriptional repression of cyclin E. Journal of Cell Biology 164(7): 985-96. Chmielnicki, E., Benraiss, A., Economides, A.N. and Goldman, S.A. (2004). Adenovirally expressed noggin and brain-derived neurotrophic factor cooperate to induce new medium spiny neurons from resident progenitor cells in the adult striatal ventricular zone. Journal of Neuroscience 24(9): 2133-42. Choi, D.W. (1988). Glutamate neurotoxicity and diseases of the nervous system. Neuron 1(8): 62334. Choi-Lundberg, D.L. and Bohn, M.C. (1995). Ontogeny and distribution of glial cell line-derived neurotrophic factor (GDNF) mRNA in rat. Brain Research. Developmental Brain Research 85(1): 80-8. Choi-Lundberg, D.L., Lin, Q., Schallert, T., Crippens, D., Davidson, B.L., Chang, Y.N., Chiang, Y.L., Qian, J., Bardwaj, L. and Bohn, M.C. (1998). Behavioral and cellular protection of rat dopaminergic neurons by an adenoviral vector encoding glial cell line-derived neurotrophic factor. Experimental Neurology 154(2): 261-75. Choo, Y.S., Johnson, G.V., MacDonald, M., Detloff, P.J. and Lesort, M. (2004). Mutant huntingtin directly increases susceptibility of mitochondria to the calcium-induced permeability transition and cytochrome c release. Human Molecular Genetics 13(14): 1407-20. 223 Literature References Cicchetti, F. and Parent, A. (1996a). Striatal interneurons in Huntington's disease: selective increase in the density of calretinin-immunoreactive medium-sized neurons. Movement Disorders 11(6): 619-26. Cicchetti, F., Gould, P.V. and Parent, A. (1996b). Sparing of striatal neurons coexpressing calretinin and substance P (NK1) receptor in Huntington's disease. Brain Research 730(1-2): 232-7. Clark, K.R., Sferra, T.J. and Johnson, P.R. (1997). Recombinant adeno-associated viral vectors mediate long-term transgene expression in muscle. Human Gene Therapy 8(6): 659-69. Cohen, G.M., Sun, X.M., Fearnhead, H., MacFarlane, M., Brown, D.G., Snowden, R.T. and Dinsdale, D. (1994). Formation of large molecular weight fragments of DNA is a key committed step of apoptosis in thymocytes. Journal of Immunology 153(2): 507-16. Connor, B. and Dragunow, M. (1998). The role of neuronal growth factors in neurodegenerative disorders of the human brain. Brain Research - Brain Research Reviews 27(1): 1-39. Conover, J.C., Ip, N.Y., Poueymirou, W.T., Bates, B., Goldfarb, M.P., DeChiara, T.M. and Yancopoulos, G.D. (1993). Ciliary neurotrophic factor maintains the pluripotentiality of embryonic stem cells. Development - Supplement 119(3): 559-65. Cory, S. and Adams, J.M. (2002). The Bcl2 family: regulators of the cellular life-or-death switch. Nature Reviews. Cancer 2(9): 647-56. Costantini, L.C., Feinstein, S.C., Radeke, M.J. and Snyder-Keller, A. (1999). Compartmental expression of trkB receptor protein in the developing striatum. Neuroscience 89(2): 505-13. Cowan, C.M. and Raymond, L.A. (2006). Selective neuronal degeneration in Huntington's disease. Current Topics in Developmental Biology 75: 25-71. Crocker, S.J., Wigle, N., Liston, P., Thompson, C.S., Lee, C.J., Xu, D., Roy, S., Nicholson, D.W., Park, D.S., MacKenzie, A., Korneluk, R.G. and Robertson, G.S. (2001). NAIP protects the nigrostriatal dopamine pathway in an intrastriatal 6-OHDA rat model of Parkinson's disease. European Journal of Neuroscience 14(2): 391-400. Croll, S.D., Suri, C., Compton, D.L., Simmons, M.V., Yancopoulos, G.D., Lindsay, R.M., Wiegand, S.J., Rudge, J.S. and Scharfman, H.E. (1999). Brain-derived neurotrophic factor transgenic mice exhibit passive avoidance deficits, increased seizure severity and in vitro hyperexcitability in the hippocampus and entorhinal cortex. Neuroscience 93(4): 1491-506. Crossman, A.R. (1987). Primate models of dyskinesia: the experimental approach to the study of basal ganglia-related involuntary movement disorders. Neuroscience 21(1): 1-40. Cudkowicz, M. and Kowall, N.W. (1990). Degeneration of pyramidal projection neurons in Huntington's disease cortex. Annals of Neurology 27(2): 200-4. Curtis, M.A., Penney, E.B., Pearson, A.G., van Roon-Mom, W.M., Butterworth, N.J., Dragunow, M., Connor, B. and Faull, R.L. (2003). Increased cell proliferation and neurogenesis in the adult human Huntington's disease brain. Proceedings of the National Academy of Sciences of the United States of America 100(15): 9023-7. Curtis, M.A., Penney, E.B., Pearson, J., Dragunow, M., Connor, B. and Faull, R.L. (2005). The distribution of progenitor cells in the subependymal layer of the lateral ventricle in the normal and Huntington's disease human brain. Neuroscience 132(3): 777-88. Cyr, M., Beaulieu, J.M., Laakso, A., Sotnikova, T.D., Yao, W.D., Bohn, L.M., Gainetdinov, R.R. and Caron, M.G. (2003). Sustained elevation of extracellular dopamine causes motor dysfunction and selective degeneration of striatal GABAergic neurons. Proceedings of the National Academy of Sciences of the United States of America 100(19): 11035-40. 224 Literature References Das, K.P., Chao, S.L., White, L.D., Haines, W.T., Harry, G.J., Tilson, H.A. and Barone, S., Jr. (2001). Differential patterns of nerve growth factor, brain-derived neurotrophic factor and neurotrophin-3 mRNA and protein levels in developing regions of rat brain. Neuroscience 103(3): 739-61. Davidson, B.L., Stein, C.S., Heth, J.A., Martins, I., Kotin, R.M., Derksen, T.A., Zabner, J., Ghodsi, A. and Chiorini, J.A. (2000). Recombinant adeno-associated virus type 2, 4, and 5 vectors: transduction of variant cell types and regions in the mammalian central nervous system. Proceedings of the National Academy of Sciences of the United States of America 97(7): 3428-32. Davies, S.W. and Roberts, P.J. (1987). No evidence for preservation of somatostatin-containing neurons after intrastriatal injections of quinolinic acid. Nature 327(6120): 326-9. Davies, S.W. and Beardsall, K. (1992). Nerve growth factor selectively prevents excitotoxin induced degeneration of striatal cholinergic neurones. Neuroscience Letters 140(2): 161-4. de Almeida, L.P., Zala, D., Aebischer, P. and Deglon, N. (2001). Neuroprotective effect of a CNTFexpressing lentiviral vector in the quinolinic acid rat model of Huntington's disease. Neurobiology of Disease 8(3): 433-46. DeChiara, T.M., Vejsada, R., Poueymirou, W.T., Acheson, A., Suri, C., Conover, J.C., Friedman, B., McClain, J., Pan, L. and Stahl, N. (1995). Mice lacking the CNTF receptor, unlike mice lacking CNTF, exhibit profound motor neuron deficits at birth. Cell 83(2): 313-22. DeLong, M.R. (1990). Primate models of movement disorders of basal ganglia origin. Trends in Neurosciences 13(7): 281-5. Detrait, E.R., Bowers, W.J., Halterman, M.W., Giuliano, R.E., Bennice, L., Federoff, H.J. and Richfield, E.K. (2002). Reporter gene transfer induces apoptosis in primary cortical neurons. Molecular Therapy 5(6): 723-30. Deveraux, Q.L., Leo, E., Stennicke, H.R., Welsh, K., Salvesen, G.S. and Reed, J.C. (1999). Cleavage of human inhibitor of apoptosis protein XIAP results in fragments with distinct specificities for caspases. EMBO Journal 18(19): 5242-51. Deveraux, Q.L. and Reed, J.C. (1999). IAP family proteins--suppressors of apoptosis. Genes & Development 13(3): 239-52. DiFiglia, M., Christakos, S. and Aronin, N. (1989). Ultrastructural localization of immunoreactive calbindin-D28k in the rat and monkey basal ganglia, including subcellular distribution with colloidal gold labeling. Journal of Comparative Neurology 279(4): 653-65. DiFiglia, M. (1990). Excitotoxic injury of the neostriatum: a model for Huntington's disease. Trends in Neurosciences 13(7): 286-9. Dobrossy, M.D. and Dunnett, S.B. (2003). Motor training effects on recovery of function after striatal lesions and striatal grafts. Experimental Neurology 184(1): 274-84. Dobrowsky, R.T., Werner, M.H., Castellino, A.M., Chao, M.V. and Hannun, Y.A. (1994). Activation of the sphingomyelin cycle through the low-affinity neurotrophin receptor. Science 265(5178): 1596-9. Dowd, E., Monville, C., Torres, E.M. and Dunnett, S.B. (2005). The Corridor Task: a simple test of lateralised response selection sensitive to unilateral dopamine deafferentation and graftderived dopamine replacement in the striatum. Brain Research Bulletin 68(1-2): 24-30. 225 Literature References Dragunow, M., Beilharz, E., Mason, B., Lawlor, P., Abraham, W. and Gluckman, P. (1993). Brainderived neurotrophic factor expression after long-term potentiation. Neuroscience Letters 160(2): 232-6. Dragunow, M., Faull, R.L., Lawlor, P., Beilharz, E.J., Singleton, K., Walker, E.B. and Mee, E. (1995). In situ evidence for DNA fragmentation in Huntington's disease striatum and Alzheimer's disease temporal lobes. Neuroreport 6(7): 1053-7. Drake, C.T., Milner, T.A. and Patterson, S.L. (1999). Ultrastructural localization of full-length trkB immunoreactivity in rat hippocampus suggests multiple roles in modulating activity-dependent synaptic plasticity. Journal of Neuroscience 19(18): 8009-26. Duan, D., Sharma, P., Yang, J., Yue, Y., Dudus, L., Zhang, Y., Fisher, K.J. and Engelhardt, J.F. (1998). Circular intermediates of recombinant adeno-associated virus have defined structural characteristics responsible for long-term episomal persistence in muscle tissue. Journal of Virology 72(11): 8568-77. Duan, D., Yan, Z., Yue, Y. and Engelhardt, J.F. (1999). Structural analysis of adeno-associated virus transduction circular intermediates. Virology 261(1): 8-14. Duan, W., Guo, Z., Jiang, H., Ware, M., Li, X.J. and Mattson, M.P. (2003). Dietary restriction normalizes glucose metabolism and BDNF levels, slows disease progression, and increases survival in huntingtin mutant mice. Proceedings of the National Academy of Sciences of the United States of America 100(5): 2911-6. Dunnett, S.B., Isacson, O., Sirinathsinghji, D.J., Clarke, D.J. and Bjorklund, A. (1988). Striatal grafts in rats with unilateral neostriatal lesions--III. Recovery from dopamine-dependent motor asymmetry and deficits in skilled paw reaching. Neuroscience 24(3): 813-20. Dure, L.S.t., Wiess, S., Standaert, D.G., Rudolf, G., Testa, C.M. and Young, A.B. (1995). DNA fragmentation and immediate early gene expression in rat striatum following quinolinic acid administration. Experimental Neurology 133(2): 207-14. During, M.J., Young, D., Baer, K., Lawlor, P. and Klugmann, M. (2003). Development and optimization of adeno-associated virus vector transfer into the central nervous system. Methods in Molecular Medicine 76: 221-36. Eberhardt, O., Coelln, R.V., Kugler, S., Lindenau, J., Rathke-Hartlieb, S., Gerhardt, E., Haid, S., Isenmann, S., Gravel, C., Srinivasan, A., Bahr, M., Weller, M., Dichgans, J. and Schulz, J.B. (2000). Protection by synergistic effects of adenovirus-mediated X-chromosome-linked inhibitor of apoptosis and glial cell line-derived neurotrophic factor gene transfer in the 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine model of Parkinson's disease. Journal of Neuroscience 20(24): 9126-34. Ellison, D.W., Beal, M.F., Mazurek, M.F., Malloy, J.R., Bird, E.D. and Martin, J.B. (1987). Amino acid neurotransmitter abnormalities in Huntington's disease and the quinolinic acid animal model of Huntington's disease. Brain 110(Pt 6): 1657-73. Emerich, D.F., Lindner, M.D., Winn, S.R., Chen, E.Y., Frydel, B.R. and Kordower, J.H. (1996). Implants of encapsulated human CNTF-producing fibroblasts prevent behavioral deficits and striatal degeneration in a rodent model of Huntington's disease. Journal of Neuroscience 16(16): 5168-81. Emerich, D.F., Cain, C.K., Greco, C., Saydoff, J.A., Hu, Z.Y., Liu, H. and Lindner, M.D. (1997a). Cellular delivery of human CNTF prevents motor and cognitive dysfunction in a rodent model of Huntington's disease. Cell Transplantation 6(3): 249-66. 226 Literature References Emerich, D.F., Winn, S.R., Hantraye, P.M., Peschanski, M., Chen, E.Y., Chu, Y., McDermott, P., Baetge, E.E. and Kordower, J.H. (1997b). Protective effect of encapsulated cells producing neurotrophic factor CNTF in a monkey model of Huntington's disease. Nature 386(6623): 3959. Emerich, D.F. (1999). Encapsulated CNTF-producing cells for Huntington's disease. Cell Transplantation 8(6): 581-2. Emerich, D.F. and Winn, S.R. (2004a). Neuroprotective effects of encapsulated CNTF-producing cells in a rodent model of Huntington's disease are dependent on the proximity of the implant to the lesioned striatum. Cell Transplantation 13(3): 253-9. Emerich, D.F. (2004b). Dose-dependent neurochemical and functional protection afforded by encapsulated CNTF-producing cells. Cell Transplantation 13(7-8): 839-44. Emerich, D.F., Thanos, C.G., Goddard, M., Skinner, S.J., Geany, M.S., Bell, W.J., Bintz, B., Schneider, P., Chu, Y., Babu, R.S., Borlongan, C.V., Boekelheide, K., Hall, S., Bryant, B. and Kordower, J.H. (2006). Extensive neuroprotection by choroid plexus transplants in excitotoxin lesioned monkeys. Neurobiology of Disease 23(2): 471-80. Engelender, S., Sharp, A.H., Colomer, V., Tokito, M.K., Lanahan, A., Worley, P., Holzbaur, E.L. and Ross, C.A. (1997). Huntingtin-associated protein 1 (HAP1) interacts with the p150Glued subunit of dynactin. Human Molecular Genetics 6(13): 2205-12. Ernfors, P., Merlio, J.P. and Persson, H. (1992). Cells Expressing mRNA for Neurotrophins and their Receptors During Embryonic Rat Development. European Journal of Neuroscience 4(11): 1140-1158. Escartin, C., Boyer, F., Bemelmans, A.P., Hantraye, P. and Brouillet, E. (2004). Insulin growth factor-1 protects against excitotoxicity in the rat striatum. Neuroreport 15(14): 2251-4. Eslamboli, A., Georgievska, B., Ridley, R.M., Baker, H.F., Muzyczka, N., Burger, C., Mandel, R.J., Annett, L. and Kirik, D. (2005). Continuous low-level glial cell line-derived neurotrophic factor delivery using recombinant adeno-associated viral vectors provides neuroprotection and induces behavioral recovery in a primate model of Parkinson's disease. Journal of Neuroscience 25(4): 769-77. Fallon, J., Reid, S., Kinyamu, R., Opole, I., Opole, R., Baratta, J., Korc, M., Endo, T.L., Duong, A., Nguyen, G., Karkehabadhi, M., Twardzik, D., Patel, S. and Loughlin, S. (2000). In vivo induction of massive proliferation, directed migration, and differentiation of neural cells in the adult mammalian brain. Proceedings of the National Academy of Sciences of the United States of America 97(26): 14686-91. Fang, W., Rivard, J.J., Mueller, D.L. and Behrens, T.W. (1994). Cloning and molecular characterization of mouse bcl-x in B and T lymphocytes. Journal of Immunology 153(10): 4388-98. Ferrante, R.J., Kowall, N.W., Beal, M.F., Martin, J.B., Bird, E.D. and Richardson, E.P., Jr. (1987). Morphologic and histochemical characteristics of a spared subset of striatal neurons in Huntington's disease. Journal of Neuropathology & Experimental Neurology 46(1): 12-27. Ferrer, I., Goutan, E., Marin, C., Rey, M.J. and Ribalta, T. (2000). Brain-derived neurotrophic factor in Huntington disease. Brain Research 866(1-2): 257-61. Figueredo-Cardenas, G., Anderson, K.D., Chen, Q., Veenman, C.L. and Reiner, A. (1994). Relative survival of striatal projection neurons and interneurons after intrastriatal injection of quinolinic acid in rats. Experimental Neurology 129(1): 37-56. 227 Literature References Figueredo-Cardenas, G., Harris, C.L., Anderson, K.D. and Reiner, A. (1998). Relative resistance of striatal neurons containing calbindin or parvalbumin to quinolinic acid-mediated excitotoxicity compared to other striatal neuron types. Experimental Neurology 149(2): 356-72. Filloux, F. and Townsend, J.J. (1993). Pre- and postsynaptic neurotoxic effects of dopamine demonstrated by intrastriatal injection. Experimental Neurology 119(1): 79-88. Frank, L., Ventimiglia, R., Anderson, K., Lindsay, R.M. and Rudge, J.S. (1996). BDNF downregulates neurotrophin responsiveness, TrkB protein and TrkB mRNA levels in cultured rat hippocampal neurons. European Journal of Neuroscience 8(6): 1220-30. Fricker, R.A., Annett, L.E., Torres, E.M. and Dunnett, S.B. (1996). The placement of a striatal ibotenic acid lesion affects skilled forelimb use and the direction of drug-induced rotation. Brain Research Bulletin 41(6): 409-16. Friedman, B., Scherer, S.S., Rudge, J.S., Helgren, M., Morrisey, D., McClain, J., Wang, D.Y., Wiegand, S.J., Furth, M.E. and Lindsay, R.M. (1992). Regulation of ciliary neurotrophic factor expression in myelin-related Schwann cells in vivo. Neuron 9(2): 295-305. Friedman, W.J., Olson, L. and Persson, H. (1991). Temporal and spatial expression of NGF receptor mRNA during postnatal rat brain development analyzed by in situ hybridization. Brain Research. Developmental Brain Research 63(1-2): 43-51. Friedman, W.J. and Greene, L.A. (1999). Neurotrophin signaling via Trks and p75. Experimental Cell Research 253(1): 131-42. Frim, D.M., Short, M.P., Rosenberg, W.S., Simpson, J., Breakefield, X.O. and Isacson, O. (1993a). Local protective effects of nerve growth factor-secreting fibroblasts against excitotoxic lesions in the rat striatum. [see comments.]. Journal of Neurosurgery 78(2): 267-73. Frim, D.M., Uhler, T.A., Short, M.P., Ezzedine, Z.D., Klagsbrun, M., Breakefield, X.O. and Isacson, O. (1993b). Effects of biologically delivered NGF, BDNF and bFGF on striatal excitotoxic lesions. Neuroreport 4(4): 367-70. Frim, D.M., Simpson, J., Uhler, T.A., Short, M.P., Bossi, S.R., Breakefield, X.O. and Isacson, O. (1993c). Striatal degeneration induced by mitochondrial blockade is prevented by biologically delivered NGF. Journal of Neuroscience Research 35(4): 452-8. Gafni, J., Hermel, E., Young, J.E., Wellington, C.L., Hayden, M.R. and Ellerby, L.M. (2004). Inhibition of calpain cleavage of huntingtin reduces toxicity: accumulation of calpain/caspase fragments in the nucleus. Journal of Biological Chemistry 279(19): 20211-20. Galpern, W.R., Matthews, R.T., Beal, M.F. and Isacson, O. (1996). NGF attenuates 3-nitrotyrosine formation in a 3-NP model of Huntington's disease. Neuroreport 7(15-17): 2639-42. Gao, G., Vandenberghe, L.H., Alvira, M.R., Lu, Y., Calcedo, R., Zhou, X. and Wilson, J.M. (2004). Clades of Adeno-associated viruses are widely disseminated in human tissues. Journal of Virology 78(12): 6381-8. Garin, M.I., Garrett, E., Tiberghien, P., Apperley, J.F., Chalmers, D., Melo, J.V. and Ferrand, C. (2001). Molecular mechanism for ganciclovir resistance in human T lymphocytes transduced with retroviral vectors carrying the herpes simplex virus thymidine kinase gene. Blood 97(1): 122-9. Garrity-Moses, M.E., Teng, Q., Liu, J., Tanase, D. and Boulis, N.M. (2005). Neuroprotective adenoassociated virus Bcl-xL gene transfer in models of motor neuron disease. Muscle and Nerve 32(6): 734-44. 228 Literature References Garrity-Moses, M.E., Teng, Q., Krudy, C., Yang, J., Federici, T. and Boulis, N.M. (2006). X-Linked inhibitor of apoptosis protein gene-based neuroprotection for the peripheral nervous system. Neurosurgery 59(1): 172-82; discussion 172-82. Gascon, E., Vutskits, L., Jenny, B., Durbec, P. and Kiss, J.Z. (2007). PSA-NCAM in postnatally generated immature neurons of the olfactory bulb: a crucial role in regulating p75 expression and cell survival. Development 134(6): 1181-90. Gauthier, L.R., Charrin, B.C., Borrell-Pages, M., Dompierre, J.P., Rangone, H., Cordelieres, F.P., De Mey, J., MacDonald, M.E., Lessmann, V., Humbert, S. and Saudou, F. (2004). Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 118(1): 127-38. Gavalda, N., Perez-Navarro, E., Gratacos, E., Comella, J.X. and Alberch, J. (2004). Differential involvement of phosphatidylinositol 3-kinase and p42/p44 mitogen activated protein kinase pathways in brain-derived neurotrophic factor-induced trophic effects on cultured striatal neurons. Molecular and Cellular Neurosciences 25(3): 460-8. Gavrieli, Y., Sherman, Y. and Ben-Sasson, S.A. (1992). Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. Journal of Cell Biology 119(3): 493501. Georgievska, B., Kirik, D. and Bjorklund, A. (2002). Aberrant sprouting and downregulation of tyrosine hydroxylase in lesioned nigrostriatal dopamine neurons induced by long-lasting overexpression of glial cell line derived neurotrophic factor in the striatum by lentiviral gene transfer. Experimental Neurology 177(2): 461-74. Gerfen, C.R. (1985). The neostriatal mosaic. I. Compartmental organization of projections from the striatum to the substantia nigra in the rat. Journal of Comparative Neurology 236(4): 454-76. Gerfen, C.R., Baimbridge, K.G. and Miller, J.J. (1985). The neostriatal mosaic: compartmental distribution of calcium-binding protein and parvalbumin in the basal ganglia of the rat and monkey. Proceedings of the National Academy of Sciences of the United States of America 82(24): 8780-4. Gerfen, C.R. (1992). The neostriatal mosaic: multiple levels of compartmental organization. Trends in Neurosciences 15(4): 133-9. Glass, M., Dragunow, M. and Faull, R.L. (2000). The pattern of neurodegeneration in Huntington's disease: a comparative study of cannabinoid, dopamine, adenosine and GABA(A) receptor alterations in the human basal ganglia in Huntington's disease. Neuroscience 97(3): 505-19. Goffredo, D., Rigamonti, D., Zuccato, C., Tartari, M., Valenza, M. and Cattaneo, E. (2005). Prevention of cytosolic IAPs degradation: a potential pharmacological target in Huntington's Disease. Pharmacological Research 52(2): 140-50. Goldstein, J.C., Rodier, F., Garbe, J.C., Stampfer, M.R. and Campisi, J. (2005). Caspaseindependent cytochrome c release is a sensitive measure of low-level apoptosis in cell culture models. Aging Cell 4(4): 217-22. Gonzalez-Garcia, M., Perez-Ballestero, R., Ding, L., Duan, L., Boise, L.H., Thompson, C.B. and Nunez, G. (1994). Bcl-XL is the major bcl-x mRNA form expressed during murine development and its product localizes to mitochondria. Development 120(10): 3033-42. Gonzalez-Garcia, M., Garcia, I., Ding, L., O'Shea, S., Boise, L.H., Thompson, C.B. and Nunez, G. (1995). Bcl-x is expressed in embryonic and postnatal neural tissues and functions to prevent neuronal cell death. Proceedings of the National Academy of Sciences of the United States of America 92(10): 4304-8. 229 Literature References Goodman, L.J., Valverde, J., Lim, F., Geschwind, M.D., Federoff, H.J., Geller, A.I. and Hefti, F. (1996). Regulated release and polarized localization of brain-derived neurotrophic factor in hippocampal neurons. Molecular & Cellular Neurosciences 7(3): 222-38. Gordon, R.J., Tattersfield, A.S., Vazey, E.M., Kells, A.P., McGregor, A.L., Hughes, S.M. and Connor, B. (2007). Temporal Profile of Subventricular Zone Progenitor Cell Migration Following Quinolinic Acid Induced Striatal Cell Loss. Neuroscience In Press. Gould, T.D. and Manji, H.K. (2005). DARPP-32: A molecular switch at the nexus of reward pathway plasticity. Proceedings of the National Academy of Sciences of the United States of America 102(2): 253-4. Goverdhana, S., Puntel, M., Xiong, W., Zirger, J.M., Barcia, C., Curtin, J.F., Soffer, E.B., Mondkar, S., King, G.D., Hu, J., Sciascia, S.A., Candolfi, M., Greengold, D.S., Lowenstein, P.R. and Castro, M.G. (2005). Regulatable gene expression systems for gene therapy applications: progress and future challenges. Molecular Therapy 12(2): 189-211. Greengard, P., Allen, P.B. and Nairn, A.C. (1999). Beyond the dopamine receptor: the DARPP32/protein phosphatase-1 cascade. Neuron 23(3): 435-47. Grieger, J.C. and Samulski, R.J. (2005). Adeno-associated virus as a gene therapy vector: vector development, production and clinical applications. Advances in Biochemical Engineering and Biotechnology 99: 119-45. Gross, A., McDonnell, J.M. and Korsmeyer, S.J. (1999). BCL-2 family members and the mitochondria in apoptosis. Genes & Development 13(15): 1899-911. Gross, A. (2001). BCL-2 proteins: regulators of the mitochondrial apoptotic program. IUBMB Life 52(3-5): 231-6. Guidetti, P., Luthi-Carter, R.E., Augood, S.J. and Schwarcz, R. (2004). Neostriatal and cortical quinolinate levels are increased in early grade Huntington's disease. Neurobiology of Disease 17(3): 455-61. Guillin, O., Diaz, J., Carroll, P., Griffon, N., Schwartz, J.C. and Sokoloff, P. (2001). BDNF controls dopamine D3 receptor expression and triggers behavioural sensitization. Nature 411(6833): 86-9. Gunawardena, S., Her, L.S., Brusch, R.G., Laymon, R.A., Niesman, I.R., Gordesky-Gold, B., Sintasath, L., Bonini, N.M. and Goldstein, L.S. (2003). Disruption of axonal transport by loss of huntingtin or expression of pathogenic polyQ proteins in Drosophila. Neuron 40(1): 25-40. Gutekunst, C.A., Li, S.H., Yi, H., Mulroy, J.S., Kuemmerle, S., Jones, R., Rye, D., Ferrante, R.J., Hersch, S.M. and Li, X.J. (1999). Nuclear and neuropil aggregates in Huntington's disease: relationship to neuropathology. Journal of Neuroscience 19(7): 2522-34. Haik, K.L., Shear, D.A., Schroeder, U., Sabel, B.A. and Dunbar, G.L. (2000). Quinolinic acid released from polymeric brain implants causes behavioral and neuroanatomical alterations in a rodent model of Huntington's disease. Experimental Neurology 163(2): 430-9. Hajnoczky, G., Davies, E. and Madesh, M. (2003). Calcium signaling and apoptosis. Biochemical and Biophysical Research Communications 304(3): 445-54. Halbert, C.L., Miller, A.D., McNamara, S., Emerson, J., Gibson, R.L., Ramsey, B. and Aitken, M.L. (2006). Prevalence of neutralizing antibodies against adeno-associated virus (AAV) types 2, 5, and 6 in cystic fibrosis and normal populations: Implications for gene therapy using AAV vectors. Human Gene Therapy 17(4): 440-7. 230 Literature References Handley, O.J., Naji, J.J., Dunnett, S.B. and Rosser, A.E. (2006). Pharmaceutical, cellular and genetic therapies for Huntington's disease. Clinical Science 110(1): 73-88. Harrington, K.M. and Kowall, N.W. (1991). Parvalbumin immunoreactive neurons resist degeneration in Huntington's disease striatum. Journal of Neuropathology & Experimental Neurology 50: 309. Hastings, T.G., Lewis, D.A. and Zigmond, M.J. (1996). Role of oxidation in the neurotoxic effects of intrastriatal dopamine injections. Proceedings of the National Academy of Sciences of the United States of America 93(5): 1956-61. Hattori, A., Luo, Y., Umegaki, H., Munoz, J. and Roth, G.S. (1998). Intrastriatal injection of dopamine results in DNA damage and apoptosis in rats. Neuroreport 9(11): 2569-72. Hauck, B., Chen, L. and Xiao, W. (2003). Generation and characterization of chimeric recombinant AAV vectors. Molecular Therapy 7(3): 419-25. Hedreen, J.C., Peyser, C.E., Folstein, S.E. and Ross, C.A. (1991). Neuronal loss in layers V and VI of cerebral cortex in Huntington's disease. Neuroscience Letters 133(2): 257-61. Helgren, M.E., Squinto, S.P., Davis, H.L., Parry, D.J., Boulton, T.G., Heck, C.S., Zhu, Y., Yancopoulos, G.D., Lindsay, R.M. and DiStefano, P.S. (1994). Trophic effect of ciliary neurotrophic factor on denervated skeletal muscle. Cell 76(3): 493-504. Henry, R.A., Hughes, S.M. and Connor, B. (2007). AAV-mediated delivery of BDNF augments neurogenesis in the normal and quinolinic acid-lesioned adult rat brain. European Journal of Neuroscience 25(12): 3513-25. Herdegen, T., Kovary, K., Buhl, A., Bravo, R., Zimmermann, M. and Gass, P. (1995). Basal expression of the inducible transcription factors c-Jun, JunB, JunD, c-Fos, FosB, and Krox-24 in the adult rat brain. Journal of Comparative Neurology 354(1): 39-56. Hersch, S.M. and Ferrante, R.J. (2004). Translating therapies for Huntington's disease from genetic animal models to clinical trials. Neurorx 1(3): 298-306. Hetman, M., Kanning, K., Cavanaugh, J.E. and Xia, Z. (1999). Neuroprotection by brain-derived neurotrophic factor is mediated by extracellular signal-regulated kinase and phosphatidylinositol 3-kinase. Journal of Biological Chemistry 274(32): 22569-80. Hickey, M.A. and Chesselet, M.F. (2003). Apoptosis in Huntington's disease. Progress in NeuroPsychopharmacology and Biological Psychiatry 27(2): 255-65. Hodgson, J.G., Agopyan, N., Gutekunst, C.A., Leavitt, B.R., LePiane, F., Singaraja, R., Smith, D.J., Bissada, N., McCutcheon, K., Nasir, J., Jamot, L., Li, X.J., Stevens, M.E., Rosemond, E., Roder, J.C., Phillips, A.G., Rubin, E.M., Hersch, S.M. and Hayden, M.R. (1999). A YAC mouse model for Huntington's disease with full-length mutant huntingtin, cytoplasmic toxicity, and selective striatal neurodegeneration. Neuron 23(1): 181-92. Hsich, G., Sena-Esteves, M. and Breakefield, X.O. (2002). Critical issues in gene therapy for neurologic disease. Human Gene Therapy 13: 579-604. Hu, Y., Leaver, S.G., Plant, G.W., Hendriks, W.T., Niclou, S.P., Verhaagen, J., Harvey, A.R. and Cui, Q. (2005). Lentiviral-mediated transfer of CNTF to schwann cells within reconstructed peripheral nerve grafts enhances adult retinal ganglion cell survival and axonal regeneration. Molecular Therapy 11(6): 906-15. Huang, D.C. and Strasser, A. (2000). BH3-Only proteins-essential initiators of apoptotic cell death. Cell 103(6): 839-42. 231 Literature References Huang, E.J. and Reichardt, L.F. (2001). Neurotrophins: roles in neuronal development and function. Annual Review of Neuroscience 24: 677-736. Hudson, J., Granholm, A.C., Gerhardt, G.A., Henry, M.A., Hoffman, A., Biddle, P., Leela, N.S., Mackerlova, L., Lile, J.D., Collins, F. and et al. (1995). Glial cell line-derived neurotrophic factor augments midbrain dopaminergic circuits in vivo. Brain Research Bulletin 36(5): 42532. Hughes, P.E., Alexi, T., Yoshida, T., Schreiber, S.S. and Knusel, B. (1996). Excitotoxic lesion of rat brain with quinolinic acid induces expression of p53 messenger RNA and protein and p53inducible genes Bax and Gadd-45 in brain areas showing DNA fragmentation. Neuroscience 74(4): 1143-60. Hughes, P.E., Alexi, T., Williams, C.E., Clark, R.G. and Gluckman, P.D. (1999). Administration of recombinant human Activin-A has powerful neurotrophic effects on select striatal phenotypes in the quinolinic acid lesion model of Huntington's disease. Neuroscience 92(1): 197-209. Huntington's Disease Collaborative Research Group (1993). A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. The Huntington's Disease Collaborative Research Group. Cell 72(6): 971-83. Ip, N.Y., McClain, J., Barrezueta, N.X., Aldrich, T.H., Pan, L., Li, Y., Wiegand, S.J., Friedman, B., Davis, S. and Yancopoulos, G.D. (1993). The alpha component of the CNTF receptor is required for signaling and defines potential CNTF targets in the adult and during development. Neuron 10(1): 89-102. Isacson, O., Dunnett, S.B. and Bjorklund, A. (1986). Graft-induced behavioral recovery in an animal model of Huntington disease. Proceedings of the National Academy of Sciences of the United States of America 83(8): 2728-32. Ito, H., Nakajima, A., Nomoto, H. and Furukawa, S. (2003). Neurotrophins facilitate neuronal differentiation of cultured neural stem cells via induction of mRNA expression of basic helixloop-helix transcription factors Mash1 and Math1. Journal of Neuroscience Research 71(5): 648-58. Ivkovic, S. and Ehrlich, M.E. (1999). Expression of the striatal DARPP-32/ARPP-21 phenotype in GABAergic neurons requires neurotrophins in vivo and in vitro. Journal of Neuroscience 19(13): 5409-19. Jana, N.R., Zemskov, E.A., Wang, G. and Nukina, N. (2001). Altered proteasomal function due to the expression of polyglutamine-expanded truncated N-terminal huntingtin induces apoptosis by caspase activation through mitochondrial cytochrome c release. Human Molecular Genetics 10(10): 1049-59. Jiang, L., Rampalli, S., George, D., Press, C., Bremer, E.G., O'Gorman, M.R. and Bohn, M.C. (2004). Tight regulation from a single tet-off rAAV vector as demonstrated by flow cytometry and quantitative, real-time PCR. Gene Therapy 11(13): 1057-67. Jing, S., Wen, D., Yu, Y., Holst, P.L., Luo, Y., Fang, M., Tamir, R., Antonio, L., Hu, Z., Cupples, R., Louis, J.C., Hu, S., Altrock, B.W. and Fox, G.M. (1996). GDNF-induced activation of the ret protein tyrosine kinase is mediated by GDNFR-alpha, a novel receptor for GDNF. Cell 85(7): 1113-24. Joel, D., Ayalon, L., Tarrasch, R., Veenman, L., Feldon, J. and Weiner, I. (1998). Electrolytic lesion of globus pallidus ameliorates the behavioral and neurodegenerative effects of quinolinic acid lesion of the striatum: a potential novel treatment in a rat model of Huntington's disease. Brain Research 787(1): 143-8. 232 Literature References Joel, D. (2001). Open interconnected model of basal ganglia-thalamocortical circuitry and its relevance to the clinical syndrome of Huntington's disease. Movement Disorders 16(3): 40723. Johnson, E.M., Jr., Chang, J.Y., Koike, T. and Martin, D.P. (1989). Why do neurons die when deprived of trophic factor? Neurobiology of Aging 10(5): 549-52; discussion 552-3. Jooss, K., Yang, Y., Fisher, K.J. and Wilson, J.M. (1998). Transduction of dendritic cells by DNA viral vectors directs the immune response to transgene products in muscle fibers. Journal of Virology 72(5): 4212-23. Kantor, O., Temel, Y., Holzmann, C., Raber, K., Nguyen, H.P., Cao, C., Turkoglu, H.O., Rutten, B.P., Visser-Vandewalle, V., Steinbusch, H.W., Blokland, A., Korr, H., Riess, O., von Horsten, S. and Schmitz, C. (2006). Selective striatal neuron loss and alterations in behavior correlate with impaired striatal function in Huntington's disease transgenic rats. Neurobiology of Disease 22(3): 538-47. Kaspar, B.K., Erickson, D., Schaffer, D., Hinh, L., Gage, F.H. and Peterson, D.A. (2002). Targeted retrograde gene delivery for neuronal protection. Molecular Therapy 5(1): 50-6. Katoh-Semba, R., Semba, R., Takeuchi, I.K. and Kato, K. (1998). Age-related changes in levels of brain-derived neurotrophic factor in selected brain regions of rats, normal mice and senescence-accelerated mice: a comparison to those of nerve growth factor and neurotrophin3. Neuroscience Research 31(3): 227-34. Kells, A.P., Fong, D.M., Dragunow, M., During, M.J., Young, D. and Connor, B. (2004). AAVmediated gene delivery of BDNF or GDNF is neuroprotective in a model of Huntington disease. Molecular Therapy 9(5): 682-8. Kells, A.P., Henry, R.A., Hughes, S.M. and Connor, B. (2006). Verification of functional AAVmediated neurotrophic and anti-apoptotic factor expression. Journal of Neuroscience Methods: doi:10.1016/j.jneumeth.2006.11.006. Kernie, S.G., Liebl, D.J. and Parada, L.F. (2000). BDNF regulates eating behavior and locomotor activity in mice. EMBO Journal 19(6): 1290-300. Kerr, J.F., Wyllie, A.H. and Currie, A.R. (1972). Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. British Journal of Cancer 26(4): 239-57. Kiechle, T., Dedeoglu, A., Kubilus, J., Kowall, N.W., Beal, M.F., Friedlander, R.M., Hersch, S.M. and Ferrante, R.J. (2002). Cytochrome C and caspase-9 expression in Huntington's disease. Neuromolecular Medicine 1(3): 183-95. Kim, R. (2005). Unknotting the roles of Bcl-2 and Bcl-xL in cell death. Biochemical and Biophysical Research Communications 333(2): 336-43. Kirik, D., Rosenblad, C. and Bjorklund, A. (2000a). Preservation of a functional nigrostriatal dopamine pathway by GDNF in the intrastriatal 6-OHDA lesion model depends on the site of administration of the trophic factor. European Journal of Neuroscience 12(11): 3871-82. Kirik, D., Rosenblad, C., Bjorklund, A. and Mandel, R.J. (2000b). Long-Term rAAV-Mediated Gene Transfer of GDNF in the Rat Parkinson’s Model: Intrastriatal But Not Intranigral Transduction Promotes Functional Regeneration in the Lesioned Nigrostriatal System. Journal of Neuroscience 20(12): 4686-4700. Kirschner, P.B., Henshaw, R., Weise, J., Trubetskoy, V., Finklestein, S., Schulz, J.B. and Beal, M.F. (1995). Basic fibroblast growth factor protects against excitotoxicity and chemical hypoxia in both neonatal and adult rats. Journal of Cerebral Blood Flow & Metabolism 15(4): 619-23. 233 Literature References Klein, R.L., Hamby, A.E., Gong, Y., Hirko, A.C., Wang, S., Hughes, J.A., King, M.A. and Meyer, E.M. (2002). Dose and promoter effects of adeno-associated viral vector for green fluorescent protein expression in the rat brain. Experimental Neurology 176: 66-72. Klugmann, M., Symes, C.W., Leichtlein, C.B., Klaussner, B.K., Dunning, J., Fong, D., Young, D. and During, M.J. (2005). AAV-mediated hippocampal expression of short and long Homer 1 proteins differentially affect cognition and seizure activity in adult rats. Molecular & Cellular Neurosciences 28(2): 347-60. Knapska, E. and Kaczmarek, L. (2004). A gene for neuronal plasticity in the mammalian brain: Zif268/Egr-1/NGFI-A/Krox-24/TIS8/ZENK? Progress in Neurobiology 74(4): 183-211. Kokaia, Z., Bengzon, J., Metsis, M., Kokaia, M., Persson, H. and Lindvall, O. (1993). Coexpression of neurotrophins and their receptors in neurons of the central nervous system. Proceedings of the National Academy of Sciences of the United States of America 90(14): 6711-5. Kokaia, Z., Andsberg, G., Yan, Q. and Lindvall, O. (1998). Rapid alterations of BDNF protein levels in the rat brain after focal ischemia: evidence for increased synthesis and anterograde axonal transport. Experimental Neurology 154(2): 289-301. Kordower, J.H., Chen, E.Y., Winkler, C., Fricker, R., Charles, V., Messing, A., Mufson, E.J., Wong, S.C., Rosenstein, J.M., Bjorklund, A., Emerich, D.F., Hammang, J. and Carpenter, M.K. (1997). Grafts of EGF-responsive neural stem cells derived from GFAP-hNGF transgenic mice: trophic and tropic effects in a rodent model of Huntington's disease. Journal of Comparative Neurology 387(1): 96-113. Kordower, J.H., Bloch, J., Ma, S.Y., Chu, Y., Palfi, S., Roitberg, B.Z., Emborg, M., Hantraye, P., Deglon, N. and Aebischer, P. (1999). Lentiviral gene transfer to the nonhuman primate brain. Experimental Neurology 160(1): 1-16. Kordower, J.H., Emborg, M.E., Bloch, J., Ma, S.Y., Chu, Y., Leventhal, L., McBride, J., Chen, E.Y., Palfi, S., Roitberg, B.Z., Brown, W.D., Holden, J.E., Pyzalski, R., Taylor, M.D., Carvey, P., Ling, Z., Trono, D., Hantraye, P., Deglon, N. and Aebischer, P. (2000). Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease. Science 290(5492): 767-73. Kotin, R.M., Linden, R.M. and Berns, K.I. (1992). Characterization of a preferred site on human chromosome 19q for integration of adeno-associated virus DNA by non-homologous recombination. EMBO Journal 11(13): 5071-8. Krestel, H.E., Mihaljevic, A.L., Hoffman, D.A. and Schneider, A. (2004). Neuronal co-expression of EGFP and beta-galactosidase in mice causes neuropathology and premature death. Neurobiology of Disease 17(2): 310-8. Krieglstein, K. (2004). Factors promoting survival of mesencephalic dopaminergic neurons. Cell and Tissue Research 318(1): 73-80. Kugler, S., Straten, G., Kreppel, F., Isenmann, S., Liston, P. and Bahr, M. (2000). The X-linked inhibitor of apoptosis (XIAP) prevents cell death in axotomized CNS neurons in vivo. Cell Death and Differentiation 7(9): 815-24. Kuipers, S.D. and Bramham, C.R. (2006). Brain-derived neurotrophic factor mechanisms and function in adult synaptic plasticity: new insights and implications for therapy. Current Opinion in Drug Discovery and Development 9(5): 580-6. Kuwana, T., Mackey, M.R., Perkins, G., Ellisman, M.H., Latterich, M., Schneiter, R., Green, D.R. and Newmeyer, D.D. (2002). Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane. Cell 111(3): 331-42. 234 Literature References Laforet, G.A., Sapp, E., Chase, K., McIntyre, C., Boyce, F.M., Campbell, M., Cadigan, B.A., Warzecki, L., Tagle, D.A., Reddy, P.H., Cepeda, C., Calvert, C.R., Jokel, E.S., Klapstein, G.J., Ariano, M.A., Levine, M.S., DiFiglia, M. and Aronin, N. (2001). Changes in cortical and striatal neurons predict behavioral and electrophysiological abnormalities in a transgenic murine model of Huntington's disease. Journal of Neuroscience 21(23): 9112-23. Landwehrmeyer, G.B., Standaert, D.G., Testa, C.M., Penney, J.B., Jr. and Young, A.B. (1995). NMDA receptor subunit mRNA expression by projection neurons and interneurons in rat striatum. Journal of Neuroscience 15(7 Pt 2): 5297-307. Lauterbach, H., Ried, C., Epstein, A.L., Marconi, P. and Brocker, T. (2005). Reduced immune responses after vaccination with a recombinant herpes simplex virus type 1 vector in the presence of antiviral immunity. Journal of General Virology 86(Pt 9): 2401-10. Leavitt, B.R., van Raamsdonk, J.M., Shehadeh, J., Fernandes, H., Murphy, Z., Graham, R.K., Wellington, C.L., Raymond, L.A. and Hayden, M.R. (2006). Wild-type huntingtin protects neurons from excitotoxicity. Journal of Neurochemistry 96(4): 1121-9. Lee, R., Kermani, P., Teng, K.K. and Hempstead, B.L. (2001). Regulation of cell survival by secreted proneurotrophins. Science 294(5548): 1945-8. Leegwater-Kim, J. and Cha, J.H. (2004). The paradigm of Huntington's disease: therapeutic opportunities in neurodegeneration. Neurorx 1(1): 128-38. Levi-Montalcini, R. and Hamburger, V. (1953). A diffusible agent of mouse sarcoma producing hyperplasia of sympathetic ganglia and hyperneurotization of viscera in the chick embryo. Journal of Experimental Zoology 123: 233-287. Levi-Montalcini, R. (1987). The nerve growth factor 35 years later. Science 237(4819): 1154-62. Levine, M.S., Klapstein, G.J., Koppel, A., Gruen, E., Cepeda, C., Vargas, M.E., Jokel, E.S., Carpenter, E.M., Zanjani, H., Hurst, R.S., Efstratiadis, A., Zeitlin, S. and Chesselet, M.F. (1999). Enhanced sensitivity to N-methyl-D-aspartate receptor activation in transgenic and knockin mouse models of Huntington's disease. Journal of Neuroscience Research 58(4): 51532. Levy, Y.S., Gilgun-Sherki, Y., Melamed, E. and Offen, D. (2005). Therapeutic potential of neurotrophic factors in neurodegenerative diseases. BioDrugs 19(2): 97-127. Li, H., Li, S.H., Johnston, H., Shelbourne, P.F. and Li, X.J. (2000). Amino-terminal fragments of mutant huntingtin show selective accumulation in striatal neurons and synaptic toxicity. Nature Genetics 25(4): 385-9. Li, S.H., Lam, S., Cheng, A.L. and Li, X.J. (2000). Intranuclear huntingtin increases the expression of caspase-1 and induces apoptosis. Human Molecular Genetics 9(19): 2859-67. Li, T., Fan, Y., Luo, Y., Xiao, B. and Lu, C. (2006). In vivo delivery of a XIAP (BIR3-RING) fusion protein containing the protein transduction domain protects against neuronal death induced by seizures. Experimental Neurology 197(2): 301-8. Lin, C.H., Tallaksen-Greene, S., Chien, W.M., Cearley, J.A., Jackson, W.S., Crouse, A.B., Ren, S., Li, X.J., Albin, R.L. and Detloff, P.J. (2001). Neurological abnormalities in a knock-in mouse model of Huntington's disease. Human Molecular Genetics 10(2): 137-44. Lin, L.F., Doherty, D.H., Lile, J.D., Bektesh, S. and Collins, F. (1993). GDNF: a glial cell linederived neurotrophic factor for midbrain dopaminergic neurons. Science 260(5111): 1130-2. 235 Literature References Linden, R.M., Winocour, E. and Berns, K.I. (1996). The recombination signals for adeno-associated virus site-specific integration. Proceedings of the National Academy of Sciences of the United States of America 93(15): 7966-72. Lindsay, R.M., Wiegand, S.J., Altar, C.A. and DiStefano, P.S. (1994). Neurotrophic factors: from molecule to man. Trends in Neurosciences 17(5): 182-90. Loeb, J.E., Cordier, W.S., Harris, M.E., Weitzman, M.D. and Hope, T.J. (1999). Enhanced expression of transgenes from adeno-associated virus vectors with the woodchuck hepatitis virus posttranscriptional regulatory element: implications for gene therapy. Human Gene Therapy 10(14): 2295-305. Loo, D.T. and Rillema, J.R. (1998). Measurement of cell death. Methods in Cell Biology 57: 25164. Lu, B. (2003). BDNF and activity-dependent synaptic modulation. Learning and Memory 10(2): 86-98. Luthi-Carter, R., Strand, A., Peters, N.L., Solano, S.M., Hollingsworth, Z.R., Menon, A.S., Frey, A.S., Spektor, B.S., Penney, E.B., Schilling, G., Ross, C.A., Borchelt, D.R., Tapscott, S.J., Young, A.B., Cha, J.H. and Olson, J.M. (2000). Decreased expression of striatal signaling genes in a mouse model of Huntington's disease. Human Molecular Genetics 9(9): 1259-71. Macdonald, V. and Halliday, G. (2002). Pyramidal cell loss in motor cortices in Huntington's disease. Neurobiology of Disease 10(3): 378-86. MacGibbon, G.A., Lawlor, P.A., Hughes, P., Young, D. and Dragunow, M. (1995). Differential expression of inducible transcription factors in basal ganglia neurons. Brain Research. Molecular Brain Research 34(2): 294-302. Maciel, E.N., Kaminski Schierle, G.S., Hansson, O., Brundin, P. and Castilho, R.F. (2003). Cyclosporin A and Bcl-2 do not inhibit quinolinic acid-induced striatal excitotoxicity in rodents. Experimental Neurology 183(2): 430-7. Malik, J.M., Shevtsova, Z., Bahr, M. and Kugler, S. (2005). Long-term in vivo inhibition of CNS neurodegeneration by Bcl-XL gene transfer. Molecular Therapy 11(3): 373-81. Mangiarini, L., Sathasivam, K., Seller, M., Cozens, B., Harper, A., Hetherington, C., Lawton, M., Trottier, Y., Lehrach, H., Davies, S.W. and Bates, G.P. (1996). Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87(3): 493-506. Marco, S., Canudas, A.M., Canals, J.M., Gavalda, N., Perez-Navarro, E. and Alberch, J. (2002). Excitatory amino acids differentially regulate the expression of GDNF, neurturin, and their receptors in the adult rat striatum. Experimental Neurology 174(2): 243-52. Martin, S.J. and Green, D.R. (1995). Protease activation during apoptosis: death by a thousand cuts? Cell 82(3): 349-52. Martinez-Serrano, A., Lundberg, C., Horellou, P., Fischer, W., Bentlage, C., Campbell, K., McKay, R.D., Mallet, J. and Bjorklund, A. (1995). CNS-derived neural progenitor cells for gene transfer of nerve growth factor to the adult rat brain: complete rescue of axotomized cholinergic neurons after transplantation into the septum. Journal of Neuroscience 15(8): 5668-80. Martinez-Serrano, A. and Bjorklund, A. (1996). Protection of the neostriatum against excitotoxic damage by neurotrophin-producing, genetically modified neural stem cells. Journal of Neuroscience 16(15): 4604-4616. 236 Literature References Martin-Iverson, M.T., Todd, K.G. and Altar, C.A. (1994). Brain-derived neurotrophic factor and neurotrophin-3 activate striatal dopamine and serotonin metabolism and related behaviors: interactions with amphetamine. Journal of Neuroscience 14(3 Pt 1): 1262-70. Martin-Iverson, M.T. and Altar, C.A. (1996). Spontaneous behaviours of rats are differentially affected by substantia nigra infusions of brain-derived neurotrophic factor and neurotrophin-3. European Journal of Neuroscience 8(8): 1696-706. Martinou, J.C. and Green, D.R. (2001). Breaking the mitochondrial barrier. Nature Reviews. Molecular Cell Biology 2(1): 63-7. Marty, S., Berzaghi Mda, P. and Berninger, B. (1997). Neurotrophins and activity-dependent plasticity of cortical interneurons. Trends in Neurosciences 20(5): 198-202. Marx, J. (2005). Neurodegeneration. Huntington's research points to possible new therapies. Science 310(5745): 43-5. Mastakov, M.Y., Baer, K., Xu, R., Fitzsimons, H. and During, M.J. (2001). Combined injection of rAAV with mannitol enhances gene expression in the rat brain. Molecular Therapy 3(2): 22532. Mastakov, M.Y., Baer, K., Symes, C.W., Leichtlein, C.B., Kotin, R.M. and During, M.J. (2002). Immunological aspects of recombinant adeno-associated virus delivery to the mammalian brain. Journal of Virology 76(16): 8446-54. Masu, Y., Wolf, E., Holtmann, B., Sendtner, M., Brem, G. and Thoenen, H. (1993). Disruption of the CNTF gene results in motor neuron degeneration. Nature 365(6441): 27-32. Matsushita, T., Elliger, S., Elliger, C., Podsakoff, G., Villarreal, L., Kurtzman, G.J., Iwaki, Y. and Colosi, P. (1998). Adeno-associated virus vectors can be efficiently produced without helper virus. Gene Therapy 5(7): 938-45. Mattson, M.P. (2000). Neuroprotective signaling and the aging brain: take away my food and let me run. Brain Research 886(1-2): 47-53. McBride, J.L., During, M.J., Wuu, J., Chen, E.Y., Leurgans, S.E. and Kordower, J.H. (2003). Structural and functional neuroprotection in a rat model of Huntington's disease by viral gene transfer of GDNF. Experimental Neurology 181(2): 213-23. McBride, J.L., Ramaswamy, S., Gasmi, M., Bartus, R.T., Herzog, C.D., Brandon, E.P., Zhou, L., Pitzer, M.R., Berry-Kravis, E.M. and Kordower, J.H. (2006). Viral delivery of glial cell linederived neurotrophic factor improves behavior and protects striatal neurons in a mouse model of Huntington's disease. Proceedings of the National Academy of Sciences of the United States of America 103(24): 9345-50. McGeorge, A.J. and Faull, R.L. (1989). The organization of the projection from the cerebral cortex to the striatum in the rat. Neuroscience 29(3): 503-37. Merlio, J.P., Ernfors, P., Kokaia, Z., Middlemas, D.S., Bengzon, J., Kokaia, M., Smith, M.L., Siesjo, B.K., Hunter, T. and Lindvall, O. (1993). Increased production of the TrkB protein tyrosine kinase receptor after brain insults. Neuron 10(2): 151-64. Merten, O.W., Geny-Fiamma, C. and Douar, A.M. (2005). Current issues in adeno-associated viral vector production. Gene Therapy 12 Suppl 1: S51-61. Meyer-Franke, A., Wilkinson, G.A., Kruttgen, A., Hu, M., Munro, E., Hanson, M.G., Jr., Reichardt, L.F. and Barres, B.A. (1998). Depolarization and cAMP elevation rapidly recruit TrkB to the plasma membrane of CNS neurons. Neuron 21(4): 681-93. 237 Literature References Miagkov, A., Turchan, J., Nath, A. and Drachman, D.B. (2004). Gene transfer of baculoviral p35 by adenoviral vector protects human cerebral neurons from apoptosis. DNA & Cell Biology 23(8): 496-501. Miranda, R.C., Sohrabji, F. and Toran-Allerand, C.D. (1993). Neuronal colocalization of mRNAs for neurotrophins and their receptors in the developing central nervous system suggests a potential for autocrine interactions. Proceedings of the National Academy of Sciences of the United States of America 90(14): 6439-43. Mitchell, I.J. (1990). Striatal outputs and dsykinesia. Journal of Psychopharmacology 4: 188-197. Mittoux, V., Joseph, J.M., Conde, F., Palfi, S., Dautry, C., Poyot, T., Bloch, J., Deglon, N., Ouary, S., Nimchinsky, E.A., Brouillet, E., Hof, P.R., Peschanski, M., Aebischer, P. and Hantraye, P. (2000). Restoration of cognitive and motor functions by ciliary neurotrophic factor in a primate model of Huntington's disease. Human Gene Therapy 11(8): 1177-87. Mittoux, V., Ouary, S., Monville, C., Lisovoski, F., Poyot, T., Conde, F., Escartin, C., Robichon, R., Brouillet, E., Peschanski, M. and Hantraye, P. (2002). Corticostriatopallidal neuroprotection by adenovirus-mediated ciliary neurotrophic factor gene transfer in a rat model of progressive striatal degeneration. Journal of Neuroscience 22(11): 4478-86. Miyoshi, H., Blomer, U., Takahashi, M., Gage, F.H. and Verma, I.M. (1998). Development of a selfinactivating lentivirus vector. Journal of Virology 72(10): 8150-7. Mizuno, K., Carnahan, J. and Nawa, H. (1994). Brain-derived neurotrophic factor promotes differentiation of striatal GABAergic neurons. Developmental Biology 165(1): 243-56. Mobley, W.C., Woo, J.E., Edwards, R.H., Riopelle, R.J., Longo, F.M., Weskamp, G., Otten, U., Valletta, J.S. and Johnston, M.V. (1989). Developmental regulation of nerve growth factor and its receptor in the rat caudate-putamen. Neuron 3(5): 655-64. Mochizuki, H., Miura, M., Shimada, T. and Mizuno, Y. (2002). Adeno-associated virus-mediated antiapoptotic gene delivery: in vivo gene therapy for neurological disorders. Methods 28(2): 248-52. Monahan, P.E. and Samulski, R.J. (2000). Adeno-associated virus vectors for gene therapy: more pros than cons? Molecular Medicine Today 6(11): 433-40. Montoya, C.P., Astell, S. and Dunnett, S.B. (1990). Effects of nigral and striatal grafts on skilled forelimb use in the rat. Progress in Brain Research 82: 459-66. Montoya, C.P., Campbell-Hope, L.J., Pemberton, K.D. and Dunnett, S.B. (1991). The "staircase test": a measure of independent forelimb reaching and grasping abilities in rats. Journal of Neuroscience Methods 36(2-3): 219-28. Morton, A.J., Lagan, M.A., Skepper, J.N. and Dunnett, S.B. (2000). Progressive formation of inclusions in the striatum and hippocampus of mice transgenic for the human Huntington's disease mutation. Journal of Neurocytology 29(9): 679-702. Mowla, S.J., Farhadi, H.F., Pareek, S., Atwal, J.K., Morris, S.J., Seidah, N.G. and Murphy, R.A. (2001). Biosynthesis and post-translational processing of the precursor to brain-derived neurotrophic factor. Journal of Biological Chemistry 276(16): 12660-6. Muchmore, S.W., Sattler, M., Liang, H., Meadows, R.P., Harlan, J.E., Yoon, H.S., Nettesheim, D., Chang, B.S., Thompson, C.B., Wong, S.L., Ng, S.L. and Fesik, S.W. (1996). X-ray and NMR structure of human Bcl-xL, an inhibitor of programmed cell death. Nature 381(6580): 335-41. 238 Literature References Mufson, E.J., Conner, J.M., Varon, S. and Kordower, J.H. (1994). Nerve growth factor-like immunoreactive profiles in the primate basal forebrain and hippocampal formation. Journal of Comparative Neurology 341(4): 507-19. Mufson, E.J., Kroin, J.S., Sendera, T.J. and Sobreviela, T. (1999). Distribution and retrograde transport of trophic factors in the central nervous system: functional implications for the treatment of neurodegenerative diseases. Progress in Neurobiology 57(4): 451-84. Murer, M.G., Yan, Q. and Raisman-Vozari, R. (2001). Brain-derived neurotrophic factor in the control human brain, and in Alzheimer's disease and Parkinson's disease. Progress in Neurobiology 63(1): 71-124. Muzyczka, N. and Warrington, K.H., Jr. (2005). Custom adeno-associated virus capsids: the next generation of recombinant vectors with novel tropism. Human Gene Therapy 16(4): 408-16. Naert, G., Ixart, G., Tapia-Arancibia, L. and Givalois, L. (2006). Continuous i.c.v. infusion of brainderived neurotrophic factor modifies hypothalamic-pituitary-adrenal axis activity, locomotor activity and body temperature rhythms in adult male rats. Neuroscience 139(2): 779-89. Nakai, M., Qin, Z.H., Wang, Y. and Chase, T.N. (1999). Free radical scavenger OPC-14117 attenuates quinolinic acid-induced NF-kappaB activation and apoptosis in rat striatum. Brain Research. Molecular Brain Research 64(1): 59-68. Nakao, N., Brundin, P., Funa, K., Lindvall, O. and Odin, P. (1995). Trophic and protective actions of brain-derived neurotrophic factor on striatal DARPP-32-containing neurons in vitro. Brain Research. Developmental Brain Research 90(1-2): 92-101. Nakao, N., Grasbon-Frodl, E.M., Widner, H. and Brundin, P. (1996). Antioxidant treatment protects striatal neurons against excitotoxic insults. Neuroscience 73(1): 185-200. Nakao, N. and Brundin, P. (1997). Effects of alpha-phenyl-tert-butyl nitrone on neuronal survival and motor function following intrastriatal injections of quinolinate or 3-nitropropionic acid. Neuroscience 76(3): 749-61. Nakao, N. and Itakura, T. (2000). Fetal tissue transplants in animal models of Huntington's disease: the effects on damaged neuronal circuitry and behavioral deficits. Progress in Neurobiology 61(3): 313-38. Natsume, A., Mata, M., Goss, J., Huang, S., Wolfe, D., Oligino, T., Glorioso, J. and Fink, D.J. (2001). Bcl-2 and GDNF delivered by HSV-mediated gene transfer act additively to protect dopaminergic neurons from 6-OHDA-induced degeneration. Experimental Neurology 169(2): 231-8. Newmeyer, D.D. and Ferguson-Miller, S. (2003). Mitochondria: releasing power for life and unleashing the machineries of death. Cell 112(4): 481-90. Nguyen, J.B., Sanchez-Pernaute, R., Cunningham, J. and Bankiewicz, K.S. (2001). Convectionenhanced delivery of AAV-2 combined with heparin increases TK gene transfer in the rat brain. Neuroreport 12(9): 1961-4. Nishino, H., Shimano, Y., Kumazaki, M. and Sakurai, T. (1995). Chronically administered 3nitropropionic acid induces striatal lesions attributed to dysfunction of the blood-brain barrier. Neuroscience Letters 186(2-3): 161-4. Norman, A.B., Norgren, R.B., Wyatt, L.M., Hildebrand, J.P. and Sanberg, P.R. (1992). The direction of apomorphine-induced rotation behavior is dependent on the location of excitotoxin lesions in the rat basal ganglia. Brain Research 569(1): 169-72. 239 Literature References Nucifora, F.C., Jr., Sasaki, M., Peters, M.F., Huang, H., Cooper, J.K., Yamada, M., Takahashi, H., Tsuji, S., Troncoso, J., Dawson, V.L., Dawson, T.M. and Ross, C.A. (2001). Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity. Science 291(5512): 2423-8. Nykjaer, A., Lee, R., Teng, K.K., Jansen, P., Madsen, P., Nielsen, M.S., Jacobsen, C., Kliemannel, M., Schwarz, E., Willnow, T.E., Hempstead, B.L. and Petersen, C.M. (2004). Sortilin is essential for proNGF-induced neuronal cell death. Nature 427(6977): 843-8. Olsson, M., Nikkhah, G., Bentlage, C. and Bjorklund, A. (1995). Forelimb akinesia in the rat Parkinson model: differential effects of dopamine agonists and nigral transplants as assessed by a new stepping test. Journal of Neuroscience 15(5 Pt 2): 3863-75. Oppenheim, R.W. (1991). Cell death during development of the nervous system. Annual Review of Neuroscience 14: 453-501. Orrenius, S., Zhivotovsky, B. and Nicotera, P. (2003). Regulation of cell death: the calciumapoptosis link. Nature Reviews. Molecular Cell Biology 4(7): 552-65. Ouimet, C.C., Miller, P.E., Hemmings, H.C., Jr., Walaas, S.I. and Greengard, P. (1984). DARPP-32, a dopamine- and adenosine 3':5'-monophosphate-regulated phosphoprotein enriched in dopamine-innervated brain regions. III. Immunocytochemical localization. Journal of Neuroscience 4(1): 111-24. Ouimet, C.C., Langley-Gullion, K.C. and Greengard, P. (1998). Quantitative immunocytochemistry of DARPP-32-expressing neurons in the rat caudatoputamen. Brain Research 808(1): 8-12. Palfi, S., Leventhal, L., Chu, Y., Ma, S.Y., Emborg, M., Bakay, R., Deglon, N., Hantraye, P., Aebischer, P. and Kordower, J.H. (2002). Lentivirally delivered glial cell line-derived neurotrophic factor increases the number of striatal dopaminergic neurons in primate models of nigrostriatal degeneration. Journal of Neuroscience 22(12): 4942-54. Pang, P.T., Teng, H.K., Zaitsev, E., Woo, N.T., Sakata, K., Zhen, S., Teng, K.K., Yung, W.H., Hempstead, B.L. and Lu, B. (2004). Cleavage of proBDNF by tPA/plasmin is essential for long-term hippocampal plasticity. Science 306(5695): 487-91. Panov, A.V., Gutekunst, C.A., Leavitt, B.R., Hayden, M.R., Burke, J.R., Strittmatter, W.J. and Greenamyre, J.T. (2002). Early mitochondrial calcium defects in Huntington's disease are a direct effect of polyglutamines. Nature Neuroscience 5(8): 731-6. Paterna, J.C., Moccetti, T., Mura, A., Feldon, J. and Bueler, H. (2000). Influence of promoter and WHV post-transcriptional regulatory element on AAV-mediated transgene expression in the rat brain. Gene Therapy 7(15): 1304-11. Paxinos, G. and Watson, C. (1986). The Rat Brain in Stereotaxic Coordinates. Academic Press. Peel, A.L. and Klein, R.L. (2000). Adeno-associated virus vectors: activity and applications in the CNS. Journal of Neuroscience Methods 98(2): 95-104. Pelleymounter, M.A., Cullen, M.J. and Wellman, C.L. (1995). Characteristics of BDNF-induced weight loss. Experimental Neurology 131(2): 229-38. Penney, J.B., Jr. and Young, A.B. (1986). Striatal inhomogeneities and basal ganglia function. Movement Disorders 1(1): 3-15. Perez-Navarro, E., Alberch, J., Arenas, E., Calvo, N. and Marsal, J. (1994). Nerve growth factor and basic fibroblast growth factor protect cholinergic neurons against quinolinic acid excitotoxicity in rat neostriatum. European Journal of Neuroscience 6(5): 706-11. 240 Literature References Perez-Navarro, E., Arenas, E., Reiriz, J., Calvo, N. and Alberch, J. (1996). Glial cell line-derived neurotrophic factor protects striatal calbindin-immunoreactive neurons from excitotoxic damage. Neuroscience 75(2): 345-52. Perez-Navarro, E., Arenas, E., Marco, S. and Alberch, J. (1999). Intrastriatal grafting of a GDNFproducing cell line protects striatonigral neurons from quinolinic acid excitotoxicity in vivo. European Journal of Neuroscience 11(1): 241-9. Perez-Navarro, E., Akerud, P., Marco, S., Canals, J.M., Tolosa, E., Arenas, E. and Alberch, J. (2000). Neurturin protects striatal projection neurons but not interneurons in a rat model of Huntington's disease. Neuroscience 98(1): 89-96. Perez-Navarro, E., Canudas, A.M., Akerund, P., Alberch, J. and Arenas, E. (2000a). Brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5 prevent the death of striatal projection neurons in a rodent model of Huntington's disease. Journal of Neurochemistry 75(5): 2190-9. Perez-Navarro, E., Gavalda, N., Gratacos, E. and Alberch, J. (2005). Brain-derived neurotrophic factor prevents changes in Bcl-2 family members and caspase-3 activation induced by excitotoxicity in the striatum. Journal of Neurochemistry 92(3): 678-91. Perrelet, D., Ferri, A., MacKenzie, A.E., Smith, G.M., Korneluk, R.G., Liston, P., Sagot, Y., Terrado, J., Monnier, D. and Kato, A.C. (2000). IAP family proteins delay motoneuron cell death in vivo. European Journal of Neuroscience 12(6): 2059-67. Perrelet, D., Ferri, A., Liston, P., Muzzin, P., Korneluk, R.G. and Kato, A.C. (2002). IAPs are essential for GDNF-mediated neuroprotective effects in injured motor neurons in vivo. Nature Cell Biology 4(2): 175-9. Petrin, D., Baker, A., Coupland, S.G., Liston, P., Narang, M., Damji, K., Leonard, B., Chiodo, V.A., Timmers, A., Hauswirth, W., Korneluk, R.G. and Tsilfidis, C. (2003a). Structural and functional protection of photoreceptors from MNU-induced retinal degeneration by the Xlinked inhibitor of apoptosis. Investigative Ophthalmology & Visual Science 44(6): 2757-63. Petrin, D., Baker, A., Brousseau, J., Coupland, S., Liston, P., Hauswirth, W.W., Komeluk, R.G. and Tsilfidis, C. (2003b). XIAP protects photoreceptors from n-methyl-n-nitrosourea-induced retinal degeneration. Advances in Experimental Medicine and Biology 533: 385-93. Pisa, M., Sanberg, P.R. and Fibiger, H.C. (1981). Striatal injections of kainic acid selectively impair serial memory performance in the rat. Experimental Neurology 74(3): 633-53. Pochon, N.A., Menoud, A., Tseng, J.L., Zurn, A.D. and Aebischer, P. (1997). Neuronal GDNF expression in the adult rat nervous system identified by in situ hybridization. European Journal of Neuroscience 9(3): 463-71. Poeschla, E.M. (2003). Non-primate lentiviral vectors. Current Opinion in Molecular Therapeutics 5(5): 529-40. Ponnazhagan, S., Erikson, D., Kearns, W.G., Zhou, S.Z., Nahreini, P., Wang, X.S. and Srivastava, A. (1997). Lack of site-specific integration of the recombinant adeno-associated virus 2 genomes in human cells. Human Gene Therapy 8(3): 275-84. Popoli, P., Pezzola, A., Domenici, M.R., Sagratella, S., Diana, G., Caporali, M.G., Bronzetti, E., Vega, J. and Scotti de Carolis, A. (1994). Behavioral and electrophysiological correlates of the quinolinic acid rat model of Huntington's disease in rats. Brain Research Bulletin 35(4): 32935. 241 Literature References Popovic, N., Maingay, M., Kirik, D. and Brundin, P. (2005). Lentiviral gene delivery of GDNF into the striatum of R6/2 Huntington mice fails to attenuate behavioral and neuropathological changes. Experimental Neurology 193(1): 65-74. Portera-Cailliau, C., Hedreen, J.C., Price, D.L. and Koliatsos, V.E. (1995). Evidence for apoptotic cell death in Huntington disease and excitotoxic animal models. Journal of Neuroscience 15(5 Pt 2): 3775-87. Prakash, N. and Wurst, W. (2006). Development of dopaminergic neurons in the mammalian brain. Cellular & Molecular Life Sciences 63(2): 187-206. Prosch, S., Stein, J., Staak, K., Liebenthal, C., Volk, H.D. and Kruger, D.H. (1996). Inactivation of the very strong HCMV immediate early promoter by DNA CpG methylation in vitro. Biological Chemistry Hoppe Seyler 377(3): 195-201. Purves, D. (1988). Body and Brain. A Trophic Theory of Neural Connections. Harvard University Press, Cambridge, MA. Qin, Y., Soghomonian, J.J. and Chesselet, M.F. (1992). Effects of quinolinic acid on messenger RNAs encoding somatostatin and glutamic acid decarboxylases in the striatum of adult rats. Experimental Neurology 115(2): 200-11. Qin, Z.H., Wang, Y. and Chase, T.N. (1996). Stimulation of N-methyl-D-aspartate receptors induces apoptosis in rat brain. Brain Research 725(2): 166-76. Qin, Z.H., Wang, J. and Gu, Z.L. (2005). Development of novel therapies for Huntington's disease: hope and challenge. Acta Pharmacologica Sinica 26(2): 129-42. Rabinowitz, J.E., Bowles, D.E., Faust, S.M., Ledford, J.G., Cunningham, S.E. and Samulski, R.J. (2004). Cross-dressing the virion: the transcapsidation of adeno-associated virus serotypes functionally defines subgroups. Journal of Virology 78(9): 4421-32. Reddy, P.H., Williams, M., Charles, V., Garrett, L., Pike-Buchanan, L., Whetsell, W.O., Jr., Miller, G. and Tagle, D.A. (1998). Behavioural abnormalities and selective neuronal loss in HD transgenic mice expressing mutated full-length HD cDNA. Nature Genetics 20(2): 198-202. Reiner, A., Albin, R.L., Anderson, K.D., D'Amato, C.J., Penney, J.B. and Young, A.B. (1988). Differential loss of striatal projection neurons in Huntington disease. Proceedings of the National Academy of Sciences of the United States of America 85(15): 5733-7. Reiner, A. (2004). Can lesions of GPe correct HD deficits? Experimental Neurology 186(1): 1-5. Reis, D.J., Gilad, G., Pickel, V.M. and Joh, T.H. (1978). Reversible changes in the activities and amounts of tyrosine hydroxylase in dopamine neurons of the substantia nigra in response to axonal injury as studied by immunochemical and immunocytochemical methods. Brain Research 144(2): 325-42. Richichi, C., Lin, E.J., Stefanin, D., Colella, D., Ravizza, T., Grignaschi, G., Veglianese, P., Sperk, G., During, M.J. and Vezzani, A. (2004). Anticonvulsant and antiepileptogenic effects mediated by adeno-associated virus vector neuropeptide Y expression in the rat hippocampus. Journal of Neuroscience 24(12): 3051-9. Rigamonti, D., Bauer, J.H., De-Fraja, C., Conti, L., Sipione, S., Sciorati, C., Clementi, E., Hackam, A., Hayden, M.R., Li, Y., Cooper, J.K., Ross, C.A., Govoni, S., Vincenz, C. and Cattaneo, E. (2000). Wild-type huntingtin protects from apoptosis upstream of caspase-3. Journal of Neuroscience 20(10): 3705-13. 242 Literature References Rigamonti, D., Sipione, S., Goffredo, D., Zuccato, C., Fossale, E. and Cattaneo, E. (2001). Huntingtin's neuroprotective activity occurs via inhibition of procaspase-9 processing. Journal of Biological Chemistry 276(18): 14545-8. Ringstedt, T., Lagercrantz, H. and Persson, H. (1993). Expression of members of the trk family in the developing postnatal rat brain. Brain Research. Developmental Brain Research 72(1): 11931. Robertson, G.S., Crocker, S.J., Nicholson, D.W. and Schulz, J.B. (2000). Neuroprotection by the inhibition of apoptosis. Brain Pathology 10(2): 283-92. Rosenblad, C., Kirik, D. and Bjorklund, A. (2000). Sequential administration of GDNF into the substantia nigra and striatum promotes dopamine neuron survival and axonal sprouting but not striatal reinnervation or functional recovery in the partial 6-OHDA lesion model. Experimental Neurology 161(2): 503-16. Rubio, N. (1997). Mouse astrocytes store and deliver brain-derived neurotrophic factor using the non-catalytic gp95trkB receptor. European Journal of Neuroscience 9(9): 1847-53. Rudge, J.S., Li, Y., Pasnikowski, E.M., Mattsson, K., Pan, L., Yancopoulos, G.D., Wiegand, S.J., Lindsay, R.M. and Ip, N.Y. (1994). Neurotrophic factor receptors and their signal transduction capabilities in rat astrocytes. European Journal of Neuroscience 6(5): 693-705. Rudge, J.S., Pasnikowski, E.M., Holst, P. and Lindsay, R.M. (1995). Changes in neurotrophic factor expression and receptor activation following exposure of hippocampal neuron/astrocyte cocultures to kainic acid. Journal of Neuroscience 15(10): 6856-67. Ryu, J.K., Kim, S.U. and McLarnon, J.G. (2003). Neuroprotective effects of pyruvate in the quinolinic acid rat model of Huntington's disease. Experimental Neurology 183(2): 700-4. Ryu, J.K., Choi, H.B. and McLarnon, J.G. (2006). Combined minocycline plus pyruvate treatment enhances effects of each agent to inhibit inflammation, oxidative damage, and neuronal loss in an excitotoxic animal model of Huntington's disease. Neuroscience 141(4): 1835-48. Saelens, X., Festjens, N., Vande Walle, L., van Gurp, M., van Loo, G. and Vandenabeele, P. (2004). Toxic proteins released from mitochondria in cell death. Oncogene 23(16): 2861-74. Sakamoto, T., Kawazoe, Y., Shen, J.S., Takeda, Y., Arakawa, Y., Ogawa, J., Oyanagi, K., Ohashi, T., Watanabe, K., Inoue, K., Eto, Y. and Watabe, K. (2003). Adenoviral gene transfer of GDNF, BDNF and TGF beta 2, but not CNTF, cardiotrophin-1 or IGF1, protects injured adult motoneurons after facial nerve avulsion. Journal of Neuroscience Methods 72(1): 54-64. Salzberg-Brenhouse, H.C., Chen, E.Y., Emerich, D.F., Baldwin, S., Hogeland, K., Ranelli, S., Lafreniere, D., Perdomo, B., Novak, L., Kladis, T., Fu, K., Basile, A.S., Kordower, J.H. and Bartus, R.T. (2003). Inhibitors of cyclooxygenase-2, but not cyclooxygenase-1 provide structural and functional protection against quinolinic acid-induced neurodegeneration. Journal of Pharmacology and Experimental Therapeutics 306(1): 218-28. Samaniego, L.A., Wu, N. and DeLuca, N.A. (1997). The herpes simplex virus immediate-early protein ICP0 affects transcription from the viral genome and infected-cell survival in the absence of ICP4 and ICP27. Journal of Virology 71(6): 4614-25. Samulski, R.J., Berns, K.I., Tan, M. and Muzyczka, N. (1982). Cloning of adeno-associated virus into pBR322: rescue of intact virus from the recombinant plasmid in human cells. Proceedings of the National Academy of Sciences of the United States of America 79(6): 2077-81. Sapp, E., Ge, P., Aizawa, H., Bird, E., Penney, J., Young, A.B., Vonsattel, J.P. and DiFiglia, M. (1995). Evidence for a preferential loss of enkephalin immunoreactivity in the external globus 243 Literature References pallidus in low grade Huntington's disease using high resolution image analysis. Neuroscience 64(2): 397-404. Sastry, P.S. and Rao, K.S. (2000). Apoptosis and the nervous system. Journal of Neurochemistry 74(1): 1-20. Saudou, F., Finkbeiner, S., Devys, D. and Greenberg, M.E. (1998). Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell 95(1): 55-66. Sauer, H. and Oertel, W.H. (1994). Progressive degeneration of nigrostriatal dopamine neurons following intrastriatal terminal lesions with 6-hydroxydopamine: a combined retrograde tracing and immunocytochemical study in the rat. Neuroscience 59(2): 401-15. Sawa, A., Wiegand, G.W., Cooper, J., Margolis, R.L., Sharp, A.H., Lawler, J.F., Jr., Greenamyre, J.T., Snyder, S.H. and Ross, C.A. (1999). Increased apoptosis of Huntington disease lymphoblasts associated with repeat length-dependent mitochondrial depolarization. Nature Medicine 5(10): 1194-8. Sawa, A., Tomoda, T. and Bae, B.I. (2003). Mechanisms of neuronal cell death in Huntington's disease. Cytogenetic and Genome Research 100(1-4): 287-95. Scattoni, M.L., Valanzano, A., Popoli, P., Pezzola, A., Reggio, R. and Calamandrei, G. (2004). Progressive behavioural changes in the spatial open-field in the quinolinic acid rat model of Huntington's disease. Behavioural Brain Research 152(2): 375-83. Schaar, D.G., Sieber, B.A., Dreyfus, C.F. and Black, I.B. (1993). Regional and cell-specific expression of GDNF in rat brain. Experimental Neurology 124(2): 368-71. Schabitz, W.R., Sommer, C., Zoder, W., Kiessling, M., Schwaninger, M. and Schwab, S. (2000). Intravenous brain-derived neurotrophic factor reduces infarct size and counterregulates Bax and Bcl-2 expression after temporary focal cerebral ischemia. Stroke 31(9): 2212-7. Schallert, T., Kozlowski, D.A., Humm, J.L. and Cocke, R.R. (1997). Use-dependent structural events in recovery of function. Advances in Neurology 73: 229-38. Schallert, T., Fleming, S.M., Leasure, J.L., Tillerson, J.L. and Bland, S.T. (2000). CNS plasticity and assessment of forelimb sensorimotor outcome in unilateral rat models of stroke, cortical ablation, parkinsonism and spinal cord injury. Neuropharmacology 39(5): 777-87. Scharfman, H.E. (1997). Hyperexcitability in combined entorhinal/hippocampal slices of adult rat after exposure to brain-derived neurotrophic factor. Journal of Neurophysiology 78(2): 108295. Schilling, G., Becher, M.W., Sharp, A.H., Jinnah, H.A., Duan, K., Kotzuk, J.A., Slunt, H.H., Ratovitski, T., Cooper, J.K., Jenkins, N.A., Copeland, N.G., Price, D.L., Ross, C.A. and Borchelt, D.R. (1999). Intranuclear inclusions and neuritic aggregates in transgenic mice expressing a mutant N-terminal fragment of huntingtin. Human Molecular Genetics 8(3): 397407. Schnepp, B.C., Jensen, R.L., Chen, C.L., Johnson, P.R. and Clark, K.R. (2005). Characterization of adeno-associated virus genomes isolated from human tissues. Journal of Virology 79(23): 14793-803. Schumacher, J.M., Short, M.P., Hyman, B.T., Breakefield, X.O. and Isacson, O. (1991). Intracerebral implantation of nerve growth factor-producing fibroblasts protects striatum against neurotoxic levels of excitatory amino acids. Neuroscience 45(3): 561-570. 244 Literature References Schwarcz, R., Fuxe, K., Agnati, L.F., Hokfelt, T. and Coyle, J.T. (1979). Rotational behaviour in rats with unilateral striatal kainic acid lesions: a behavioural model for studies on intact dopamine receptors. Brain Research 170(3): 485-95. Schwarcz, R. and Kohler, C. (1983). Differential vulnerability of central neurons of the rat to quinolinic acid. Neuroscience Letters 38(1): 85-90. Schwarcz, R., Foster, A.C., French, E.D., Whetsell, W.O., Jr. and Kohler, C. (1984). Excitotoxic models for neurodegenerative disorders. Life Sciences 35(1): 19-32. Schwarcz, R., Tamminga, C.A., Kurlan, R. and Shoulson, I. (1988). Cerebrospinal fluid levels of quinolinic acid in Huntington's disease and schizophrenia. Annals of Neurology 24(4): 580-2. Selemon, L.D. and Goldman-Rakic, P.S. (1985). Longitudinal topography and interdigitation of corticostriatal projections in the rhesus monkey. Journal of Neuroscience 5(3): 776-94. Shear, D.A., Dong, J., Gundy, C.D., Haik-Creguer, K.L. and Dunbar, G.L. (1998). Comparison of intrastriatal injections of quinolinic acid and 3-nitropropionic acid for use in animal models of Huntington's disease. Progress in Neuropsychopharmacology & Biological Psychiatry 22(7): 1217-40. Shelbourne, P.F., Killeen, N., Hevner, R.F., Johnston, H.M., Tecott, L., Lewandoski, M., Ennis, M., Ramirez, L., Li, Z., Iannicola, C., Littman, D.R. and Myers, R.M. (1999). A Huntington's disease CAG expansion at the murine Hdh locus is unstable and associated with behavioural abnormalities in mice. Human Molecular Genetics 8(5): 763-74. Shimazaki, K., Urabe, M., Monahan, J., Ozawa, K. and Kawai, N. (2000). Adeno-associated virus vector-mediated bcl-2 gene transfer into post-ischemic gerbil brain in vivo: prospects for gene therapy of ischemia-induced neuronal death. Gene Therapy 7(14): 1244-9. Shirao, T., Evinger, M.J., Iacovitti, L. and Reis, D.J. (1992). Lesions of nigrostriatal pathway reduce expression of tyrosine hydroxylase gene in residual dopaminergic neurons of substantia nigra. Neuroscience Letters 141(2): 208-12. Simons, M., Beinroth, S., Gleichmann, M., Liston, P., Korneluk, R.G., MacKenzie, A.E., Bahr, M., Klockgether, T., Robertson, G.S., Weller, M. and Schulz, J.B. (1999). Adenovirus-mediated gene transfer of inhibitors of apoptosis protein delays apoptosis in cerebellar granule neurons. Journal of Neurochemistry 72(1): 292-301. Sleeman, M.W., Anderson, K.D., Lambert, P.D., Yancopoulos, G.D. and Wiegand, S.J. (2000). The ciliary neurotrophic factor and its receptor, CNTFR alpha. Pharmaceutica Acta Helvetiae 74(2-3): 265-72. Smith, Y., Bevan, M.D., Shink, E. and Bolam, J.P. (1998). Microcircuitry of the direct and indirect pathways of the basal ganglia. Neuroscience 86(2): 353-87. Sofroniew, M.V., Howe, C.L. and Mobley, W.C. (2001). Nerve growth factor signaling, neuroprotection, and neural repair. Annual Review of Neuroscience 24: 1217-81. Soule, J., Messaoudi, E. and Bramham, C.R. (2006). Brain-derived neurotrophic factor and control of synaptic consolidation in the adult brain. Biochemical Society Transactions 34(Pt 4): 6004. Springer, J.E., Mu, X., Bergmann, L.W. and Trojanowski, J.Q. (1994). Expression of GDNF mRNA in rat and human nervous tissue. Experimental Neurology 127(2): 167-70. Srinivasula, S.M., Gupta, S., Datta, P., Zhang, Z., Hegde, R., Cheong, N., Fernandes-Alnemri, T. and Alnemri, E.S. (2003). Inhibitor of apoptosis proteins are substrates for the mitochondrial serine protease Omi/HtrA2. Journal of Biological Chemistry 278(34): 31469-72. 245 Literature References Srivastava, A., Lusby, E.W. and Berns, K.I. (1983). Nucleotide sequence and organization of the adeno-associated virus 2 genome. Journal of Virology 45(2): 555-64. Steffan, J.S., Kazantsev, A., Spasic-Boskovic, O., Greenwald, M., Zhu, Y.Z., Gohler, H., Wanker, E.E., Bates, G.P., Housman, D.E. and Thompson, L.M. (2000). The Huntington's disease protein interacts with p53 and CREB-binding protein and represses transcription. Proceedings of the National Academy of Sciences of the United States of America 97(12): 6763-8. Steininger, T.L., Wainer, B.H., Klein, R., Barbacid, M. and Palfrey, H.C. (1993). High-affinity nerve growth factor receptor (Trk) immunoreactivity is localized in cholinergic neurons of the basal forebrain and striatum in the adult rat brain. Brain Research 612(1-2): 330-5. Strauss, S., Otten, U., Joggerst, B., Pluss, K. and Volk, B. (1994). Increased levels of nerve growth factor (NGF) protein and mRNA and reactive gliosis following kainic acid injection into the rat striatum. Neuroscience Letters 168(1-2): 193-6. Stromberg, I., Bjorklund, L., Johansson, M., Tomac, A., Collins, F., Olson, L., Hoffer, B. and Humpel, C. (1993). Glial cell line-derived neurotrophic factor is expressed in the developing but not adult striatum and stimulates developing dopamine neurons in vivo. Experimental Neurology 124(2): 401-12. Summerford, C. and Samulski, R.J. (1998). Membrane-associated heparan sulfate proteoglycan is a receptor for adeno-associated virus type 2 virions. Journal of Virology 72(2): 1438-45. Sun, Y., Savanenin, A., Reddy, P.H. and Liu, Y.F. (2001). Polyglutamine-expanded huntingtin promotes sensitization of N-methyl-D-aspartate receptors via post-synaptic density 95. Journal of Biological Chemistry 276(27): 24713-8. Sun, Z., Xie, J. and Reiner, A. (2002). The differential vulnerability of striatal projection neurons in 3-nitropropionic acid-treated rats does not match that typical of adult-onset Huntington's disease. Experimental Neurology 176(1): 55-65. Takahashi, R., Yokoji, H., Misawa, H., Hayashi, M., Hu, J. and Deguchi, T. (1994). A null mutation in the human CNTF gene is not causally related to neurological diseases. [see comments.] [erratum appears in Nat Genet 1994 Jun;7(2):215.]. Nature Genetics 7(1): 79-84. Tan, S.A. and Aebischer, P. (1996). The problems of delivering neuroactive molecules to the CNS. Ciba Foundation Symposium 196: 211-36; discussion 236-9. Tang, T.S., Slow, E., Lupu, V., Stavrovskaya, I.G., Sugimori, M., Llinas, R., Kristal, B.S., Hayden, M.R. and Bezprozvanny, I. (2005). Disturbed Ca2+ signaling and apoptosis of medium spiny neurons in Huntington's disease. Proceedings of the National Academy of Sciences of the United States of America 102(7): 2602-7. Tattersfield, A.S., Croon, R.J., Liu, Y.W., Kells, A.P., Faull, R.L. and Connor, B. (2004). Neurogenesis in the striatum of the quinolinic acid lesion model of Huntington's disease. Neuroscience 127(2): 319-32. Temel, Y., Cao, C., Vlamings, R., Blokland, A., Ozen, H., Steinbusch, H.W., Michelsen, K.A., von Horsten, S., Schmitz, C. and Visser-Vandewalle, V. (2006). Motor and cognitive improvement by deep brain stimulation in a transgenic rat model of Huntington's disease. Neuroscience Letters 406(1-2): 138-41. Teng, H.K., Teng, K.K., Lee, R., Wright, S., Tevar, S., Almeida, R.D., Kermani, P., Torkin, R., Chen, Z.Y., Lee, F.S., Kraemer, R.T., Nykjaer, A. and Hempstead, B.L. (2005). ProBDNF induces neuronal apoptosis via activation of a receptor complex of p75NTR and sortilin. Journal of Neuroscience 25(22): 5455-63. 246 Literature References Testa, C.M., Standaert, D.G., Landwehrmeyer, G.B., Penney, J.B., Jr. and Young, A.B. (1995). Differential expression of mGluR5 metabotropic glutamate receptor mRNA by rat striatal neurons. Journal of Comparative Neurology 354(2): 241-52. Thomas, C.E., Schiedner, G., Kochanek, S., Castro, M.G. and Lowenstein, P.R. (2000). Peripheral infection with adenovirus causes unexpected long-term brain inflammation in animals injected intracranially with first-generation, but not with high-capacity, adenovirus vectors: toward realistic long-term neurological gene therapy for chronic diseases. Proceedings of the National Academy of Sciences of the United States of America 97(13): 7482-7. Thomas, C.E., Ehrhardt, A. and Kay, M.A. (2003). Progress and problems with the use of viral vectors for gene therapy. Nature Reviews Genetics 4(5): 346-58. Thomas, L.B., Gates, D.J., Richfield, E.K., O'Brien, T.F., Schweitzer, J.B. and Steindler, D.A. (1995). DNA end labeling (TUNEL) in Huntington's disease and other neuropathological conditions. Experimental Neurology 133(2): 265-72. Timpe, J., Bevington, J., Casper, J., Dignam, J.D. and Trempe, J.P. (2005). Mechanisms of adenoassociated virus genome encapsidation. Current Gene Therapy 5(3): 273-84. Tobin, A.J. and Signer, E.R. (2000). Huntington's disease: the challenge for cell biologists. Trends in Cell Biology 10(12): 531-6. Tomac, A., Widenfalk, J., Lin, L.F., Kohno, T., Ebendal, T., Hoffer, B.J. and Olson, L. (1995). Retrograde axonal transport of glial cell line-derived neurotrophic factor in the adult nigrostriatal system suggests a trophic role in the adult. Proceedings of the National Academy of Sciences of the United States of America 92(18): 8274-8. Tongiorgi, E., Righi, M. and Cattaneo, A. (1997). Activity-dependent dendritic targeting of BDNF and TrkB mRNAs in hippocampal neurons. Journal of Neuroscience 17(24): 9492-505. Treanor, J.J., Goodman, L., de Sauvage, F., Stone, D.M., Poulsen, K.T., Beck, C.D., Gray, C., Armanini, M.P., Pollock, R.A., Hefti, F., Phillips, H.S., Goddard, A., Moore, M.W., Buj-Bello, A., Davies, A.M., Asai, N., Takahashi, M., Vandlen, R., Henderson, C.E. and Rosenthal, A. (1996). Characterization of a multicomponent receptor for GDNF. Nature 382(6586): 80-3. Trottier, Y., Devys, D., Imbert, G., Saudou, F., An, I., Lutz, Y., Weber, C., Agid, Y., Hirsch, E.C. and Mandel, J.L. (1995). Cellular localization of the Huntington's disease protein and discrimination of the normal and mutated form. [see comments.]. Nature Genetics 10(1): 10410. Troy, C.M., Friedman, J.E. and Friedman, W.J. (2002). Mechanisms of p75-mediated death of hippocampal neurons. Role of caspases. Journal of Biological Chemistry 277(37): 34295-302. Trupp, M., Belluardo, N., Funakoshi, H. and Ibanez, C.F. (1997). Complementary and overlapping expression of glial cell line-derived neurotrophic factor (GDNF), c-ret proto-oncogene, and GDNF receptor-alpha indicates multiple mechanisms of trophic actions in the adult rat CNS. Journal of Neuroscience 17(10): 3554-67. Ungerstedt, U. (1971). Striatal dopamine release after amphetamine or nerve degeneration revealed by rotational behaviour. Acta Physiologica Scandinavica. Supplementum 367: 49-68. Vaillant, A.R., Mazzoni, I., Tudan, C., Boudreau, M., Kaplan, D.R. and Miller, F.D. (1999). Depolarization and neurotrophins converge on the phosphatidylinositol 3-kinase-Akt pathway to synergistically regulate neuronal survival. Journal of Cell Biology 146(5): 955-66. Valjent, E., Pascoli, V., Svenningsson, P., Paul, S., Enslen, H., Corvol, J.C., Stipanovich, A., Caboche, J., Lombroso, P.J., Nairn, A.C., Greengard, P., Herve, D. and Girault, J.A. (2005). Regulation of a protein phosphatase cascade allows convergent dopamine and glutamate 247 Literature References signals to activate ERK in the striatum. Proceedings of the National Academy of Sciences of the United States of America 102(2): 491-6. Van Ooyen, A. (2005). Competition in neurite outgrowth and the development of nerve connections. Progress in Brain Research 147: 81-99. Venero, J.L., Beck, K.D. and Hefti, F. (1994). Intrastriatal infusion of nerve growth factor after quinolinic acid prevents reduction of cellular expression of choline acetyltransferase messenger RNA and trkA messenger RNA, but not glutamate decarboxylase messenger RNA. Neuroscience 61(2): 257-68. Ventimiglia, R., Mather, P.E., Jones, B.E. and Lindsay, R.M. (1995). The neurotrophins BDNF, NT3 and NT-4/5 promote survival and morphological and biochemical differentiation of striatal neurons in vitro. European Journal of Neuroscience 7(2): 213-22. Vis, J.C., Schipper, E., de Boer-van Huizen, R.T., Verbeek, M.M., de Waal, R.M., Wesseling, P., ten Donkelaar, H.J. and Kremer, B. (2005). Expression pattern of apoptosis-related markers in Huntington's disease. Acta Neuropathologica 109(3): 321-8. von Horsten, S., Schmitt, I., Nguyen, H.P., Holzmann, C., Schmidt, T., Walther, T., Bader, M., Pabst, R., Kobbe, P., Krotova, J., Stiller, D., Kask, A., Vaarmann, A., Rathke-Hartlieb, S., Schulz, J.B., Grasshoff, U., Bauer, I., Vieira-Saecker, A.M., Paul, M., Jones, L., Lindenberg, K.S., Landwehrmeyer, B., Bauer, A., Li, X.J. and Riess, O. (2003). Transgenic rat model of Huntington's disease. Human Molecular Genetics 12(6): 617-24. Vonsattel, J.P., Myers, R.H., Stevens, T.J., Ferrante, R.J., Bird, E.D. and Richardson, E.P., Jr. (1985). Neuropathological classification of Huntington's disease. Journal of Neuropathology & Experimental Neurology 44(6): 559-77. Vonsattel, J.P. and DiFiglia, M. (1998). Huntington disease. Journal of Neuropathology & Experimental Neurology 57(5): 369-84. Waldvogel, H.J., Faull, R.L., Williams, M.N. and Dragunow, M. (1991). Differential sensitivity of calbindin and parvalbumin immunoreactive cells in the striatum to excitotoxins. Brain Research 546(2): 329-35. Wang, G.H., Mitsui, K., Kotliarova, S., Yamashita, A., Nagao, Y., Tokuhiro, S., Iwatsubo, T., Kanazawa, I. and Nukina, N. (1999). Caspase activation during apoptotic cell death induced by expanded polyglutamine in N2a cells. Neuroreport 10(12): 2435-8. Watabe, K., Ohashi, T., Sakamoto, T., Kawazoe, Y., Takeshima, T., Oyanagi, K., Inoue, K., Eto, Y. and Kim, S.U. (2000). Rescue of lesioned adult rat spinal motoneurons by adenoviral gene transfer of glial cell line-derived neurotrophic factor. Journal of Neuroscience Research 60(4): 511-9. Watson, D.J., Kobinger, G.P., Passini, M.A., Wilson, J.M. and Wolfe, J.H. (2002). Targeted transduction patterns in the mouse brain by lentivirus vectors pseudotyped with VSV, Ebola, Mokola, LCMV, or MuLV envelope proteins. Molecular Therapy 5(5 Pt 1): 528-37. Watson, F.L., Heerssen, H.M., Moheban, D.B., Lin, M.Z., Sauvageot, C.M., Bhattacharyya, A., Pomeroy, S.L. and Segal, R.A. (1999). Rapid nuclear responses to target-derived neurotrophins require retrograde transport of ligand-receptor complex. Journal of Neuroscience 19(18): 7889-900. Wellington, C.L., Ellerby, L.M., Gutekunst, C.A., Rogers, D., Warby, S., Graham, R.K., Loubser, O., van Raamsdonk, J., Singaraja, R., Yang, Y.Z., Gafni, J., Bredesen, D., Hersch, S.M., Leavitt, B.R., Roy, S., Nicholson, D.W. and Hayden, M.R. (2002). Caspase cleavage of mutant 248 Literature References huntingtin precedes neurodegeneration in Huntington's disease. Journal of Neuroscience 22(18): 7862-72. Wheeler, V.C., White, J.K., Gutekunst, C.A., Vrbanac, V., Weaver, M., Li, X.J., Li, S.H., Yi, H., Vonsattel, J.P., Gusella, J.F., Hersch, S., Auerbach, W., Joyner, A.L. and MacDonald, M.E. (2000). Long glutamine tracts cause nuclear localization of a novel form of huntingtin in medium spiny striatal neurons in HdhQ92 and HdhQ111 knock-in mice. Human Molecular Genetics 9(4): 503-13. Widmer, H.R. and Hefti, F. (1994). Stimulation of GABAergic neuron differentiation by NT-4/5 in cultures of rat cerebral cortex. Brain Research. Developmental Brain Research 80(1-2): 279-84. Widmer, H.R., Schaller, B., Meyer, M. and Seiler, R.W. (2000). Glial cell line-derived neurotrophic factor stimulates the morphological differentiation of cultured ventral mesencephalic calbindinand calretinin-expressing neurons. Experimental Neurology 164(1): 71-81. Winkler, C., Gil, J.M., Araujo, I.M., Riess, O., Skripuletz, T., von Horsten, S. and Petersen, A. (2006). Normal sensitivity to excitotoxicity in a transgenic Huntington's disease rat. Brain Research Bulletin 69(3): 306-10. Wiznerowicz, M. and Trono, D. (2005). Harnessing HIV for therapy, basic research and biotechnology. Trends in Biotechnology 23(1): 42-7. Wong, L.F., Azzouz, M., Walmsley, L.E., Askham, Z., Wilkes, F.J., Mitrophanous, K.A., Kingsman, S.M. and Mazarakis, N.D. (2004). Transduction patterns of pseudotyped lentiviral vectors in the nervous system. Molecular Therapy 9(1): 101-11. Wong, L.F., Ralph, G.S., Walmsley, L.E., Bienemann, A.S., Parham, S., Kingsman, S.M., Uney, J.B. and Mazarakis, N.D. (2005). Lentiviral-mediated delivery of Bcl-2 or GDNF protects against excitotoxicity in the rat hippocampus. Molecular Therapy 11(1): 89-95. Wong, L.F., Goodhead, L., Prat, C., Mitrophanous, K.A., Kingsman, S.M. and Mazarakis, N.D. (2006). Lentivirus-mediated gene transfer to the central nervous system: therapeutic and research applications. Human Gene Therapy 17(1): 1-9. Wu, Z., Asokan, A. and Samulski, R.J. (2006). Adeno-associated virus serotypes: vector toolkit for human gene therapy. Molecular Therapy 14(3): 316-27. Xiao, X., Li, J. and Samulski, R.J. (1998). Production of high-titer recombinant adeno-associated virus vectors in the absence of helper adenovirus. Journal of Virology 72(3): 2224-32. Xu, D., Bureau, Y., McIntyre, D.C., Nicholson, D.W., Liston, P., Zhu, Y., Fong, W.G., Crocker, S.J., Korneluk, R.G. and Robertson, G.S. (1999). Attenuation of ischemia-induced cellular and behavioral deficits by X chromosome-linked inhibitor of apoptosis protein overexpression in the rat hippocampus. Journal of Neuroscience 19(12): 5026-33. Xu, D.G., Crocker, S.J., Doucet, J.P., St-Jean, M., Tamai, K., Hakim, A.M., Ikeda, J.E., Liston, P., Thompson, C.S., Korneluk, R.G., MacKenzie, A. and Robertson, G.S. (1997). Elevation of neuronal expression of NAIP reduces ischemic damage in the rat hippocampus. Nature Medicine 3(9): 997-1004. Xu, L., Daly, T., Gao, C., Flotte, T.R., Song, S., Byrne, B.J., Sands, M.S. and Parker Ponder, K. (2001). CMV-beta-actin promoter directs higher expression from an adeno-associated viral vector in the liver than the cytomegalovirus or elongation factor 1 alpha promoter and results in therapeutic levels of human factor X in mice. Human Gene Therapy 12(5): 563-73. Xu, R., Janson, C.G., Mastakov, M., Lawlor, P., Young, D., Mouravlev, A., Fitzsimons, H., Choi, K.L., Ma, H., Dragunow, M., Leone, P., Chen, Q., Dicker, B. and During, M.J. (2001). 249 Literature References Quantitative comparison of expression with adeno-associated virus (AAV-2) brain-specific gene cassettes. Gene Therapy 8(17): 1323-32. Yamada, M., Oligino, T., Mata, M., Goss, J.R., Glorioso, J.C. and Fink, D.J. (1999). Herpes simplex virus vector-mediated expression of Bcl-2 prevents 6-hydroxydopamine-induced degeneration of neurons in the substantia nigra in vivo. Proceedings of the National Academy of Sciences of the United States of America 96(7): 4078-83. Yan, Q., Matheson, C., Sun, J., Radeke, M.J., Feinstein, S.C. and Miller, J.A. (1994). Distribution of intracerebral ventricularly administered neurotrophins in rat brain and its correlation with trk receptor expression. Experimental Neurology 127(1): 23-36. Yang, C.C., Xiao, X., Zhu, X., Ansardi, D.C., Epstein, N.D., Frey, M.R., Matera, A.G. and Samulski, R.J. (1997). Cellular recombination pathways and viral terminal repeat hairpin structures are sufficient for adeno-associated virus integration in vivo and in vitro. Journal of Virology 71(12): 9231-47. Young, K.M., Merson, T.D., Sotthibundhu, A., Coulson, E.J. and Bartlett, P.F. (2007). p75 neurotrophin receptor expression defines a population of BDNF-responsive neurogenic precursor cells. Journal of Neuroscience 27(19): 5146-55. Yu, T., Scully, S., Yu, Y., Fox, G.M., Jing, S. and Zhou, R. (1998). Expression of GDNF family receptor components during development: implications in the mechanisms of interaction. Journal of Neuroscience 18(12): 4684-96. Zeron, M.M., Chen, N., Moshaver, A., Lee, A.T., Wellington, C.L., Hayden, M.R. and Raymond, L.A. (2001). Mutant huntingtin enhances excitotoxic cell death. Molecular and Cellular Neurosciences 17(1): 41-53. Zeron, M.M., Hansson, O., Chen, N., Wellington, C.L., Leavitt, B.R., Brundin, P., Hayden, M.R. and Raymond, L.A. (2002). Increased sensitivity to N-methyl-D-aspartate receptor-mediated excitotoxicity in a mouse model of Huntington's disease. Neuron 33(6): 849-60. Zhang, X.D., Gillespie, S.K. and Hersey, P. (2004). Staurosporine induces apoptosis of melanoma by both caspase-dependent and -independent apoptotic pathways. Molecular Cancer Therapeutics 3(2): 187-97. Zigova, T., Pencea, V., Wiegand, S.J. and Luskin, M.B. (1998). Intraventricular administration of BDNF increases the number of newly generated neurons in the adult olfactory bulb. Molecular and Cellular Neurosciences 11(4): 234-45. Zimmermann, K.C., Bonzon, C. and Green, D.R. (2001). The machinery of programmed cell death. Pharmacological & Therapeutics 92(1): 57-70. Zolotukhin, S., Byrne, B.J., Mason, E., Zolotukhin, I., Potter, M., Chesnut, K., Summerford, C., Samulski, R.J. and Muzyczka, N. (1999). Recombinant adeno-associated virus purification using novel methods improves infectious titer and yield. Gene Therapy 6(6): 973-85. Zoltick, P.W., Chirmule, N., Schnell, M.A., Gao, G.P., Hughes, J.V. and Wilson, J.M. (2001). Biology of E1-deleted adenovirus vectors in nonhuman primate muscle. Journal of Virology 75(11): 5222-9. Zuccato, C., Ciammola, A., Rigamonti, D., Leavitt, B.R., Goffredo, D., Conti, L., MacDonald, M.E., Friedlander, R.M., Silani, V., Hayden, M.R., Timmusk, T., Sipione, S. and Cattaneo, E. (2001). Loss of huntingtin-mediated BDNF gene transcription in Huntington's disease. Science 293(5529): 493-8. Zuccato, C., Tartari, M., Crotti, A., Goffredo, D., Valenza, M., Conti, L., Cataudella, T., Leavitt, B.R., Hayden, M.R., Timmusk, T., Rigamonti, D. and Cattaneo, E. (2003). Huntingtin interacts 250 Literature References with REST/NRSF to modulate the transcription of NRSE-controlled neuronal genes. Nature Genetics 35(1): 76-83. Zuccato, C., Liber, D., Ramos, C., Tarditi, A., Rigamonti, D., Tartari, M., Valenza, M. and Cattaneo, E. (2005). Progressive loss of BDNF in a mouse model of Huntington's disease and rescue by BDNF delivery. Pharmacological Research 52(2): 133-9. Zufferey, R., Dull, T., Mandel, R.J., Bukovsky, A., Quiroz, D., Naldini, L. and Trono, D. (1998). Self-inactivating lentivirus vector for safe and efficient in vivo gene delivery. Journal of Virology 72(12): 9873-80. 251
© Copyright 2025