doi:10.1093/brain/aws195 Brain 2012: 135; 2826–2837 | 2826 BRAIN A JOURNAL OF NEUROLOGY Immunoglobulin G Fc receptor deficiency prevents Alzheimer-like pathology and cognitive impairment in mice Paula Fernandez-Vizarra,1,* Oscar Lopez-Franco,1 Ben˜at Mallavia,1 Alejandro Higuera-Matas,3 Virginia Lopez-Parra,1 Guadalupe Ortiz-Mun˜oz,1 Emilio Ambrosio,3 Jesus Egido,1 Osborne F. X. Almeida2 and Carmen Gomez-Guerrero1 1 Renal and Vascular Inflammation, Nephrology Department, IIS-Fundacion Jimenez Diaz, Autonoma University, Avda. Reyes Catolicos 2, 28040 Madrid, Spain 2 NeuroAdaptations Group, Max Planck Institute for Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany 3 Department of Psychobiology, School of Psychology, UNED, C/Juan del Rosal 10, 28040 Madrid, Spain *Present address: NeuroAdaptations Group, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany Correspondence may also be addressed to: Paula Fernandez-Vizarra, PhD, NeuroAdaptations Group, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany. E-mail: [email protected] Alzheimer’s disease is a severely debilitating disease of high and growing proportions. Hypercholesterolaemia is a key risk factor in sporadic Alzheimer’s disease that links metabolic disorders (diabetes, obesity and atherosclerosis) with this pathology. Hypercholesterolaemia is associated with increased levels of immunoglobulin G against oxidized lipoproteins. Patients with Alzheimer’s disease produce autoantibodies against non-brain antigens and specific receptors for the constant Fc region of immunoglobulin G have been found in vulnerable neuronal subpopulations. Here, we focused on the potential role of Fc receptors as pathological players driving hypercholesterolaemia to Alzheimer’s disease. In a well-established model of hypercholesterolaemia, the apolipoprotein E knockout mouse, we report increased brain levels of immunoglobulin G and upregulation of activating Fc receptors, predominantly of type IV, in neurons susceptible to amyloid b accumulation. In these mice, gene deletion of -chain, the common subunit of activating Fc receptors, prevents learning and memory impairments without influencing cholesterolaemia and brain and serum immunoglobulin G levels. These cognition-protective effects were associated with a reduction in synapse loss, tau hyperphosphorylation and intracellular amyloid b accumulation both in cortical and hippocampal pyramidal neurons. In vitro, activating Fc receptor engagement caused synapse loss, tau hyperphosphorylation and amyloid b deposition in primary neurons by a mechanism involving mitogen-activated protein kinases and b-site amyloid precursor protein cleaving enzyme 1. Our results represent the first demonstration that immunoglobulin G Fc receptors contribute to the development of hypercholesterolaemia-associated features of Alzheimer’s disease and suggest a new potential target for slowing or preventing Alzheimer’s disease in hypercholesterolaemic patients. Keywords: Alzheimer’s disease; hypercholesterolaemia; apolipoprotein E; immunoglobulin receptors; cognitive dysfunction Received June 8, 2011. Revised June 8, 2012. Accepted June 10, 2012 ß The Author (2012). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: [email protected] Downloaded from by guest on October 21, 2014 Correspondence to: Carmen Gomez-Guerrero, PhD, Renal and Vascular Inflammation IIS-Fundacion Jimenez Diaz Avda. Reyes Catolicos 2, 28040 Madrid, Spain E-mail: [email protected] or [email protected] FcR in sporadic Alzheimer-like disease Brain 2012: 135; 2826–2837 | 2827 Abbreviations: apoE = apolipoprotein E; BACE1 = b-site amyloid precursor protein cleaving enzyme 1; JNK = c-Jun N-terminal kinase; DKO = double knockout; ERK = extracellular regulated mitogen-activated protein kinase; GFAP = glial fibrillary acidic protein; IgG = immunoglobulin G; MCP1 = monocyte chemoattractant protein 1; FcR = receptor for the constant Fc region of immunoglobulin G; TNF = tumour necrosis factor Introduction Materials and methods Reagents Soluble IgG immune complexes (Oreskes and Mandel, 1983) were obtained by heat aggregation (30 min at 63 C) of monomeric mouse IgG (Cappel, MP Biomedicals) and subsequent centrifugation to eliminate insoluble immune complexes as described (Hernandez-Vargas et al., Downloaded from by guest on October 21, 2014 The pre-dementia phase of Alzheimer’s disease can be modelled in transgenic mice carrying human mutations (Ashe and Zahs, 2010). Such models are considered useful in the development of therapeutics aimed at slowing the transition from asymptomatic Alzheimer’s disease—in which the neuropathology is already initiated—to full-blown Alzheimer’s disease. On the other hand, the amplifying effects of secondary changes are now thought to confound therapeutic interventions and given that Alzheimer’s disease develops over decades, it is clearly important to improve understanding of the mechanisms that initiate this pathology. Animal models burdened with risks associated with the development of Alzheimer’s disease serve this goal better than those involving overexpression of molecules that are causally associated with Alzheimer’s disease. The neuropathological characteristics of Alzheimer’s disease include amyloid b depositions and accumulations of abnormally hyperphosphorylated tau protein in selected brain regions; in addition, Alzheimer’s disease brains show abundant signs of microvascular damage and pronounced inflammation (Bertram et al., 2010). Current evidence suggests that amyloid b initiates a disease process that progresses to cognitive impairment and that tau mediates cognitive dysfunction (Ashe and Zahs, 2010). Although the pathways contributing to increased amyloid b levels appear to differ according to predisposing risk factors, it is reasonable to assume that these pathways converge at some point before triggering unbalanced amyloid b metabolism; identification of such an upstream hub would facilitate research and therapeutic developments. Patients suffering from sporadic Alzheimer’s disease show elevated levels of immunoglobulin G (IgG) autoantibodies against non-brain antigens (Ounanian et al., 1990), and express receptors for the constant Fc region of IgG (FcR) (Bouras et al., 2005) in neuronal populations that are vulnerable to the disease (Morrison and Hof, 2002). Importantly, increased circulating and brain IgG levels are found in individuals at risk of developing Alzheimer’s disease (Ballard et al., 2011), including advanced age (Listı` et al., 2006), stress (Nakata et al., 2000) and metabolic disorders such as diabetes, obesity and atherosclerosis (Lu et al., 2001; Turk et al., 2001; Haroun and El-Sayed, 2007; Okamatsu et al., 2009; Winer et al., 2011). Abnormal modifications of proteins, such as oxidation and non-enzymatic glycosylation, are also observed during early stages of metabolic disorders (Haroun and El-Sayed, 2007; Winer et al., 2011); these result in an adaptive immune response with subsequently sustained high levels of circulating autoantibodies (mainly IgG isotype) (Doyle and Mamula, 2001) and the corresponding immune complexes formed by antibody–antigen interaction. Antibody penetrance into the brain is severely limited in physiological conditions; however, weakening of the blood–brain barrier allows entry of immunoglobulins and immune complexes, as has been observed in individuals with metabolic syndrome, and risk for Alzheimer’s disease, and also in experimental models of Alzheimer’s disease-related pathologies (Methia et al., 2001; Kuang et al., 2004; Hafezi-Moghadam et al., 2007; Bake et al., 2009; Diamond et al., 2009). Thus, we hypothesize that brain FcR activation may be a major convergence point for sporadic Alzheimer’s disease triggered by the occurrence of two pathological phenomena: IgG overproduction and blood–brain barrier leakage. In the mouse, four FcR classes that differ in affinity, specificity and function have been described. Activating FcRs (I, III and IV) associate with the common -chain which harbours the immunoreceptor tyrosine-based activation motif and elicit an immune response upon ligand (IgG or immune complexes) binding; in contrast, the inhibitory FcRIIb contains the immunoreceptor tyrosine-based inhibition motif and nullifies cell activation (Nimmerjahn et al., 2005; Nimmerjahn and Ravetch, 2008). FcRs have been localized in neurons, microglia, astroglia and oligodendrocytes (Bouras et al., 2005) and have been shown to play an important role in myelination (Nakahara et al., 2003). Hypercholesterolaemia, a common feature in metabolic disorders, is characterized by autoimmune responses triggered by most forms of modified low density lipoproteins (Burut et al., 2010). Interestingly, hypercholesterolaemic mice display several neuropathological hallmarks of Alzheimer’s disease, including increased levels of amyloid precursor protein and amyloid b, abnormally hyperphosphorylated tau and inflammation, together with cognitive impairments (Oitzl et al., 1997; Veinbergs et al., 1999; Crisby et al., 2004; Rahman et al., 2005; Bjelik et al., 2006). In this study, we investigated the potential role of FcR in the aetiopathogenesis of Alzheimer’s disease. Given that a multiplicity of risk factors may be represented in hypercholesterolaemia and the fact that Alzheimer’s disease is associated with IgG overproduction and blood–brain barrier leakage, studies were performed in apolipoprotein E (apoE) knockout mice, which display overt hypercholesterolaemia (Zhang et al., 1992). Therefore, the features characteristic of Alzheimer’s disease were comparatively studied between single apoE knockout (apoE / ) mice and double knockout (DKO) mice (Hernandez-Vargas et al., 2006), which are gene deficient in both apoE and -chain, the common subunit necessary for assembly, cell-surface localization and functionality of activating FcRs (Nimmerjahn and Ravetch, 2008). 2828 | Brain 2012: 135; 2826–2837 2006). Culture media, supplements and transfection reagent were purchased from Lonza and Life Technologies; SP600125 and U0126 from Stressgen Bioreagents Corp.; small interfering RNA from Santa Cruz Biotechnologies. The 9E9 FcR blocking antibody was generously provided by Dr J.V. Ravetch (The Rockefeller University, NY) (Nimmerjahn et al., 2005). Primary antibodies used were: synaptophysin, microtubule associated protein 2, and tau phosphorylated at Ser199/202 (Millipore); tau doubly phosphorylated at Ser202/Thr205, Ser199/202 and Ser205/208 (AT8 antibody) (Pierce Biotechnology Inc.); glial fibrillary acidic protein (GFAP; Sigma-Aldrich); a neoepitope in amyloid b, to exclude cross-reactions with its precursor amyloid precursor protein (IBL); FcRIV (Santa Cruz Biotechnologies); mouse IgG (Amersham); b-site amyloid precursor protein cleaving enzyme 1 (BACE1; ProSci Inc.); insulin degrading enzyme (Abcam); CD11b (Abcam); -tubulin (Sigma-Aldrich); and glyceraldehyde-3-phosphate dehydrogenase (GAPDH, Millipore). Assays for real-time PCR were from Life Technologies. Mice Behavioural tests Mice were housed three to five per cage and allowed to habituate to the cage environment for 2 weeks before behavioural testing. Housing conditions were kept constant until the end of behavioural testing. All behavioural tests were carried out under dim light in a sound-proof room by the same researcher and were conducted in the following order: elevated plus maze, object location test. Elevated plus maze Mice were allowed to freely explore the maze for 8 min. Anxiety index was calculated as the time spent in open arms relative to the total time spent in closed and open arms and in the centre. Locomotor activity index was calculated as frequency of entry into closed arms. Object location test One day after the elevated plus maze test, mice were habituated to the empty arena placed in a room with extra-maze cues for 15 min during 5 days before training. In the training phase, mice were exposed to two identical objects for 5 min. After a delay of 14 min, the time spent exploring the objects in a new (novel) and in the old (familiar) locations was recorded during 3 min (test phase). To analyse cognitive performance, a location index (or relative exploration time) was calculated as previously described (Murai et al., 2007) with a slight modification: to/Te, where to = time spent exploring either the displaced or the non-displaced object and Te = total time spent exploring both objects. Biochemistry Total serum cholesterol concentrations were measured using standard enzymatic methods (Invitrogen). Total serum immunoglobulins were measured by sandwich ELISA using specific antibodies recognizing mouse IgG1, IgG2a/c and IgG3 (BD Biosciences). Primary neuronal cultures Mouse cortical neurons from prenatal embryonic Day 17 were mechanically dissociated and plated on poly-L-lysine-coated coverslips or plates. Cell suspension (106 cells/ml) was seeded in Dulbecco’s modified Eagle medium containing 5% foetal bovine serum, 0.05% GlutaMAXTM and 1% penicillin/streptomycin, and then cultured in NeurobasalÕ medium supplemented with 2% B27, 1% GlutaMAXTM and 0.1% penicillin/streptomycin. On Day 5 in vitro, 10 mM cytosine arabinoside was added. Contaminating glial cells in neuronal cultures accounted for 52% (astroglia and microglia combined) on the day of use. Cultures at Day 12 were stimulated with IgG immune complexes (150 mg/ml). In some experiments, cells were pretreated for 1 h with mitogen-activated protein kinases inhibitors [c-Jun N-terminal kinase (JNK) inhibitor, SP600125, 5 10 5 M; extracellular regulated mitogen-activated protein kinase (ERK) inhibitor, U0126, 10 5 M] or FcRIV blocking antibody (9E9 or irrelevant hamster IgG, 1 mg/106 cells; Nimmerjahn et al., 2005) before stimulation. For knock-down experiments, cells were transfected with 60 pM of small interfering RNA (FcRIV or irrelevant) and 8 ml of LipofectamineTM 2000 in culture medium. Small interfering RNA knocking-down efficiency checked by real-time PCR was 72.1 0.3% at 24 h after transfection (n = 3). Immunocytochemistry Brains were fixed in 4% paraformaldehyde for 4 h. Cryostat sections (10 mm) on slice were used for immunohistochemistry. For antigenretrieval in amyloid b immunostaining, sections were pretreated with 88% formic acid for 5 min. Non-specific binding sites were blocked by incubating sections in 1% bovine serum albumin and 5% pre-immune serum diluted in 0.5% Tween-20 in PBS for 1 h at room temperature in slight orbital agitation. Primary antibody was incubated in 0.5% Tween-20 (FcRIV 1:50, amyloid b 1:100, GFAP 1:2000; CD11b 1:20) for three overnights at 4 C in slight orbital agitation. For immunocytochemistry, fixation in 2% paraformaldehyde (10 min, room temperature) was performed before incubation of primary antibodies in PBS overnight at 4 C in slight agitation (FcRIV 1:50, microtubule associated protein 2 1:100, amyloid b 1:100, GFAP 1:2000, CD11b 1:20). For both in vivo and in vitro studies, fluorescent secondary antibodies (1:100 in PBS) were incubated at room temperature for 1 h with orbital agitation. Incubation with 40 6-diamidino-2-phenylindole (DAPI; 1:10 000 in PBS) was then performed (10 min, room temperature). Confocal images were obtained by sequential scan. Western blot Cytosolic proteins were extracted from cells (Ortiz-Munoz et al., 2010) and the cerebral cortex from mice (Rodrigo et al., 2004) and resolved on SDS–PAGE, transferred onto polyvinylidene fluoride membranes and immunoblotted with specific antibodies (Ortiz-Munoz et al., 2010). Optical densities of individual bands were normalized to loading controls (GAPDH or -tubulin) and n-fold changes were obtained by normalization against results obtained under basal conditions (untreated cells) or control (wild-type) mice, as appropriate. Downloaded from by guest on October 21, 2014 All animal studies conformed to Directive 2010/63/EU of the European Parliament and were approved by the Institutional Animal Care and Use Committee. Wild-type and apoE knockout mice were purchased from Jackson Laboratory; the DKO mice (gene deficient in both apoE and -chain) were generated in our laboratory as previously described (Hernandez-Vargas et al., 2006). Wild-type, apoE / and DKO mice were fed a standard mouse lab chow diet (Panlab) and allowed to age. Mice from each group were sacrificed at 12 months (wild-type, n = 5; apoE / , n = 8; DKO, n = 7) and 21 months (wild-type, n = 5; apoE / , n = 12; DKO, n = 11). Mice brains were obtained after deep anaesthesia with a mixture (0.01 ml/g body weight, intraperitoneally) of ketamine (10 mg/ml) and xylazine (1 mg/ml) and perfusion with saline. P. Fernandez-Vizarra et al. FcR in sporadic Alzheimer-like disease Brain 2012: 135; 2826–2837 | 2829 Results FcR deficiency restores cognitive and synaptic status and attenuates Alzheimer-like pathology in hypercholesterolaemic mice hypercholesterolaemic and control mice at middle-age (12-month-old). (A) Western blot of IgG in the cerebral cortex of wild-type (WT), apoE / and DKO mice. Representative blots from each group (wild-type, n = 5; apoE / , n = 8; DKO, n = 7) are shown. Summary of densitometric analysis is expressed in arbitrary units (a.u.). (B) Immunoglobulin isotype distribution in mouse sera measured by ELISA. Values are expressed as absorbance units at = 450 nm. Values are mean SEM of five animals per group. (C) Real-time PCR analysis of activating and inhibitory FcRs in the hippocampus and cerebral cortex of mice. Values are mean SEM of studied animals per group (wild-type, n = 5; apoE / , n = 8; DKO, n = 7). *P 5 0.05 and **P 5 0.01 versus wild-type; #P 5 0.05 versus apoE / . Messenger RNA expression Total RNA from cells and tissues was extracted and retro-transcribed as previously described (Ortiz-Munoz et al., 2010). Gene expression was analysed in duplicate by real-time PCR on a TaqManÕ ABI 7500 sequence detection system (Applied Biosystems) and normalized to housekeeping 18S transcripts. Results are given as n-fold changes, relative to control groups. The relative expression levels of activating and inhibitory FcR were calculated according to the formula (FcRI + FcRIII + FcRIV)/FcRIIb and expressed as a percentage of control values. Statistics Statistical significance was tested by unpaired Student’s t-test and one-way ANOVA followed by an appropriate post hoc least significant difference comparison test (GraphPad Prism software). A value of P 5 0.05 was considered to be statistically significant. Downloaded from by guest on October 21, 2014 Figure 1 Levels of IgG and expression of FcR isoforms in Western blot studies in brain lysates demonstrated very low levels of IgG in middle-aged (12-month-old) wild-type mice and high levels in apoE / and DKO (deficient in apoE and -chain genes) mice, with both knockout strains showing a similar content (Fig. 1A). ApoE / and DKO groups showed comparable serum cholesterol levels (460 27 mg/dl versus 475 20 mg/dl, P 4 0.05). Furthermore, serum antibody levels confirmed increased brain IgG levels in apoE / and DKO mice, as compared to wild-type mice. The two strains of knockout mice (apoE / and DKO) did not differ in their IgG subclass (IgG1, IgG2 and IgG3) expression profiles (Fig. 1B), i.e. they shared a similar immune imprint. Given that high levels of IgG reportedly upregulate FcR expression in different inflammatory disease models (Nimmerjahn and Ravetch, 2008), we next examined whether experimental hypercholesterolaemia altered FcR gene expression in the brain. Consistent with the increase in IgG levels, we found an increased expression of activating and inhibitory FcRs, mainly of the FcRIV isoform, in brains from apoE / animals, when compared with wild-type mice (Fig. 1C); this resulted in a net activating profile (activating:inhibitory ratio, percentage versus wild-type mice: cortex, 6.06; hippocampus, 13.99). In contrast, apart from changes in the expression of FcRIII, hypercholesterolaemic DKO mice did not show any difference in FcR expression in the cortex and hippocampus, compared to wild-type animals (Fig. 1C). Middle-aged hypercholesterolaemic apoE / mice showed intracellular amyloid b immunostaining in pyramidal neurons of the hippocampus and temporal cortex (Fig. 2A and C), consistent with the early neuroanatomical and cellular distribution of intracellular amyloid b deposition found in patients with Alzheimer’s disease (Gouras et al., 2000; Gyure et al., 2001; Fernandez-Vizarra et al., 2004). Immunohistochemical localization of amyloid b was similar to that of FcRIV (Fig. 2D and E), the major receptor expressed in hypercholesterolaemic mice brains; FcRIV staining was observed in pyramidal neurons of the hippocampus, temporal cortex, but also of the cingulate cortex (data not shown) of apoE / mice, but not in other types of neuron or glial cells. ApoE / mice also showed a significant increase in tau hyperphosphorylation in the cerebral cortex, as compared to age-matched wild-type controls (Fig. 2H). Importantly, deletion of activating FcR in DKO mice attenuated amyloid b immunostaining (Fig. 2A and C) and tau hyperphosphorylation (Fig. 2H) in all neuroanatomical areas studied. Interestingly, FcR deficiency was also protective against amyloid b deposition in aged (21-month-old) mice (Fig. 2B). Furthermore, lack of functional FcRs significantly reduced the gene expression of the inflammatory cytokine tumour necrosis factor (TNF) and the monocyte chemoattractant protein 1 (MCP1, also known as Ccl2; 2830 | Brain 2012: 135; 2826–2837 P. Fernandez-Vizarra et al. Downloaded from by guest on October 21, 2014 Figure 2 Functional deficiency in activating FcRs protects against amyloid b deposition, tau phosphorylation, astrogliosis and cytokine expression in hypercholesterolaemic mice. Role of FcRIV. (A and B) Representative confocal images (scale bar = 20 mm) and magnification details showing amyloid b (Ab) immunostaining (green) and nuclei (blue) taken from the hippocampus of apoE / and DKO mice aged 12 and 21 months, as indicated. (C) Amyloid b immunostaining in the cerebral cortex from middle-aged (12-month-old) mice. Note the increase in both intracellular and extracellular amyloid b immunostaining in apoE / brains. (D and E) Representative FcRIV immunofluorescence in hippocampus (D, scale bar = 10 mm) and cerebral cortex (E, scale bar = 20 mm) from middle-aged mice, showing FcRIV in green and cell nuclei in blue. Note the similar cellular distribution of amyloid b and FcRIV and the absence of FcRIV in glial cells. (F and G) Representative confocal images of astrogliosis (F, red) and microgliosis (G, green) in the hippocampus from middle-aged mice (nuclear staining in blue). (H and I) Western blot analysis of tau phosphorylation (ptau, H) and astrogliosis (GFAP marker, I) in the cerebral cortex of middle-aged mice. Representative blots from each group are shown. Summary of densitometric analysis is expressed as fold increases. (J) Cytokine gene expression in the hippocampus and cerebral cortex of middle-aged mice measured by real-time PCR. IL-6 = interleukin 6; IFN- = interferon . Values are mean SEM of studied animals per group [for 12-month-old mice: wild-type (WT), n = 5; apoE / , n = 8, DKO, n = 7; for 21-month-old mice: **P 5 0.01, n = 5; apoE / , n = 12; DKO, n = 11]. *P 5 0.05 and **P 5 0.01 versus wild-type; #P 5 0.05 versus apoE / . Fig. 2J) as well as the astroglial marker GFAP (Fig. 2I) in cerebral cortex of hypercholesterolaemic mice. Immunohistochemical studies revealed that astrogliosis was restricted to the hippocampus (Fig. 2F), the region most affected by amyloid b deposition. By contrast, microgliosis, detected in the hippocampus (Fig. 2G) and across several cortical areas (data not shown), did not correlate with the pattern of amyloid b deposition or FcRIV overexpression and was not affected by FcR deficiency. A robust loss of synapses was found in apoE / mice (Fig. 3A), as judged by the expression of synaptophysin; currently, synaptic FcR in sporadic Alzheimer-like disease Brain 2012: 135; 2826–2837 | 2831 Figure 3 Synapse loss and cognitive dysfunction are prevented by FcR deletion. (A) Representative immunoblots and quantification of synaptophysin (syn) expression in the cerebral cortex of middle-aged mice. Values are mean SEM of the total of studied animals per group: wild-type (WT), n = 5; apoE / , n = 8; DKO, n = 7. **P 5 0.01 versus wild-type; #P 5 0.05 versus apoE / . (B) Spatial learning and memory measured using the object location test index (exploration time of a given location relative to total object exploration time). (C) Anxiety levels and locomotor activity measured with the elevated plus maze test. Anxiety index was calculated as the relative time in open arms, and locomotor activity as frequency of entry into closed arms. All behavioural studies were performed in middle-aged mice (wild-type, n = 9; apoE / , n = 9; DKO, n = 10). **P 5 0.01 versus familiar location. Engagement of neuronal FcR in primary cultures induces Alzheimer-like pathology The role of neuronal FcR in hypercholesterolaemia-associated Alzheimer-like pathology was further studied in primary neuronal cultures from wild-type, apoE / and DKO mice. In wild-type and apoE / neurons, IgG immune complexes induced a large increase in the expression of genes for all activating FcR isoforms, mainly FcRIV (IV 4 I III); smaller effects were observed with regard to the inhibitory FcRIIb isoform, resulting in a net activating profile (activating/inhibitory ratio, n-fold versus basal: wild-type, 2.58; apoE / , 1.57; Fig. 4A). Immmunofluorescence confirmed the induction of the activating FcRIV isoform by IgG immune complexes (Fig. 4B). In contrast, immune complexes failed to induce activating FcR expression in neurons from DKO mice; these neurons showed an upregulation of inhibitory FcRIIb only (Fig. 4A and B). In parallel, amyloid b accumulation was observed after FcR engagement in wild-type pyramidal neurons (Fig. 5A). Immune complexes also induced tau hyperphosphorylation at epitopes that are considered important in Alzheimer’s disease (Fig. 5B and C). Furthermore, significant synaptic loss was found in immune complex-stimulated primary neurons from wild-type and apoE / mice (Fig. 5D). Importantly, FcR deficiency attenuated amyloid b deposition and tau hyperphosphorylation (Fig. 5A–C), protected primary neurons against synapse loss (Fig. 5D) and significantly blocked the expression of inflammation-related genes after stimulation with immune complexes (Fig. 5F). To directly assess the involvement of FcRIV, the most inducible activating FcR found in vivo and in vitro, inhibition experiments were performed with small interfering RNA and neutralizing antibody. FcRIV small interfering RNA efficiently abrogated intracellular amyloid b accumulation (Fig. 5E), tau phosphorylation (Fig. 5C) and synaptic loss (Fig. 5D) in response to immune complexes. However, FcRIV small interfering RNA partially reduced MCP1 gene expression without significant effects on TNF (Fig. 5F and G). Similarly, blocking FcRIV with a highly specific antibody prevented tau phosphorylation and synaptic loss and attenuated MCP1 protein expression (Fig. 5C, D and G). Furthermore, and consistently with our in vivo results, confocal microscopic studies using specific markers of astro- and microglial cells (GFAP and CD11b, respectively) failed to reveal FcRIV immunostaining in either astroglia or microglia (data not shown, note that cultures contained 52% of glial cells), suggesting that Downloaded from by guest on October 21, 2014 loss appears to be the best correlate of cognitive dysfunction in Alzheimer’s disease (Masliah et al., 2001; Reddy et al., 2005; Gomez et al., 2010). Importantly, FcR deletion completely rescued synaptophysin protein levels in hypercholesterolaemic mice (Fig. 3A). This recovery was shown to be functionally relevant as shown by performance in the object location test, a measure of hippocampus-medial temporal lobe related learning and memory (Eichenbaum et al., 2007). In contrast to apoE / mice, DKO mice and wild-type mice discriminated between novel and familiar locations to similar extents, indicating intact cognitive function in the DKO mice (Fig. 3B). Specifically, differences observed in spatial learning and memory were not a function of total object exploration times in the training (wild-type, 15.7 6.1 s; apoE / , 17.3 5.9 s; DKO, 5.3 1.2 s, n = 4–10) and test phases (wild-type, 10.3 3.2 s; apoE / , 7.5 2.1 s; DKO, 4.0 1.6 s, n = 4–10) or of different relative exploration times of the two objects during the acquisition phase of the task (P 4 0.05 in all cases). Furthermore, the observed differences in cognitive performance could not be explained by increased anxiety or decreased locomotor activity, as revealed in the elevated plus maze test (Fig. 3C). 2832 | Brain 2012: 135; 2826–2837 P. Fernandez-Vizarra et al. Figure 4 Immune complexes induce FcR overexpression in primary neurons. (A) The expression profile of activating FcRs (I, III and IV) and inhibitory FcRIIb in primary neuronal cultures from wild-type (WT), apoE / , and DKO mice was measured by real-time PCR after stimulation with immune complexes (IC). Basal is represented as a dashed line. Data are mean SEM of three to six experiments. *P 5 0.05 and **P 5 0.01 versus basal; #P 5 0.05 versus stimulated wild-type cells. (B) Immunofluorescence of FcRIV (red) and the neuronal marker microtubule associated protein 2 (MAP-2, green) in wild-type and DKO neuronal cultures under basal conditions and after 24-h treatment with IC. Representative of three independent experiments. Scale bar = 20 mm. neurons are responsible for the observed responses following treatment with immune complexes. Role of BACE1, JNK and ERK pathways in the neuroprotective effects of FcR deficiency Both increased production and decreased clearance of amyloid b have been implicated in sporadic Alzheimer’s disease in humans Discussion This study used an animal model of hypercholesterolaemia to investigate the upstream pathways that lead to neuropathological features characteristic of Alzheimer’s disease, i.e. those preceding inappropriate increases in amyloid b. Targeting early pathological mechanisms before the onset of accumulation of amyloid b and associated amplifying events, such as neuroinflammation and vascular changes, can be expected to facilitate therapeutic interventions. While genetic and molecular data indicate the important role of amyloid b in the initiation of Alzheimer’s disease (Selkoe, 2001; Hardy, 2006), a number of recent clinical trials, aimed at reducing amyloid b burden in mild-to-moderate stages of dementia, have failed (Shepardson et al., 2011b). Accordingly, the importance of focusing on factors that induce amyloid b accumulation and in advance of secondary changes has become even more evident. It is now recognized that hypercholesterolaemia, often found in diabetes, obesity and atherosclerosis, is a key risk factor in sporadic Alzheimer’s disease (Ballard et al., 2011; Shepardson et al. 2011a). In support of this, several studies have shown that the molecular, neuroanatomical and cognitive changes found in Alzheimer’s disease are recapitulated in hypercholesterolaemic mice (Oitzl et al., 1997; Veinbergs et al., 1999; Crisby et al., 2004; Rahman et al., 2005; Bjelik et al., 2006). The work presented here represents the first to suggest a pivotal role for brain FcR in the pathogenesis of Downloaded from by guest on October 21, 2014 (Yang et al., 2003; Li et al., 2004; Mawuenyega et al., 2010). Accordingly, we investigated whether the enzymes BACE1 and insulin degrading enzyme were modulated by hypercholesterolaemia and FcR functional deletion. Western blot analysis for BACE1 in the cerebral cortex from apoE / mice revealed a significant increase in the 140-kDa band, with a parallel decrease in the 70-kDa band (Fig. 6A), suggestive of an increase in homodimeric BACE1 activity in hypercholesterolaemia (Schmechel et al., 2004; Jin et al., 2010). Interestingly, the effects of hypercholesterolaemia on BACE1 were abolished when the FcR gene was deleted (Fig. 6A). On the other hand, insulin degrading enzyme levels remained unchanged under hypercholesterolaemic conditions and were unaffected by FcR deletion, as shown by western blot analysis (Fig. 6B). Activation of JNK and ERK pathways have been implicated in synaptic plasticity (Curtis and Finkbeiner, 1999), amyloid b production and tau phosphorylation (Sato et al., 2002; Colucci-D’Amato et al., 2003) and are known to be involved in FcR signalling (Song et al., 2002; Luo et al., 2010). In our in vivo model, hypercholesterolaemia was associated with activation of JNK and ERK pathways; in contrast, JNK and ERK activation levels were similar in wild-type and FcR-deficient mice (Fig. 7A). In vitro, immune complexes induced a sustained activation of both JNK and ERK; these changes were seen before and during expression of Alzheimer’s disease markers and synaptic loss in wild-type cultures, and were precluded by FcR deficiency (Fig. 7B). Moreover, pretreatment of neurons with specific inhibitors of JNK and ERK significantly attenuated the tau hyperphosphorylation and synaptic loss induced by immune complex treatment (Fig. 7C and D). FcR in sporadic Alzheimer-like disease Brain 2012: 135; 2826–2837 | 2833 Representative immunofluorescence images from three independent experiments showing amyloid b (A) and phosphorylated tau (ptau, B) in red combined with the neuronal marker microtubule associated protein 2 (MAP-2, green) and cell nuclei (blue), both in wild-type (WT) and DKO neurons under basal conditions or following a 48-h exposure to immune complexes (IC). Arrow indicates typical intracellular amyloid b deposit in a pyramidal neuron resembling a senile plaque. Arrowheads indicate basal and apical dendrites from pyramidal neurons stained for AT8 in stimulated cells. Scale bars = 10 mm. (C and D) Western blot analysis of phosphorylated tau (ptau, C) and synaptophysisn (syn, D) in primary neurons after a 48-h treatment with immune complexes. The effect of FcRIV inhibition in wild-type neurons was assessed with small interfering RNA (siRNA) and blocking antibody (Block. Ab). Representative immunoblots of three to six independent experiments are shown. (E) Photomicrographs of triple-colour immunofluorescence (red, amyloid b; green, microtubule associated protein 2; and blue, nuclei) from wild-type neurons transfected with FcRIV small interfering RNA before stimulation with immune complexes. Arrowhead denotes neuronal debris (irregular morphology without nuclear staining) strongly stained for amyloid b, suggestive of a senile plaque derived from intracellular deposits. Scale bar = 20 mm; representative of three independent experiments. (F) Time-dependent induction of TNF and MCP1 by immune complexes in primary cultures from WT, apoE / and DKO mice, measured by real-time PCR. The involvement of FcRIV was assessed with small interfering RNA. (G) MCP1 protein production in wild-type neurons was measured by ELISA. Dashed lines represent basal conditions. Data are mean SEM of three to six experiments per group. *P 5 0.05 and **P 5 0.01 versus basal; #P 5 0.05 and ##P 5 0.01 versus stimulated wild-type cells. sporadic Alzheimer’s disease. We show that hypercholesterolaemia results in a significant increase in brain IgG levels with a parallel increase in activating FcR gene expression. Furthermore, hypercholesterolaemic mice exhibited increased protein expression of FcRIV isoform in pyramidal neurons vulnerable to intraneuronal amyloid b deposition in the hippocampus and temporal cortex, regions similarly affected in the brains of patients with Alzheimer’s disease (Gouras et al., 2000; Gyure et al., 2001; Fernandez-Vizarra et al., 2004). Notably, we show that FcRIV is more widely expressed than amyloid b; besides the hippocampus and temporal cortex, we observed FcRIV staining in the cingulate cortex, an area that is affected in early-stage Alzheimer’s disease (Barnes et al., 2007; Pengas et al., 2010). These findings are complemented by the observation that cross-linking of FcR with IgG immune complexes leads to amyloid b accumulation, hyperphosphorylation of tau at sites associated with Alzheimer’s disease and synaptic loss in primary neuronal cultures. An important finding to emerge from this study is that deletion of FcR function in hypercholesterolaemic mice prevents learning and memory impairments. These cognition-protective effects were associated with a reduction in amyloid b deposition, abnormal tau hyperphosphorylation and synapse loss. Interestingly, the above mentioned behavioural and neuronal consequences of FcR deletion appear to occur independently of brain IgG and Downloaded from by guest on October 21, 2014 Figure 5 Over-activation of neuronal FcRs, and particularly the FcRIV isoform, induces Alzheimer-like pathology in vitro. 2834 | Brain 2012: 135; 2826–2837 P. Fernandez-Vizarra et al. Figure 6 FcR deletion precludes the apparent increase in active BACE1 induced by hypercholesterolaemia. Western blot analysis of BACE1 (A) and insulin degrading enzyme (IDE, B) protein levels in the cerebral cortex from middle-aged mice. Representative blots from each group are shown. Values are mean SEM (wild-type, n = 5; apoE / , n = 8; DKO, n = 7). *P 5 0.05 versus wild-type; ##P 5 0.01 versus apoE / . study in mice showed that disruption of the blood–brain barrier, and subsequent deposition of IgG in the brain, causes synaptic loss and memory impairment (Bell et al., 2010). Loss of integrity of the blood–brain barrier, as well as IgG deposits and/or FcR expression, are also associated with Alzheimer’s disease risk factors other than ageing, namely atherosclerosis, obesity and diabetes (Methia et al., 2001; Kuang et al., 2004; Hafezi-Moghadam et al., 2007; Bake et al., 2009; Diamond et al., 2009). Moreover, the brain is among the most commonly affected organs in patients with immune-mediated diseases, such as systemic lupus erythematosus, subacute endocarditis and hepatitis C infection (Harris and Cobbs, 1996; Lister and Hickey, 2006; Carvalho-Filho et al., 2012), in which neuronal dysfunction may result from direct immune effects (autoantibody binding to cell surface, circulating immune complex deposition and inflammation) on brain resident cells via FcR and complement activation. Although these immune conditions might influence the likelihood of developing Alzheimer’s disease and other dementias, further research in this field is clearly worthwhile. In light of strong evidence that amyloid b production is increased (Yang et al., 2003; Li et al., 2004) and cleared with reduced efficiency (Mawuenyega et al., 2010) in sporadic Alzheimer’s disease, we here explored the mechanisms through which FcR activation may lead to increased levels of cerebral amyloid b. We focused on BACE1, whose homodimerization and association with amyloid precursor protein leads to the initial cleavage of amyloid precursor protein (Schmechel et al., 2004; Jin et al., 2010) and whose expression and activity is upregulated in patients with sporadic Alzheimer’s disease (Yang et al., 2003; Li et al., 2004). We observed increased amounts of homodimeric BACE1 in hypercholesterolaemic mice, an effect that was abolished when the FcR gene was deleted. Further, in line with previous work that reported that BACE1 homodimerization depends on its phosphorylation at specific serine/threonine residues (Walter et al., 2001), we observed sustained activation of JNK and ERK in the brains of hypercholesterolaemic apoE / mice, and in cultured neurons that were exposed to immune complexes; activated JNK Downloaded from by guest on October 21, 2014 blood cholesterol levels since neither of these parameters are altered in FcR null mutant mice. Support for the view that the FcRIV isoform is responsible for mediating the detrimental effects of immune complexes on neurons stems from experiments in which selective FcRIV inhibition (small interfering RNA silencing and blocking antibody) rescued immune complex-induced synaptic loss and the accumulation of amyloid b and hyperphosphorylated tau. It is important to note that our analysis revealed that FcRIV expression is restricted to neurons both in vivo and in vitro, as no signal was detected in astro- and microglia. The results, obtained in primary neuronal cultures, indicate that MCP1 and TNF are unlikely to exert substantial influence over the development of amyloid b accumulation, tau pathology and synaptic loss in primary neurons: although FcRIV knock-down in neurons did not preclude cytokine (MCP1 and TNF) induction by immune complexes, this inflammatory response was not followed by the aforementioned neuropathological features. Nevertheless, a potentially protective role of these cytokines cannot be ruled out at present. In this context, it is worth mentioning that MCP1, a cytokine involved in the recruitment, proliferation and activation of glial cells, may trigger gliosis in the hippocampus; since astro- and microglia do not express the only hypercholesterolaemia-responsive FcR isoform (FcRIV), gliosis in the hippocampus cannot be attributed to activation of FcR. If early inflammatory responses serve to retard hypercholesterolaemia-associated Alzheimer’s disease pathology, interesting therapeutic opportunities could be explored by exploiting the differential regulation of inflammation and Alzheimer-related pathology by FcR isoforms; importantly, these would not necessarily interfere with the potentially protective functions of glial cells. Our hypothesis that IgG is involved in the onset of certain sporadic forms of Alzheimer’s disease is consistent with recent data suggesting that factors that induce weakening of the blood–brain barrier may be causally related to neurodegenerative disease; since this barrier gets leaky during normal ageing (Hafezi-Moghadam et al., 2007), hypercholesterolaemia would add to or multiply the risk of developing Alzheimer’s disease. Indeed, a recent FcR in sporadic Alzheimer-like disease Brain 2012: 135; 2826–2837 | 2835 Downloaded from by guest on October 21, 2014 Figure 7 JNK and ERK activation are involved in the neuroprotective actions of FcR deletion. (A) JNK and ERK activity was measured by western blot analysis of phosphorylated proteins (p-proteins: pJNK and pERK) in the cerebral cortex from middle-aged mice. (B) JNK and ERK activation in primary neurons under basal conditions (B) or following 48-h stimulation with immune complexes (IC). (C and D) Western blot analysis of tau phosphorylation (ptau, C) and synaptophysin levels (syn, D) in wild-type neurons pretreated with inhibitors of JNK (SP600125, SP) and ERK (U0126, U) before stimulation. Basal condition is represented as a dashed line. Summary of densitometric analysis is expressed as fold increases. Representative blots from in each group are shown. Values are mean SEM of studied animals per group (wild-type, n = 5; apoE / , n = 8; DKO, n = 7; A) and of three to six independent experiments (B–D). *P 5 0.05 versus wild-type (WT) mice or basal cells; #P 5 0.05 versus apoE / mice or stimulated wild-type cells. and ERK have been previously implicated in the generation of amyloid b and the abnormal hyperphosphorylation of tau (Sato et al., 2002; Colucci-D’Amato et al., 2003). In summary, our study describes a novel mechanism that may trigger certain sporadic forms of Alzheimer’s disease. The results point to FcR as a potential target for preventative intervention, at least in cases where hypercholesterolaemia is a risk factor. Although preventive approaches in those patients may primarily include reduction of hypercholesterolaemia, recent evidence indicates that lipid-lowering drugs are inefficacious treatments for Alzheimer’s disease (Shepardson et al., 2011b). Alternatively, strategies targeting antibody production and FcR balance and activities could determine the net functional effect and hence retard disease onset and progression. In future, it will be also important to determine whether levels of IgG immune complexes can serve as early biomarkers of sporadic Alzheimer’s disease and to assess the potential contribution of FcR signalling to forms of Alzheimer’s disease that are not directly associated with hypercholesterolaemia. Acknowledgements The authors thank Dr J. V. Ravetch (The Rockefeller University, New York) for generously providing FcRIV blocking antibody, Dr 2836 | Brain 2012: 135; 2826–2837 M.P. Sanchez and A.M. Garcia-Cabrero (Laboratory of Neurology, IIS-FJD, Madrid) and Drs C. Lopez-Menendez and L. Sanchez-Ruiloba (IIB Alberto Sols, CSIC-UAM) for antibody samples and skilled advice with mice and primary cultures. Funding Spanish Ministry of Science (SAF2009/11794), Fondo de Investigaciones Sanitarias (FIS PI10/00072, RECAVA RD06/ 0014/0035), Fundacion Renal In˜igo Alvarez de Toledo, Spanish Society of Nephrology and Lilly Foundation. P.F.-V. was supported by postdoctoral fellowships from FIS (Sara Borrell program) and Caja Madrid Foundation. References and PHF in clinically evaluated cases of Alzheimer’s disease. Histol Histopathol 2004; 19: 823–44. Gomez RM, Rosso OA, Berretta R, Moscato P. Uncovering molecular biomarkers that correlate cognitive decline with the changes of hippocampus’ gene expression profiles in Alzheimer’s disease. PLoS One 2010; 5: e10153. Gordon I, Genis I, Grauer E, Sehayek E, Michaelson DM. Biochemical and cognitive studies of apolipoprotein-E-deficient mice. Mol Chem Neuropathol 1996; 28: 97–103. Gouras GK, Tsai J, Naslund J, Vincent B, Edgar M, Checler F, et al. Intraneuronal Abeta42 accumulation in human brain. Am J Pathol 2000; 156: 15–20. Gyure KA, Durham R, Stewart WF, Smialek JE, Troncoso JC. Intraneuronal abeta-amyloid precedes development of amyloid plaques in Down syndrome. Arch Pathol Lab Med 2001; 125: 489–92. Hafezi-Moghadam A, Thomas KL, Wagner DD. ApoE deficiency leads to a progressive age dependent blood-brain barrier leakage. Am J Physiol Cell Physiol 2007; 292: C1256–62. Hardy J. Has the amyloid cascade hypothesis for Alzheimer’s disease been proved? Curr Alzheimer Res 2006; 3: 71–3. Haroun M, El-Sayed M. Measurement of IgG levels can serve as a biomarker in newly diagnosed diabetic children. J Clin Biochem Nutr 2007; 40: 56–61. Harris PS, Cobbs CG. Cardiac, cerebral, and vascular complications of infective endocarditis. Cardiol Clin 1996; 14: 437–50. Hernandez-Vargas P, Ortiz-Mun˜oz G, Lopez-Franco O, Suzuki Y, Gallego-Delgado J, Sanjuan G, et al. Fcgamma receptor deficiency confers protection against atherosclerosis in apolipoprotein E knockout mice. Circ Res 2006; 99: 1188–96. Jin S, Agerman K, Kolmodin K, Gustafsson E, Dahlqvist C, Jureus A, et al. Evidence for dimeric BACE-mediated APP processing. Biochem Biophys Res Commun 2010; 393: 21–7. Kuang F, Wang BR, Zhang P, Fei LL, Jia Y, Duan XL, et al. Extravasation of blood-borne immunoglobulin G through blood-brain barrier during adrenaline-induced transient hypertension in the rat. Int J Neurosci 2004; 114: 575–91. Li R, Lindholm K, Yang LB, Yue X, Citron M, Yan R, et al. Amyloid beta peptide load is correlated with increased betasecretase activity in sporadic Alzheimer’s disease patients. Proc Natl Acad Sci USA 2004; 101: 3632–7. Lister KJ, Hickey MJ. Immune complexes alter cerebral microvessel permeability: roles of complement and leukocyte adhesion. Am J Physiol Heart Circ Physiol 2006; 291: H694–704. Listı` F, Candore G, Modica MA, Russo M, Di Lorenzo G, EspositoPellitteri M, et al. A study of serum immunoglobulin levels in elderly persons that provides new insights into B cell immunosenescence. Ann N Y Acad Sci 2006; 1089: 487–95. Lu J, Moochhala S, Kaur C, Ling EA. Cellular inflammatory response associated with breakdown of the blood-brain barrier after closed head injury in rats. J Neurotrauma 2001; 18: 399–408. Luo Y, Pollard JW, Casadevall A. Fcgamma receptor cross-linking stimulates cell proliferation of macrophages via the ERK pathway. J Biol Chem 2010; 285: 4232–42. Masliah E, Mallory M, Alford M, DeTeresa R, Hansen LA, McKeel DW Jr, et al. Altered expression of synaptic proteins occurs early during progression of Alzheimer’s disease. Neurology 2001; 56: 127–9. Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, et al. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science 2010; 330: 1774. Methia N, Andre P, Hafezi-Moghadam A, Economopoulos M, Thomas KL, Wagner DD. ApoE deficiency compromises the blood brain barrier especially after injury. Mol Med 2001; 7: 810–5. Morrison JH, Hof PR. Selective vulnerability of corticocortical and hippocampal circuits in aging and Alzheimer’s disease. Prog Brain Res 2002; 136: 467–86. Murai T, Okuda S, Tanaka T, Ohta H. Characteristics of object location memory in mice: Behavioral and pharmacological studies. Physiol Behav 2007; 90: 116–24. Downloaded from by guest on October 21, 2014 Ashe KH, Zahs KR. Probing the biology of Alzheimer’s disease in mice. Neuron 2010; 66: 631–45. Bake S, Friedman JA, Sohrabji F. Reproductive age-related changes in the blood brain barrier: expression of IgG and tight junction proteins. Microvasc Res 2009; 78: 413–24. Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer’s disease. Lancet 2011; 377: 1019–31. Barnes J, Godbolt AK, Frost C, Boyes RG, Jones BF, Scahill RI, et al. Atrophy rates of the cingulate gyrus and hippocampus in Alzheimer’s disease and FTLD. Neurobiol Aging 2007; 28: 20–28. Bell RD, Winkler EA, Sagare AP, Singh I, LaRue B, Deane R, et al. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron 2010; 68: 409–27. Bertram L, Lill CM, Tanzi RE. The genetics of Alzheimer disease: back to the future. Neuron 2010; 68: 270–81. Bjelik A, Bereczki E, Gonda S, Juha´sz A, Rimano´czy A, Zana M, et al. Human apoB overexpression and a high-cholesterol diet differently modify the brain APP metabolism in the transgenic mouse model of atherosclerosis. Neurochem Int 2006; 49: 393–400. Bouras C, Riederer BM, Kovari E, Hof PR, Giannakopoulos P. Humoral immunity in brain aging and Alzheimer’s disease. Brain Res Brain Res Rev 2005; 48: 477–87. Burut DF, Karim Y, Ferns GA. The role of immune complexes in atherogenesis. Angiology 2010; 61: 679–89. Carvalho-Filho RJ, Narciso-Schiavon JL, Tolentino LH, Schiavon LL, Ferraz ML, Silva AE. Central nervous system vasculitis and polyneuropathy as first manifestations of hepatitis C. World J Gastroenterol 2012; 18: 188–91. Colucci-D’Amato L, Perrone-Capano C, di PU. Chronic activation of ERK and neurodegenerative diseases. Bioessays 2003; 25: 1085–95. Crisby M, Rahman SM, Sylven C, Winblad B, Schultzberg M. Effects of high cholesterol diet on gliosis in apolipoprotein E knockout mice. Implications for Alzheimer’s disease and stroke. Neurosci Lett 2004; 369: 87–92. Curtis J, Finkbeiner S. Sending signals from the synapse to the nucleus: possible roles for CaMK, Ras/ERK, and SAPK pathways in the regulation of synaptic plasticity and neuronal growth. J Neurosci Res 1999; 58: 88–95. Diamond B, Huerta PT, Mina-Osorio P, Kowal C, Volpe BT. Losing your nerves? Maybe it’s the antibodies. Nat Rev Immunol 2009; 9: 449–56. Doyle HA, Mamula MJ. Post-translational protein modifications in antigen recognition and autoimmunity. Trends Immunol 2001; 22: 443–9. Eichenbaum H, Yonelinas AP, Ranganath C. The medial temporal lobe and recognition memory. Annu Rev Neurosci 2007; 30: 123–52. Ferna´ndez-Vizarra P, Ferna´ndez AP, Castro-Blanco S, Serrano J, Bentura ML, Martı´nez-Murillo R, et al. Intra- and extracellular Abeta P. Fernandez-Vizarra et al. FcR in sporadic Alzheimer-like disease | 2837 Sato S, Tatebayashi Y, Akagi T, Chui DH, Murayama M, Miyasaka T, et al. Aberrant tau phosphorylation by glycogen synthase kinase-3beta and JNK3 induces oligomeric tau fibrils in COS-7 cells. J Biol Chem 2002; 277: 42060–5. Schmechel A, Strauss M, Schlicksupp A, Pipkorn R, Haass C, Bayer TA, et al. Human BACE forms dimers and colocalizes with APP. J Biol Chem 2004; 279: 39710–7. Selkoe DJ. Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev 2001; 81: 741–66. Shepardson NE, Shankar GM, Selkoe DJ. Cholesterol level and statin use in Alzheimer disease: I. Review of epidemiological and preclinical studies. Arch Neurol 2011a; 68: 1239–44. Shepardson NE, Shankar GM, Selkoe DJ. Cholesterol level and statin use in Alzheimer Disease: II. Review of human trials and recommendations. Arch Neurol 2011b; 68: 1385–92. Song X, Shapiro S, Goldman DL, Casadevall A, Scharff M, Lee SC. Fcgamma receptor I- and III-mediated macrophage inflammatory protein 1alpha induction in primary human and murine microglia. Infect Immun 2002; 70: 5177–84. Turk Z, Ljubic S, Turk N, Benko B. Detection of autoantibodies against advanced glycation endproducts and AGE-immune complexes in serum of patients with diabetes mellitus. Clin Chim Acta 2001; 303: 105–15. Veinbergs I, Mante M, Jung MW, Van UE, Masliah E. Synaptotagmin and synaptic transmission alterations in apolipoprotein E-deficient mice. Prog Neuropsychopharmacol Biol Psychiatry 1999; 23: 519–31. Walter J, Fluhrer R, Hartung B, Willem M, Kaether C, Capell A, et al. Phosphorylation regulates intracellular trafficking of betasecretase. J Biol Chem 2001; 276: 14634–41. Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 2011; 17: 610–17. Yang LB, Lindholm K, Yan R, Citron M, Xia W, Yang XL, et al. Elevated beta-secretase expression and enzymatic activity detected in sporadic Alzheimer disease. Nat Med 2003; 9: 3–4. Zhang SH, Reddick RL, Piedrahita JA, Maeda N. Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E. Science 1992; 258: 468–71. Downloaded from by guest on October 21, 2014 Nakahara J, Tan-Takeuchi K, Seiwa C, Gotoh M, Kaifu T, Ujike A, et al. Signaling via immunoglobulin Fc receptors induces oligodendrocyte precursor cell differentiation. Dev Cell 2003; 4: 841–52. Nakata A, Araki S, Tanigawa T, Miki A, Sakurai S, Kawakami N, et al. Decrease of suppressor-inducer (CD4 + CD45RA) T lymphocytes and increase of serum immunoglobulin G due to perceived job stress in Japanese nuclear electric power plant workers. J Occup Environ Med 2000; 42: 143–50. Nimmerjahn F, Bruhns P, Horiuchi K, Ravetch JV. FcgammaRIV: a novel FcR with distinct IgG subclass specificity. Immunity 2005; 23: 41–51. Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol 2008; 8: 34–47. Oitzl MS, Mulder M, Lucassen PJ, Havekes LM, Grootendorst J, de Kloet ER. Severe learning deficits in apolipoprotein E-knockout mice in a water maze task. Brain Res 1997; 752: 189–96. Okamatsu Y, Matsuda K, Hiramoto I, Tani H, Kimura K, Yada Y, et al. Ghrelin and leptin modulate immunity and liver function in overweight children. Pediatr Int 2009; 51: 9–13. Oreskes I, Mandel D. Size fractionation of thermal aggregates of immunoglobulin G. Anal Biochem 1983; 134: 199–204. Ortiz-Mun˜oz G, Lopez-Parra V, Lopez-Franco O, Fernandez-Vizarra P, Mallavia B, Flores C, et al. Suppressors of cytokine signalling abrogate diabetic nephropathy. J Am Soc Nephrol 2010; 21: 763–72. Ounanian A, Guilbert B, Renversez JC, Seigneurin JM, Avrameas S. Antibodies to viral antigens, xenoantigens, and autoantigens in Alzheimer’s disease. J Clin Lab Anal 1990; 4: 367–75. Pengas G, Hodges JR, Watson P, Nestor PJ. Focal posterior cingulate atrophy in incipient Alzheimer’s disease. Neurobiol Aging 2010; 31: 25–33. Rahman A, Akterin S, Flores-Morales A, Crisby M, Kivipelto M, Schultzberg M, et al. High cholesterol diet induces tau hyperphosphorylation in apolipoprotein E deficient mice. FEBS Lett 2005; 579: 6411–16. Reddy PH, Mani G, Park BS, Jacques J, Murdoch G, Whetsell W Jr, et al. Differential loss of synaptic proteins in Alzheimer’s disease: implications for synaptic dysfunction. J Alzheimers Dis 2005; 7: 103–17. Rodrigo J, Ferna´ndez-Vizarra P, Castro-Blanco S, Bentura ML, Nieto M, Go´mez-Isla T, et al. Nitric oxide in the cerebral cortex of amyloid-precursor protein (SW) Tg2576 transgenic mice. Neuroscience 2004; 128: 73–89. Brain 2012: 135; 2826–2837
© Copyright 2024