Small Intestinal Neuroendocrine Tumor Analyses Somatostatin Analog Effects and MicroRNA Profiling

Digital Comprehensive Summaries of Uppsala Dissertations
from the Faculty of Medicine 1039
Small Intestinal Neuroendocrine
Tumor Analyses
Somatostatin Analog Effects and MicroRNA Profiling
SU-CHEN LI
ACTA
UNIVERSITATIS
UPSALIENSIS
UPPSALA
2014
ISSN 1651-6206
ISBN 978-91-554-9058-4
urn:nbn:se:uu:diva-233207
Dissertation presented at Uppsala University to be publicly examined in Enghoffsalen, Entrance 50, Uppsala University Hospital, Uppsala, Thursday, 20 November 2014 at 09:15 for
the degree of Doctor of Philosophy (Faculty of Medicine). The examination will be conducted
in English. Faculty examiner: Associate Professor Marco Volante (Department of Clinical and
Biological Sciences, University of Turin).
Abstract
Li, S.-C. 2014. Small Intestinal Neuroendocrine Tumor Analyses. Somatostatin Analog
Effects and MicroRNA Profiling. Digital Comprehensive Summaries of Uppsala
Dissertations from the Faculty of Medicine 1039. 48 pp. Uppsala: Acta Universitatis
Upsaliensis. ISBN 978-91-554-9058-4.
Small intestinal neuroendocrine tumors (SI-NETs) originate from serotonin-producing
enterochromaffin (EC) cells in the intestinal mucosa. Somatostatin analogs (SSAs) are mainly
used to control hormonal secretion and tumor growth. However, the molecular mechanisms
leading to the control of SI-NETs are unknown. Although microRNAs (miRNAs) are post
transcriptional regulators deeply studied in many cancers, are not well-defined in SI-NETs.
We adopted a two-pronged strategy to investigate SSAs and miRNAs: first, to provide novel
insights into how SSAs control NET cells, and second, to identify an exclusive SI-NET miRNA
expression, and investigate the biological functions of miRNA targets.
To accomplish the first aim, we treated CNDT2.5 cells with octreotide for 16 months.
Affymetrix microarray was performed to study gene variation of CNDT2.5 cells in the presence
or absence of octreotide. The study revealed that octreotide induces six genes, ANXA1,
ARHGAP18, EMP1, GDF15, TGFBR2 and TNFSF15.
To accomplish the second aim, SI-NET tissue specimens were used to run genome-wide
Affymetrix miRNA arrays. The expression of five miRNAs (miR-96, -182, -183, -196a and
-200a) was significantly upregulated in laser capture microdissected (LCM) tumor cells versus
LCM normal EC cells, whereas the expression of four miRNAs (miR-31, -129-5p, -133a
and -215) was significantly downregulated in LCM tumor cells. We also detected nine tissue
miRNAs in serum samples, showing that the expression of five miRNAs is significantly
increased in SSA treated patients versus untreated patients. Conversely, SSAs do not change
miRNA expression of four low expressed miRNAs. Silencing miR-196a expression was used
to investigate functional activities in NET cells. This experimental approach showed that four
miR-196a target genes, HOXA9, HOXB7, LRP4 and RSPO2, are significantly upregulated in
silenced miR-196a NET cells.
In conclusion, ANXA1, ARHGAP18, EMP1, GDF15, TGFBR2 and TNFSF15 genes might
regulate cell growth and differentiation in NET cells, and play a role in an innovative octreotide
signaling pathway. The global SI-NET miRNA profiling revealed that nine selected miRNAs
might be involved in tumorigenesis, and play a potential role as novel markers for follow-up.
Indeed, silencing miR-196a demonstrated that HOXA9, HOXB7, LRP4 and RSPO2 genes are
upregulated at both transcriptional and translational levels.
Keywords: Small intestinal neuroendocrine tumors, Somatostatin analogs, Laser-capture
microdissection, Microarray profiling and microRNA profiling
Su-Chen Li, Endocrine oncology, Akademiska sjukhuset, Uppsala University, SE-751 85
Uppsala, Sweden.
© Su-Chen Li 2014
ISSN 1651-6206
ISBN 978-91-554-9058-4
urn:nbn:se:uu:diva-233207 (http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-233207)
To my family
不要問我從哪裡來 我的故鄉在遠方
為什麼流浪 流浪遠方 流浪
為了天空飛翔的小鳥
為了山間輕流的小溪
為了寬闊的草原 流浪遠方 流浪
還有還有 為了夢中的橄欖樹橄欖樹
橄欖樹
三毛
List of Papers
This thesis is based on the following papers, which are referred to in the text
by their Roman numerals.
I
Li SC, Martijn C, Cui T, Essaghir A, Luque RM, Demoulin
JB, Castaño JP, Öberg K, Giandomenico V. The somatostatin
analogue octreotide inhibits growth of small intestine
neuroendocrine tumour cells. PLoS One. 2012. 7(10):e48411.
II
Li SC, Martijn C, Essaghir A, Lloyd RV, Demoulin JB, Öberg
K, Giandomenico V. Global microRNA profiling of welldifferentiated small intestinal neuroendocrine tumors. Modern
Pathology 2013; 26:685-696.
III
Li SC, Khan M, Caplin M, Meyer T, Öberg K, Giandomenico
V. Circulating microRNA detection in small intestinal
neuroendocrine tumor patients treated with somatostatin
analogs. Submitted manuscript.
IV
Li SC, Shi H, Khan M, Caplin M, Meyer T, Öberg K,
Giandomenico V. Functional role of miR-196a in neuroendocrine tumors. Submitted manuscript.
Reprints were made with permission from the respective publishers.
I
II
Open access under the terms of the Creative Commons
Attribution (CC BY) License.
Open access under the terms of Creative Commons
Attribution-Noncommercial-No Derivative Works Unported
License (CC BY-NC-ND).
Additional publications
I
II
III
Cui T, Hurtig M, Elgue G, Li SC, Veronesi G, Essaghir A,
Demoulin JB, Pelosi G, Alimohammadi M, Öberg K, Giandomenico
V. Paraneoplastic antigen Ma2 autoantibodies as specific blood
biomarkers for detection of early recurrence of small intestine
neuroendocrine tumors. PLoS One. 2010 Dec 30; 5(12):e16010.
Cui T*, Tsolakis AV*, Li SC, Cunningham JL, Lind T, Öberg K,
Giandomenico V. Olfactory receptor 51E1 protein as a potential
novel tissue biomarker for small intestine neuroendocrine
carcinomas. Eur J Endocrinol. 2013 Jan 17; 168(2):253-61.
Darmanis S, Cui T, Drobin K, Li SC, Öberg K, Nilsson P, Schwenk
JM, Giandomenico V. Identification of candidate serum proteins for
classifying well-differentiated small intestinal neuroendocrine
tumors. PLoS One. 2013 Nov 25; 8(11):e81712.
Supervisors
Associate Professor Valeria Giandomenico, PhD
Department of Medical Sciences, Endocrine Oncology, Uppsala University,
Uppsala, Sweden
Professor Emeritus Kjell Öberg, MD, PhD
Department of Medical Sciences, Endocrine Oncology, Uppsala University,
Uppsala, Sweden
Chair
Professor Gunnar Westin, PhD
Department of Surgical Sciences, Endocrine Surgery, Uppsala University, Uppsala,
Sweden
Faculty Opponent
Associate Professor Marco Volante, MD, PhD
Department of Clinical and Biological Sciences, University of Turin,
San Luigi Hospital, Turin, Italy
Committee members
Professor Aristidis Moustakas, PhD
Department of Medical Biochemistry and Microbiology, Uppsala University,
Uppsala, Sweden
Associate Professor Johan Lennartsson, PhD
Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden
Associate Professor Rainer Heuchel, PhD
Pancreas Research Lab, Karolinska Institute, Huddinge, Sweden
Contents
Introduction ................................................................................................... 11
Gastrointestinal neuroendocrine tumors ................................................... 11
Neuroendocrine cells ........................................................................... 11
Gastrointestinal endocrine cells ........................................................... 11
Neuroendocrine tumor overview ......................................................... 11
Diagnosis of GI-NETs ......................................................................... 12
The management of GI-NETs ............................................................. 13
Somatostatin ............................................................................................. 14
Somatostatin and somatostatin receptors ............................................. 14
Somatostatin analogs ........................................................................... 16
MicroRNA................................................................................................ 17
MicroRNA overview ........................................................................... 17
Prediction of miRNA targets ............................................................... 19
Functional validation of miRNA targets.............................................. 19
Aims of the study .......................................................................................... 21
Materials and Methods .................................................................................. 22
Human NET cell lines (Papers I and IV) ................................................. 22
Human tissue specimens and human serum samples ............................... 22
Tissue specimens ................................................................................. 23
Serum specimens ................................................................................. 23
Gene and miRNA expression ................................................................... 23
RNA isolation and cDNA synthesis .................................................... 23
Quantitative real-time PCR (QRT-PCR) ............................................. 24
Northern blot analysis (Paper II) ......................................................... 24
Microarray data analysis ...................................................................... 24
MicroRNA array data analysis ............................................................ 25
Protein expression .................................................................................... 25
Western blot analysis (Papers I and IV) .............................................. 25
Immunohistochemistry (Paper I) ......................................................... 25
Laser capture microdissection of SI-NET cells (Papers I and II) ............. 26
Cell proliferation assay............................................................................. 26
Network analysis of microRNA target genes (Paper III) ......................... 26
Transfection (Paper IV) ............................................................................ 27
Statistical analysis (Papers I, II, III and IV) ............................................. 27
Results and Discussion ................................................................................. 28
Paper I: Octreotide inhibits growth of CNDT2.5 cells ............................ 28
Paper II: Global miRNA profiling of SI-NETs ........................................ 30
Paper III: Serum miRNA expression of SI-NETs .................................... 32
Paper IV: MiR-196a target genes in NET cells ........................................ 35
Concluding Remarks and Perspectives ......................................................... 37
Acknowledgements ....................................................................................... 40
References ..................................................................................................... 42
Abbreviations
5-HIAA
AC
Ala
ANXA1
ARHGAP18
Asn
BMP4
Ca2+
cAMP
C. elegans
CTNNB1
C-terminus
Cys
D cell
DMEM
DMSO
EC
ECL
EMP1
ETS1
FBS
FZD
GDF15
GI
Gly
GPCR
HD
HMGA2
HOX
kDa
Ki-67
LAN
LCM
LM
LNM
5-hydroxyindoleacetic acid
Adenylyl cyclase
Alanine
Annexin A1
Rho GTPase-activating protein 18
Asparagine
Bone morphogenetic protein 4
Calcium ion
Cyclic adenosine mono-phosphate
Caenorhabditis elegans
Catenin (Cadherin-associated protein), Beta 1
Carboxyl-terminus
Cysteine
Delta cell
Dulbecco’s Modification of Eagle’s Medium
Dimethyl Sulfoxide
Enterochromaffin
Enhanced chemoluminescence
Epithelial membrane protein 1
V-Ets Avian Erythroblastosis Virus
Fetal bovine serum
Frizzled
Growth/differentiation factor 15
Gastrointestinal
Glycine
G-protein-coupled receptors
Healthy donor
High mobility group AT-hook 2
Homeobox
Kilo Dalton
Antigen KI-67
Lock nucleic acids
Laser capture microdissection
Liver metastases
Lymph node metastases
LRP
Lys
NET
NT
MAPK
MAS
miRNA
MirOB
MM
mRNA
N-terminus
One-Way ANOVA
Phe
Pri-miRNA
Pre-miRNA
PT
PTPase
QRT-PCR
RECIST
RISC
RNase
RSPO
SD
Ser
SI
SSA
SST
SSTR
TGFBR2
Thr
TME
TNFSF15
TNM
Trp
Tyr
UCL
UTR
UU
Val
Vs
WHO
Low density lipoprotein receptor-related protein
Lysine
Neuroendocrine tumor
Nucleotides
Mitogen-activated protein kinases
Molecule Annotation System
MicroRNA
MicroRNA OncoBase
Mesentery metastases
Messenger RNA
Amine-terminus
One-way analysis of variance
Phenylalanine
Primary microRNA
MicroRNA precursors
Primary tumor
Phosphotyrosine phosphatase
Quantitative real-time PCR
Response Evaluation Criteria in Solid Tumors
RNA-induced silencing complex
Ribonuclease
R-spondin
Standard deviation
Serine
Small intestine
Somatostatin analog
Somatostatin
Somatostatin receptor
TGF-beta type II receptor
Threonine
Tumor microenvironment
Tumor necrosis factor superfamily member 15
Tumor node metastasis
Tryptophan
Tyrosine
University College of London
Untranslated region
Uppsala University Hospital
Valine
Versus
World Health Organization
Introduction
Gastrointestinal neuroendocrine tumors
Neuroendocrine cells
Neuroendocrine cells are widely distributed in various parts of the human
body, including the central and peripheral nervous systems, thyroid,
gastrointestinal and respiratory tracts, pancreas and skin. They are also
known as neurosecretory cells, which release hormones into the blood to
affect their endocrine targets [1,2]. The neuroendocrine system has a
functional interaction between the endocrine system and the central and
peripheral nervous systems, which are incorporated into the diffuse
neuroendocrine system, according to their biological function [3]. These
cells are therefore able to produce a variety of bioactive products, and their
secretion is then regulated by G-protein-coupled receptors (GPCR), iongated receptors and receptors with tyrosine-kinase activity [4,5].
Gastrointestinal endocrine cells
Gastrointestinal (GI) endocrine cells are differentiated epithelial cells
diffused among mucosa cells in the GI tract. They contain regulatory peptide
hormones and/or biogenic amines. The GI tract contains at least 14 different
kinds of neuroendocrine cell types. They produce a variety of bioactive
peptides or amines [6]. Among them, serotonin is the main secretory product
of the enterochromaffin (EC) cells, and it represents more than 90% of all
serotonin synthesized in the body. Serotonin is produced from tryptophan,
by hydroxylation and decarboxylation in the cytoplasm of EC cells, and is
then transported into the secretory granules by a transport protein called the
vesicular monoamine transporter [7].
Neuroendocrine tumor overview
Neuroendocrine tumors (NETs) are a rare disease. The tumors are often
small and indolent, and show no specific symptoms during the early disease
stage. The lack of sensitive and specific methods for early clinical detection
results in any diagnosis being delayed by up to several years. Most patients
thus present metastatic disease in various organs at the time of diagnosis.
11
Metastases are often found in regional lymph nodes and liver [8,9]. The first
World Health Organization (WHO) classification of NETs was reported in
1980 and then updated in 2000, 2004 and 2010 [10,11]. NETs can be
categorized into well-differentiated NETs with either low or intermediate
grade, and poorly differentiated NETs with high grade [10,12]. The tumor
node metastasis (TNM) system classification guidelines from the European
Neuroendocrine Tumor Society are also relevant for the staging and grading
of NETs, and are based on the WHO system [13,14].
GI-NETs occur in a variety of organs from different cell types, which are
characterized as hormone-producing cells. These tumor cells have the ability
to produce and release hormones into the body that affect the normal
functions of the organs and might result in different hormonal syndromes.
GI-NETs are divided into functioning or nonfunctioning tumors according to
secretory symptoms. Functioning GI-NETs often release bioactive products,
such as serotonin, into the systemic circulation, that are associated with
carcinoid syndrome. Symptoms include flushing, diarrhea and abdominal
pain. In contrast, non-functioning GI-NETs do not develop any distinct
hormonal syndrome [5,13].
Diagnosis of GI-NETs
GI-NETs can be diagnosed by clinical symptoms, hormone levels,
pathology, and radiological and nuclear imaging. Biochemical GI-NET
markers are divided into general and specific markers. General markers,
such as chromogranins, pancreatic polypeptide, neuro-specific enolase and
subunits of glycoproteins, are used to detect diverse clinical hormone-related
syndromes [13,15]. Specific markers including immuno-histochemical tumor
markers, chromo-granin A and synaptophysin, are routinely used today [16].
When patients present with carcinoid syndrome, urinary levels of 5-hydroxyindoleacetic acid (5-HIAA) are increased. The 5-HIAA is a final serotonin
metabolite commonly used to identify NETs, generally small intestinal
NETs (SI-NETs). In addition, the proliferation marker antigen KI-67 (Ki-67)
is used to determine tumor prognosis and the lower proliferation indicated by
Ki-67 correlates with longer survival [17]. Imaging is commonly used to
identify the tumor position in the body and the stage of disease. Today, a
variety of techniques, such us computed tomography, magnetic resonance
image scans, positron emission tomography, endoscopic ultrasonography
and somatostatin receptor scintigraphy, are used specifically to localize
NETs [8,18].
12
The management of GI-NETs
The management of GI-NETs depends on the disease stage and the
symptoms related to the hormonal secretion. The ideal treatment option is
surgery during the primary stage of the disease. This means that surgery
should be considered as the first-line therapy [13,18]. However, curative
surgery is unavailable to the majority of patients, who usually present with
advanced disease at the time of diagnosis. Therefore, palliative therapies,
such as somatostatin analogs (SSAs) and interferon alpha are used to reduce
symptoms and inhibit tumor growth for NET patients [13,19,20]. Recently
new drugs, such as everolimus and sunitinib, have also been used to inhibit
tumor growth. Everolimus is a mammalian target of rapamycin inhibitor that
is mainly used in controlling cell growth, angiogenesis and cell metabolism
[21]. Sunitinib is a tyrosine-kinase inhibitor and its main actions are
inhibition of tumor angiogenesis and antitumor effect [22,23]. Although the
clinical response to chemotherapy is often ineffective on indolent
differentiated rare tumors, combined chemo-therapy using temozolomide
along with either capecitabine or bevacizumab has recently become an
effective and innovative therapy for poorly differentiated NETs [13,18]. The
response of NETs to radionuclide therapy is limited. However, peptide
receptor radio-therapy has shown positive effects in patients with
unresectable somatostatin receptor-positive tumors [13,24].
13
Somatostatin
Somatostatin and somatostatin receptors
Somatostatin (SST) was initially isolated from the hypothalamus and
recognized as an inhibitor factor of growth hormone release [25]. SST is also
present in the SST cells, named D-cells that produce SST [26,27]. SST is
released into two distinct types of polypeptide chains. The first one is SST14, which comprises the 14-amino-acids form and the second one is SST-28,
a chain of 28 amino-acids. SST-14 contains a disulfide bridge which allows
its cyclic structure (Fig. 1A), and plays important roles in glucagon
inhibition and gastric acid secretion [26,27]. SST-28 is a specific growth
hormone inhibitor that inhibits insulin release. Indeed, gastrin produced by
G-cells in the GI tract is able to reduce pancreatic secretion of insulin
[26,28].
Figure 1. Molecular structure of SST-14 and SSA. (A) SST-14 consists of 14
amino acids with a disulfide bridge to form the cyclic structure. The amino acid
residues, Phe, Trp, Lys and Thr, are required to interact with the somatostatin
receptors. (B) Octreotide changes its form from Trp to D-Trp. (C) Lanreotide
changes Phe to Tyr and Thr to Val. Ala = alanine; Asn = asparagine; Cys = cysteine;
Gly = glycine; Lys = lysine; Phe = phenylalanine; Ser = serine; Thr = threonine; Trp
= tryptophan; Tyr = tyrosine; Val = Valine. The figure is modified from Modlin et
al., 2010 [29].
14
SST-14 and SST-28 exert their functions by binding to five somatostatin
receptors (SSTRs) with high and specific affinity. These receptors belong to
the GPCR family which comprises seven trans-membrane domain proteins
[30]. The five SSTRs are encoded by five different genes named SSTR1,
SSTR2, SSTR3, SSTR4 and SSTR5. SST binds to SSTRs to initiate a complex
signaling pathway, which is firstly triggered by the interaction of activated
G-protein though binding to SST [31]. The reduction of cyclic adenosine
mono-phosphate and calcium ion (Ca2+) then results in the inhibition of
hormone secretion (Fig. 2) [32,33]. Moreover, phosphotyrosine phosphatase
(PTPase) can be activated by G-proteins and generate cytostatic actions,
such as cell growth and apoptosis control (Fig. 2) [34,35].
Figure 2. Multifaceted signaling pathways are activated by SST binding to
SSTRs. SST inhibits hormone secretion by reducing cAMP and calcium ion (Ca2+).
PTPase is activated by G-proteins leading to cytostatic actions, such as cell growth
and apoptosis. AC = adenylyl cyclase; cAMP = cyclic adenosine mono-phosphate;
Gα, Gβ and Gγ = G protein subunit; MAPK = mitogen-activated protein kinases;
PTPase = phosphotyrosine phosphatase. The figure is modified from Ferone et al.,
2009 and Öberg et al., 2010 [36,37].
15
Somatostatin analogs
Physiological SST is not used for clinical therapy, due to its rapid
degradation and a short half-life. Therefore, synthetic SSAs have been
developed as potential clinical drugs. They are SST-like structures with a
shorter peptide length, maintaining the conserved region for biological
activity (Fig. 1A). Although the native SST has a naturally higher affinity to
all the five SSTRs, synthetic SSAs increased their half-life from 2-3 minutes
to approximately 90 minutes [38]. Octreotide and lanreotide are two SSAs in
common clinical use. Compared to SST, octreotide is an octapeptide,
including D-Phe at the N-terminus, L-Thr at the C-terminus, but with D-Trp
replaced from L-Trp in the conserved region (Fig. 1B) [29]. Lanreotide is
also an octapeptide comprising D-βNal at the N-terminus, L-Thr at the Cterminus, but where L-Trp is replaced by D-Trp, Phe by Tyr and Thr by Val
in the conserved region (Fig. 1C) [29].
Abdominal pain, diarrhea, nausea and vomiting often occur as SSA side
effects. The majority of NET patients discontinue SSA treatment within
months. This phenomenon is named tachyphylaxis, and might be due to the
fact that patients become unresponsive to the SSA treatment after a certain
period of time. The potential mechanism of the secretion inhibition
desensitization of tumor correlated hormones is unknown [29].
Octreotide was the first SSA approved by the Food and Drug
Administration for functioning NETs with hormone-related symptoms. The
drug has a high affinity to SSTR2 and SSTR5, medium affinity to SSTR3
but low affinity to SSTR1 and SSTR4 [29,37,39]. A related clinical trial
recently demonstrated that octreotide is also able to inhibit tumor growth
[40]. Octreotide is also used in nuclear medicine imaging. New clinical
approaches include the use of SST-based radio-peptides, which have a high
affinity to SSTR2 [37].
16
MicroRNA
MicroRNA overview
MicroRNAs (miRNAs) are small non-coding RNA of 18-25 nucleotides (nt),
which were first identified from Caenorhabditis elegans (C. elegans) by
Ambrose et al., in 1993 [41]. The group found that lin-4 does not encode a
protein but is pivotal in C. elegans development. Two short lin-4 transcripts
sequence (22 and 61 nt) are complementary to a repeated sequence in the 3’
untranslated region (UTR) of lin-14 messenger RNA (mRNA) that results in
lin-4 repression of lin-14 translation through an antisense RNA-RNA
interaction [41].
MiRNAs originate in the nucleus and are transcribed by RNA polymerase
II or polymerase III into a longer precursor known as primary miRNA (primiRNA), which comprises several stem-loop structures [42,43]. Pri-miRNA
is then cleaved by RNase III enzyme Drosha to release a second precursor
of circa 70 to 90 nt, named miRNA precursor (pre-miRNA) [44]. The
process continues with the exportation of pre-miRNAs into cytoplasm by
exportin-5 protein and pre-miRNAs are then cleaved to release doublestranded mature miRNAs of between 19 nt and 24 nt by the RNase III
enzyme Dicer [45,46]. One strand of the double-stranded mature miRNA is
associated with RNA-induced silencing complex (RISC), which then binds
to target mRNA to trigger the cellular effects. The miRNA biogenesis is
shown in Fig. 3 [47].
The function of miRNAs appears to be in gene regulation. An mRNA is
transcribed from the DNA and is later translated into protein. The 3’-UTR is
the section of mRNA that immediately follows the translation termination
codon. Several regions of the mRNA are not translated into protein including
the 5’ cap, 5’-UTR, 3’-UTR, and the poly (A) tail. Animal miRNAs are
usually complementary to a site in the 3’-UTR, whereas plant miRNAs are
usually complementary to coding regions of mRNAs. In animals miRNAs
more often have only part of the right sequence of nucleotides to bond with
the target mRNA, known as imperfect matched. Although partially
complementary miRNAs are common, nucleotides 2–8 of the miRNA
(called seed region) still have to be perfectly complementary in order to
recognize their targets. In conclusion, partial complementarity between
miRNAs and their target 3’-UTR induce translational repression of target
mRNAs, whereas perfect complementary between miRNAs and their targets
induce RISC to cleave target mRNAs [48].
17
Figure 3. Biogenesis of miRNAs. MiRNAs are first transcribed into pri-miRNA by
RNA polymerases. This produces several hairpin structures in the nucleus, which are
cleaved by Drosha to release a stem loop of 70-90 nt, called pre-miRNAs. The premiRNAs are exported to the cytoplasm by exportin-5 protein, and then cleaved by
Dicer to release a 19-24 nt mature miRNA that is incorporated into RISC. The
mature miRNA leads RISC to either cleave the mRNA or induce translational
repression according to the degree of complementarity. The figure is modified from
He et al., 2004 [47].
Several studies have shown that miRNAs play pivotal roles in cell
proliferation, development, differentiation and apoptosis [49-53]. Moreover,
miRNAs function either as tumor suppressors or oncogenes [54,55]. Indeed,
a downregulated miRNA may act as a tumor suppressor, while an
upregulated miRNA may act as an oncogene in cancer [56]. MiRNA
expression profiling of healthy and diseased specimens showed different
levels of miRNA expression in the two groups thus revealing insights into
the development of a variety of diseases. Based on the deregulation of
miRNA expression in cancer, miRNA expression profiles may be used to
explore biomarkers for cancer diagnosis, prognosis and response to
treatment [57]. Although the majority of miRNAs are intracellular, some of
them have been found outside the cells, such as in body fluids, mainly in
serum and plasma. The majority of miRNAs have the capacity to resist
natural ribonuclease (RNase) activity [58,59]. In addition, miRNAs may be
used as targets for molecular therapy using two main strategies, including
miRNA inhibition/silencing and miRNA mimic/over-expression. AntimiRNA oligonucleotides can be used to compete with the interactions of
miRNAs and cause their target miRNA depletion, whereas viral or liposomal
delivery systems are able to increase miRNA expression levels, which might
restore tumor inhibition functions in cancer cells [60-62].
18
Prediction of miRNA targets
Prediction of miRNA targets in animals is more complicated than in plants.
The interaction between miRNA and mRNA is almost perfectly
complementary in plants, whereas it is partially complementary in animals
[63]. Computational analyses show that a single miRNA may regulate
several hundred targets and a single mRNA may be controlled by more than
one miRNA [64]. Recently, several computational programs have been
developed to predict miRNA targets, such as DIANA LAB, PicTar, miRDB
and TargetScan [65]. The principles of miRNA target prediction are quite
comparable by using different approaches. For example in animal miRNA
detection, the miRNA sequence is complementary to the 3’ UTR sequence
of its target genes. In particular, target mRNA binds to the conserved
complementary 5’ miRNA at the seed region nucleotide position between
two and eight. In addition, prediction criteria include the use of free energy
between miRNAs and their target genes. The predicted efficacy between
miRNAs and mRNAs can be calculated by using RNA folding programs
[65,66].
Functional validation of miRNA targets
MiRNAs regulation role is correlated to the biological functions of their
targets. A single miRNA is predicted to target a large amount of mRNAs and
a single target is also regulated by more than one miRNA [64]. Therefore, it
is important to investigate the biological functions of miRNAs and their
targets and to filter out the false positive prediction results. The typical
approach is to investigate whether miRNAs are able to alter the mRNA
expression levels of their target genes. This means that either miRNA overexpression or miRNA loss-function is used to study the biological functions
of target genes.
MiRNA over-expression can be achieved in vitro by transfection of the
chemically synthesized double strand RNA precursors that might mimic
endogenous miRNAs [67]. MiRNA expression plasmids and viral vectors
are used to produce numerous miRNAs. Introducing miRNA precursor into
DNA plasmids is required for producing mature miRNAs [68]. The adenoassociated virus and lentivirus delivered systems were developed to improve
transfection efficiency [69,70]. The reporter system is also often used to
confirm the functions of miRNAs by either increasing or decreasing reporter
translational expression levels [66].
Gene knockout and antisense techniques are also applied to silence
miRNA functions [71]. Oligoribonucleotides complementary to miRNAs
inhibit miRNA expression. Indeed, the 2’-O-methyl modification enables
irreversible small RNA function inhibition [72]. Moreover, lock nucleic
acids (LANs) are conformational restricted nucleotide analogs that increase
19
the affinity of oligonucleotides to their complementary DNA or RNA.
Therefore, LANs can be used to modify oligoribonucleotides to silence
miRNA expression [73]. In addition, miRNA sponges and miRNA-masks
are used as miRNA antisense oligonucleotides. The miRNA sponges contain
multiple sites for tandem-binding to miRNA targets. Sponges repress
miRNA targets when vectors encoding the sponges are transfected into
cultured cells. The expression of sponges from stably integrated transgenes
may be used for studying miRNA functions in vivo [74,75]. Unlike miRNA
sponges, miRNA-masks comprise single-stranded 2’-O-methyl-modified
antisense oligonucleotides that are perfect complementary to 3’-UTR of
predicted miRNA targets. Therefore, miRNA-masks occupy the miRNAbinding site to inhibit miRNA functions [76].
20
Aims of the study
The overall aim of my study was to investigate somatostatin analog effects
and the potential roles of microRNAs in small intestinal neuroendocrine
tumors in order to improve patient management and disease monitoring.
The specific aims of the study are elucidated in the papers included:
Paper I. To provide fresh insights into how somatostatin analogs control
neuroendocrine tumor cells using CNDT2.5 cells as a novel in vitro model.
Paper II. To identify an exclusive microRNA profile of small intestinal
neuroendocrine tumors, as this might play a critical role in SI-NET
progression.
Paper III. To investigate the expression of the selected microRNAs from
Paper II by using serum samples to extend our findings from tissue
specimens to blood samples.
Paper IV. To explore the biological functions of miR-196a target genes
using CNDT2.5 and NCI-H727 NET cells as in vitro models. The final goal
was to model a microRNA biological functions investigation system that
might be applied and to perform analyses on other microRNAs.
21
Materials and Methods
The majority of the materials and methods included in this thesis are fully
described in the papers listed. However, the most important methods used
during the experimental work are briefly described as follows.
Human NET cell lines (Papers I and IV)
The human SI-NET cells CNDT2.5 and KRJ-1, derived from midgut
carcinoids, were cultured in a 1:1 mixture of Dulbecco’s Modification of
Eagle’s Medium (DMEM) and Ham’s F-12 medium supplemented with 10%
of fetal bovine serum (FBS), 1% penicillin-streptomycin, 1% sodium
pyruvate, 1% MEM vitamin solution, 1% L-glutamine, 1% HEPES buffer
and 1% nonessential amino acids. The human endocrine pancreatic
carcinoma cells QGP-1, derived from pancreatic islet cell carcinoma; the
human atypical bronchial carcinoid cells NCI-H720, and the human typical
bronchial carcinoid cells NCI-H727, were cultured in RPMI 1640 medium
supplemented with 10% FBS, 1% penicillin-streptomycin and 1% Lglutamine. The cells were cultured at 37˚C and a 5% CO2-humidified
atmosphere. All the culture reagents were from Life Technologies, Carlsbad,
CA, USA.
Human tissue specimens and human serum samples
The human tumor specimens (Papers I and II) and serum samples (Papers III
and IV) were collected at Uppsala University Hospital, Uppsala, Sweden,
and were approved by the regional Ethics Committee at Uppsala University
Hospital (Dnr 2011/426). The human serum samples (Papers III and IV)
collected at University College of London, London, UK, were approved by
the London Local Ethics Committee and all donors provided written
informed consent. Eligible patients had pathologically confirmed NETs
categorized according to their primary site of origin with metastatic disease
measurable by Response Evaluation Criteria in Solid Tumors (RECIST).
22
Tissue specimens
Thirty snap-frozen specimens from SI-NET patients were used to isolate
total RNA from laser capture microdissected tumor cells (Papers I and II).
Twelve formalin–fixed, paraffin-embedded SI-NET tissues were used for
immunohistochemistry analysis (Paper I).
Serum specimens
Seven healthy donor (HD) serum samples and 42 SI-NET serum samples,
including 14 primary tumor (PT), 14 lymph node metastases (LNM) and 14
liver metastases (LM) were collected at Uppsala University Hospital (UU).
Three HD serum samples and six SI-NET serum samples with LM were
collected at University College of London (UCL). All the serum samples
were used to detect miRNA expression and miR-196a target gene expression
by QRT-PCR analysis (Paper III and IV).
Gene and miRNA expression
RNA isolation and cDNA synthesis
The Ambion PARIS kit was used to isolate total RNA from human NET
cells (Paper I). The RNAqueous-Micro Kit was used to prepare total RNA
from laser capture microdissected tumor cells (Papers I and II). The mirVana
miRNA kit was used to isolate total RNA from tissue specimens (Papers II
and IV). The mirVana PARIS Kit was also used to isolate total RNA from
serum samples (Papers III and IV). All the RNA isolation kits were from
Life Technologies. Total RNA was converted to cDNA with the iScript
cDNA synthesis Kit (Bio-Rad, Hercules, CA, USA) (Papers I and IV). Total
RNA, including miRNA, was converted to cDNA by the QuantiMir RT Kit
(System Biosciences, Mount View, CA, USA) (Papers II and IV). Total
RNA of serum samples was reverse transcribed to cDNA using the TaqMan
miRNA Reverse Transcription Kit (Life Technologies) (Paper III).
23
Quantitative real-time PCR (QRT-PCR)
The Brilliant SYBR Green QPCR Master Mix (Agilent Technologies,
Waldbronn, Germany) was used to detect mRNA level (Paper I) and miRNA
level (Paper II) from NET cells and tissue specimens. TaqMan Universal
PCR Master Mix II without UNG (Life Technologies) was used to detect
miRNA expression from serum samples (Paper III) and SsoAdvanced
Universal SYBR Green Supermix (Bio-Rad) was used to detect gene
expression of serum samples (Paper IV). The QRT-PCR analyses were
performed by using Stratagene Mx3005P real-time PCR System (Agilent
Technologies).
Northern blot analysis (Paper II)
Northern blot analysis was performed with the mirVana miRNA detection
kit (Life Technologies) that is a hybridization-based solution to analyze
miRNA expression. Total RNA from snap-frozen SI-NET specimens was
incubated with a 32[P]-labeled RNA probe (Perkin Elmer, Boston, MA,
USA) followed by RNase digestion. The radioisotope-labeled RNA
fragments were prepared by using the mirVana miRNA Probe Construction
Kit (Life Technologies). The radiolabeled RNA samples were analyzed on
15% TBE-Urea gels (Bio-Rad) and detected by Fuji radiography film
(Fujifilm, Tokyo, Japan). The developed radiography films of northern blot
analyses were scanned for semi-quantitative analysis using the Molecular
Imager ChemiDoc XRS+ System (Bio-Rad) and Image Lab Software (BioRad).
Microarray data analysis
Total RNA was hybridized onto the Affymetrix Human Gene 1.0 ST Array
(Affymetrix, Santa Clara, CA, USA) by Uppsala Array Platform, Uppsala
University Hospital, Uppsala, Sweden. Scanned images of microarray chips
were analyzed by GeneChip Operating Software (Affymetrix). The
hierarchical clustering algorithm was applied to a group of genes and
samples according to similarities in expression. Genes differentially
expressed were clustered using Euclidian distance with average linkage
clustering (genes and samples). Gene function based on gene ontology
analysis was performed by using the Molecule Annotation System (MAS)
version 3.0 (www.bioinfo.capitalbio.com/).
24
MicroRNA array data analysis
Total RNA was hybridized onto the Affymetrix GeneChip miRNA 1.0 Array
(Affymetrix) by the Bioinformatics and Expression Analysis core facility,
Karolinska Institute, Huddinge, Sweden. Scanned images of microarray
chips were analyzed using the Affymetrix miRNA QC Tool (Affymetrix).
The raw data have been normalized by using a quantile algorithm.
Expression levels in the different groups were compared by using the twosided unpaired Student’s t-test and false discovery rates (q-values) were
estimated by means of the q-value package in R (www.r-project.org). The
hierarchical clustering was performed by using the MeV software
(www.tm4.org). The hierarchical clustering algorithm was applied,
according to similarities in expression, to the group of miRNAs from all
samples, which were clustered according to Pearson correlation distance.
Protein expression
Western blot analysis (Papers I and IV)
Whole-cell protein lysates were extracted by using radio-immunoprecipitation assay buffer. Protein concentrations were determined using
Coomassie-Plus Better Bradford Assay (Thermo Scientific, Rockford, IL,
USA). Protein lysates were resolved by either 7.5% Mini-PROTEA TGX
precast gels or any kDa Mini-PROTEA TGX precast gels (Bio-Rad)
according to the protein molecular weight, and then transferred to 0.45-µm
nitrocellulose membranes (Bio-Rad). Precision Plus Protein Dual Color
Standards (Bio-Rad) was used to calculate and confirm the apparent size of
proteins. The membranes were blocked by Western Blocking Reagent
(Roche Applied Science) overnight and then blotted with the primary
antibody overnight at 4˚C. They were then washed, incubated with the
horseradish peroxidase conjugated secondary antibody, and washed again.
The blots were visualized by using Lumigen ECL Ultra TMA-6 (Beckman
Coulter, Brea, CA, USA) and the semi-quantitative analysis by the
Molecular Imager ChemiDoc XRS+ System (Bio-Rad) and Image Lab
Software (Bio-Rad).
Immunohistochemistry (Paper I)
Paraffin-embedded tissue slides were used to investigate the differentially
expressed markers. After staining, the slides were evaluated by Axiophot
light microscope and AxioVision Rel.4.5 software (Carl Zeiss AG,
Oberkochen, Germany).
25
Laser capture microdissection of SI-NET cells (Papers I
and II)
Snap-frozen specimens of SI-NETs were cut to 8-µm sections by a
microtome cryostat (Leica, Wetzlar, Germany) and then adhered to
polyethylene naphthalate membrane frame slides (Life Technologies).
Tumor cells were collected with the ArcturusXT Microdissection System
(Life Technologies).
Cell proliferation assay
The metabolic proliferation reagent WST-1 (Roche Applied Science,
Mannheim, Germany) was used to measure cell proliferation rate in Paper I.
The principle of this assay relies on the cleavage of the stable tetrazolium
salt WST-1 to a soluble formazan by a complex enzymatic cellular
mechanism. This bio-reduction depends mainly on the glycolytic production
of NAD (P) H in viable cells. Thus, the amount of formazan dye formed
correlates directly to the number of metabolically active cells in the culture
and the concentration determined by optical density at 450 nm. The Vybrant
MTT cell proliferation assay (Life Technologies) was also used to detect cell
growth rate in Paper IV. The principle of this assay relies on the conversion
of the water soluble 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium
bromide to an insoluble formazan. The formazan is then solubilized by using
dimethyl sulfoxide (DMSO) and the concentration determined by optical
density at 570 nm. Human NET cells were seeded in 96-well plates with 100
µL per well and then the cells were cultured for up to seven days. The
absorbance of the samples against a background control (medium) as blank
was measured by using a Multiskan Ascent microplate ELISA reader
(Thermo Scientific, Rockford, IL, USA).
Network analysis of microRNA target genes (Paper III)
Cytoscape software (version 3.1.1) was used to analyze the global network
between miRNAs and miRNA-regulated genes. MicroRNA OncoBase
(MirOB) (www.mirob.interactome.ru/) software was used to identify
biologically relevant gene networks and any potential miRNA target gene’s
ontology information.
26
Transfection (Paper IV)
Anti-196a, anti-let-7, used as positive controls and miRNA antisense
oligonucleotides negative control (Life Technologies, Carlsbad, CA, USA)
were used to transfect NET cells. MiR-196a antisense are chemically
modified, single-stranded oligonucleotides specifically designed to bind to
and inhibit endogenous miR-196a resulting in artificial upregulation of target
mRNA translation. Endogenous let-7 induces HMGA2 gene downregulation.
Therefore, introduction of the anti-let-7 may induce a significant
upregulation in the HMGA2 gene. MiRNA antisense oligonucleotides
negative control has a unique sequence design that does not target any
human genes and has no measurable effects on known miRNA functions.
The miRNA inhibition experiment was performed by the reverse transfection
procedure, using Lipofectamine RNAiMAX (Life Technologies).
Transfected NET cells were incubated in 5% CO2 and a humidified
atmosphere at 37°C for 24 and 48 hours.
Statistical analysis (Papers I, II, III and IV)
The statistical significance of the difference between two groups was
evaluated by two-tailed Student’s t-test. The statistical significance of the
difference in more than two groups was evaluated by either One-Way
ANOVA or Two-Way ANOVA followed by either Dunnett’s test or the
Bonferroni test using GraphPad Prism 5 (Graph Pad, Software, La Jolla CA,
USA); p value < 0.05 is considered significant.
27
Results and Discussion
Paper I: Octreotide inhibits growth of CNDT2.5 cells
Octreotide is a widely used synthetic SSA that significantly improves the
management of NETs and it acts through SSTR signaling. CNDT2.5 cells
were treated with 1 µM of octreotide from 1 day up to 16 months. The
Affymetrix GeneChip® Human Gene 1.0 ST Array was used to study gene
expression variation of CNDT2.5 cells in the presence or absence of
octreotide, which followed a precise long kinetic time. QRT-PCR and
western blot analyses were used to validate Affymetrix microarray in silico
data. The WST-1 cell proliferation assay was used to evaluate cell growth of
CNDT2.5 cells in the presence or absence of 1 µM octreotide at different
time points. Laser capture microdissected tumor cells and paraffin-embedded
tissue slides from SI-NETs at different disease stages were used to detect
octreotide effects on transcriptional and translational levels.
Microarray analyses revealed no relevant changes in SSTR expression
levels. However, octreotide stimulated six genes, ANXA1, ARHGAP18,
EMP1, GDF15, TGFBR2 and TNFSF15, which were not previously
considered essential in the octreotide signaling pathway. The six genes were
significantly upregulated after being treated with octreotide for 10 and 16
months (Fig. 4). Moreover, these six genes were detected in tumor tissue
specimens both at transcriptional and translational levels. ANXA1, EMP1,
TGFBR2 and TNFSF15 play major roles in cell proliferation and apoptosis
[77-80], whereas ARHGAP18 and GDF15 play main roles in signal
transduction [81,82]. ARHGAP18 and EMP1 might work as tumor
suppressors [83,84].
In conclusion, we presume that these novel genes might regulate cell
growth and differentiation on normal and tumor neuroendocrine cells.
Therefore, investigation of how octreotide is associated with triggering these
six genes to crosstalk may clarify a novel pathway associated with these
genes in controlling normal and NET cell growth.
28
Figure 4. QRT-PCR analysis of CNDT2.5 cells in the absence or presence of 1
µM octreotide. ANXA1, ARHGAP18, EMP1, GDF15, TGFBR2 and TNFSF15 were
analyzed using total RNA at 1 week (wk), 4 months, 10 months and 16 months (mo)
of culture by QRT-PCR. Results were plotted using the 2-∆∆Ct method with β-actin
expression (set to 1) from each individual sample as endogenous reference. Plotted
results are mean ± SD for triplicate wells. Significance was calculated by Two-Way
ANOVA followed by the Bonferroni test, comparing with untreated CNDT2.5 cells.
*** = p < 0.001
29
Paper II: Global miRNA profiling of SI-NETs
Fifteen snap-frozen specimens of SI-NETs at different disease stages, five
PT, five MM and five LM, were included in the study. Total RNA was
hybridized onto Affymetrix GeneChip® miRNA arrays for genome-wide
profiling. Differentially expressed miRNAs were then confirmed by using
northern blot analysis from the initial specimens and QRT-PCR analysis
from immuno-LCM normal EC cells and LCM SI-NET cells at different
stages of disease.
We also filtered the significant miRNAs from a global miRNA profiling,
which are differentially expressed in PT versus (vs) MM and LM. Thus, 33
differentially expressed miRNAs were selected for further analysis. The
bioinformatics clustering approach showed that the 33 differentially
expressed miRNAs clustered in a specific manner according to different
stages of disease. We also restricted up to nine selected miRNAs for further
investigations, to explore which ones were significantly altered between
primary tumor and metastasis groups.
Further investigation of the nine selected miRNAs by using LCM cells
showed that miR-96, -182, -183, -196a and -200a expression was
significantly upregulated in LCM tumor cells vs LCM normal EC cells,
whereas miR-31, -129-5p, -133a and -215 expression was significantly
downregulated in LCM tumor cells (Fig. 5). Online software programs, such
as DIANA LAB, PicTar, miRDB and TargetScan, were used to predict
miRNA target genes. The already published SI-NETs microarray profiling
data [85] were then used as a reference to filter miRNA potential target
genes.
In conclusion, little evidence of miRNA expression and deregulation in
SI-NETs has been previously reported. Thus, our genome-wide SI-NET
miRNA profiles have provided relevant information about potentially pivotal
miRNAs that might play roles in tumor progression and which might be
developed as therapeutics targets in the future. Further analyses to clarify
which genes are regulated by the selected miRNAs became our new goal.
These analyses might further illuminate the biological functions of the nine
selected miRNAs to improve our understanding of tumorigenesis and tumor
progression in SI-NETs. It is also critically important to develop blood
biomarkers to create less invasive tests to monitor SI-NETs.
30
Figure 5. QRT-PCR analysis validated the expression of nine selected miRNAs.
Total RNA was isolated from microdissected tumor cells and microdissected normal
EC cells. Analysis was run using three normal EC cell, three primary tumor, three
mesentery metastasis and three liver metastasis samples. (A) Upregulated miRNA
expression in tumor cells vs normal EC cells. (B) Downregulated miRNA
expression in tumor cells vs normal EC cells. Results were plotted using the 2-∆∆Ct
method with RNU48 expression (set to 1) from each individual sample for
normalization. Significance was calculated by One-Way ANOVA followed by the
Bonferroni test. * = p < 0.05, ** = p < 0.01 and *** = p < 0.001.
31
Paper III: Serum miRNA expression of SI-NETs
In Paper II, we investigated nine miRNAs in SI-NET tissue specimens.
Thus, the aims of this study are to investigate whether SI-NET tissue
miRNAs are expressed in patient serum samples, to explore a potential SSAs
role in miRNAs regulation in treated patients and to elucidate the serum
miRNA expression pattern reproducibility using serum samples collected in
different hospitals. Forty-nine serum samples were collected at UU,
including seven HD as normal controls, 14 PT, 14 LNM and 14 LM. In
addition, three HD and six LM serum samples were collected at UCL.
Target-specific stem-loop reverse transcription primers were used to detect
the nine tissue miRNAs in serum samples.
QRT-PCR analysis results show that miR-96, -182, -183, -196a and -200a
expression is lower in SI-NET untreated patients than in SSA-treated
patients at all different stages. One of the most remarkable findings is that
miR-200a shows an atypical behavior. Indeed, it is highly expressed in both
untreated and SSA-treated LM patients and unequivocally never at the
earlier stages (Fig. 6). Conversely, miR-31, -129-5p, -133a and -215
expression does not show any difference in untreated SI-NET patients and
SSA-treated patients at all different stages. Serum samples collected at UCL
were used to confirm the miRNA expression findings breakthrough from UU
collected samples. Four out of five highly expressed miRNAs were
significantly increased in SSA-treated LM patients vs untreated LM patients
and HD. However, miR-200a expression was significantly upregulated in
untreated patients and SSA-treated LM patients vs HD, as previously shown
in Figure 6 (Fig. 7). The expression of the other four miRNAs was
significantly decreased in both untreated and SSA-treated LM patients vs
HD.
In conclusion, SI-NET tissue miRNAs are expressed in serum samples. In
addition, SSAs play a pivotal role in increasing miRNAs expression of SSAtreated patient. The study enlightens that miR-200a might be involved in the
last phase of SI-NET progression. Moreover, the miRNA expression pattern
in serum samples from two different hospital collections, characterized by
matched medication, stage and grade, is almost identical. Therefore, a
further biological functional study is essential to better understand the
possible role of miR-200a in regulating the SI-NET metastatic process.
32
Figure 6. QRT-PCR analysis of serum miRNA expression in untreated and
SSA-treated SI-NET patients at all stages. MiR-96, -182, -183, -196a and -200a
expression is significantly increased at all stages of SSA-treated patients vs
untreated patients. Remarkably, miR-200a shows an atypical behavior because it is
increased in both untreated LM patients and SSA-treated LM patients. The QRTPCR analysis includes 21 untreated (7 PT, 7 LNM and 7 LM) and 21 SSA-treated
patients (7 SSA-PT, 7 SSA-LNM and 7 SSA-LM). Each group includes seven serum
samples. Results were plotted using the 2-∆∆Ct method with miR-16 expression (set to
1) from each individual sample for normalization. Significance was calculated by
One-Way ANOVA followed by the Dunnett’s test. * = p < 0.05, ** = p < 0.01 and
*** = p < 0.001.
33
Figure 7. QRT-PCR analysis of serum miRNA expression in untreated and
SSA-treated SI-NET patients at LM stage, samples collected at UU and UCL.
(A) MiR-96, -182, -183 and -196a expression is significantly increased in 6 SSAtreated LM patients vs 6 untreated LM patients and 6 HD. (B) MiR-200a shows an
atypical behavior has already been shown in Figure 6. Among the six samples of
each group, three samples were collected at UU and three were collected at UCL.
Results were plotted using the 2-∆∆Ct method with miR-16 expression (set to 1) from
each individual sample for normalization. Significance was calculated by One-Way
ANOVA followed by the Bonferroni test. * = p < 0.05, ** = p < 0.01 and *** = p <
0.001.
34
Paper IV: MiR-196a target genes in NET cells
In Papers II and III, we showed that miR-196a is highly expressed in SINETs vs normal controls by using both tissue specimens and serum samples.
The innovative aim of Paper IV was to investigate the biological functions of
miR-196a in vitro. The established human NET cell lines, CNDT2.5 and
NCI-H727 cells, which express miR-196a highly, were used as in vitro
models for SI-NETs and lung NETs. Anti-miR-196a was used to transfect
and to silence potential miR-196a effects in NET cells.
The miRNA target prediction analysis suggested several miR-196a target
genes, such as HOXA9, HOXB7, LRP4 and RSPO2, for further investigation.
The transcriptional level analysis showed that the expression of the four
target genes was significantly upregulated in miR-196a silenced NET cells
vs negative control cells (Fig. 8). The same upregulation effect was also
shown at the translational level.
Because four target genes significantly increased gene expression in miR196a silenced NET cells, we further investigated whether they regulated any
downstream genes. Braig et al [86] showed that silencing miR-196a resulted
in increasing HOXB7 at the transcriptional and translational levels in
melanoma cells. The upregulation effects also include ETS1 increased
activity, which induced BMP4 expression. Our unique findings showed that
miR-196a silencing also increased BMP4 and ETS1 gene expression in
CNDT2.5 and NCI-H727 cells. In addition, several indications have shown
that RSPO proteins bind to FZD receptors to enhance WNT-β-catenin
signaling [87] and that WNT co-receptors LRP5 and LRP6 are required for
FZD proteins to activate WNT signaling [87,88]. Our unprecedented
findings on downstream genes revealed that miR-196a silencing also
increased CTNNB1, FZD5, LRP5 and LRP6 gene expression, which might
be involved in WNT signaling pathways.
In conclusions, we have identified four miR-196a target genes that
regulate six downstream genes by using miR-196a silencing in SI-NET and
lung NET cells. Further investigation is essential to understand how target
genes regulate the downstream genes and their biological functions.
35
Figure 8. Silencing miR-196a induced HOXA9, HOXB7, LRP4 and RSPO2 gene
expression upregulation in two NET cell lines. CNDT2.5 cells and NCI-H727
cells were transfected with either anti-196a or negative control for 24 and 48 hours.
(A) The gene expression of four miR-196a target genes HOXA9, HOXB7, LRP4 and
RSPO2 was significantly upregulated in anti-196a transfected CNDT2.5 cells versus
the negative control. (B) The anti-miR-196a transfected NCI-H727 cells show a
significant gene expression upregulation of the four potential miR-196a target genes
mentioned in Figure 8A. Results were plotted using the 2-∆∆Ct method with β-actin
expression (set to 1) from each individual sample for normalization Plotted results
are means ± SEM from triplicate wells. Significance was calculated by One-Way
ANOVA followed by Dunnett’s test. * = p < 0.05, ** = p < 0.01 and *** = p <
0.001.
36
Concluding Remarks and Perspectives
Octreotide is a commonly used synthetic SSA that significantly improves the
management of NET patients. However, the molecular mechanisms leading
either to decreased symptoms or to successful tumor growth control are
largely unexplained. We treated midgut carcinoid cells CNDT2.5 from 1 day
up to 16 months with octreotide. We then performed Affymetrix microarray
analysis to provide unique insights into how octreotide might control SINET cells. Six genes (ANXA1, ARHGAP18, EMP1, GDF15, TGFBR2 and
TNFSF15) might regulate cell growth and differentiation in normal and NET
cells, as reported in Paper I.
Although abnormal miRNA expression is associated with several cancers,
scientific literature does not elucidate much about SI-NETs miRNA
expression and regulation. This suggested performing a global tissue miRNA
profiling using SI-NET specimens to restrict up to 33 differentially
expressed miRNAs. Selection of nine miRNAs out of 33 was important to
further study their relevant importance, as shown in Paper II. Studying LCM
cells revealed that the expression of five miRNAs (miR-96, -182, -183, 196a and -200a) was significantly upregulated in LCM tumor cells vs LCM
normal EC cells, whereas four miRNAs (miR-31, -129-5p, -133a and -215)
were significantly downregulated in LCM tumor cells vs LCM normal EC
cells.
We moved our central interest from tissue specimens to serum samples in
Paper III to further investigate the Paper II findings. The main goal was to
expand the scientific value of our previous investigation because to collect
patient blood samples is less invasive than collecting tissue specimens. This
study showed that the five upregulated and the four downregulated
expressed tissue miRNAs are also detectable in serum samples. In addition,
the serum miRNA expression pattern is very similar in the samples collected
in different hospitals, i.e. UU and UCL. One of the major findings of Paper
III is that miR-200a showed an atypical expression behavior at the LM stage.
Indeed, no significant difference is shown between untreated and SSAtreated samples at the LM stage, and miR-200a is highly increased in both
kinds of samples.
Among the nine selected miRNAs, we chose miR-196a to elucidate its
biological functions in vitro in Paper IV. Previously published data have
shown aberrant miR-196a expression in several malignancies, such as gastric
37
cancers, gastrointestinal stromal cancers and pancreatic cancers. We thus
silenced miR-196a in CNDT2.5 and NCI-H727 cells to investigate how
miR-196a might regulate its targets and control their biological functions.
Four target genes (HOXA9, HOXB7, LRP4 and RSPO2) and six downstream
regulated ones (BMP4, ETS1, CTNNB1, FZD5, LRP5 and LRP6) are altered
at transcriptional and translational levels, by silencing miR-196a expression.
Unexpectedly, miR-196a showed no impact on cell growth control either in
SI-NET or lung NET cells, and this finding led to questions about the
molecular mechanisms behind NET cell growth control.
In my opinion, I have identified from the findings in the four papers that
make up my PhD thesis, four challenging new tasks that require potential
future investigation. First, it would be important to further investigate the
network of the six genes that have been associated with a potential novel
octreotide signal cascade, as reported for the first time in Paper I. Antitumor
drugs are delivered efficiently through the tumor vasculature and cross the
vessel wall to reach the tumor tissue. However, heterogeneity within the
tumor micro-environment (TME) may affect the sensitivity of the tumor
cells to drug treatment [89]. Since four out of six genes, ANXA1, GDF15,
TGFBR2 and TNFSF15, have been reported in TME studies [90-93], it is
essential to explore the role of the TME, which might interpret the complex
gene and encoded protein crosstalk in controlling NET cell biology. Further
studies might clarify a novel pathway and whether these genes might play an
important role in controlling normal and NET cell growth.
Second, a deeper investigation of the regulatory roles of the other eight
selected miRNAs, as reported in Paper II, is one of my major interests. One
potential approach might be to use different NET cells as in vitro models to
study different kinds of NETs, such as pancreatic carcinoma cells, BON-1
and QGP-1.
Third, to cast light on potential reasons behind the atypical behavior of
miR-200a at the LM stage, as reported in Paper III, might help to better
understand whether and how to administer SSA to patients to improve their
medical management. It is therefore important to enlarge the serum sample
size to identify miR-200a expression at different stages of NETs. This might
provide significant statistical evidence to understand whether miR-200a can
be used as a stratification and/or follow-up biomarker for NET patients.
Meanwhile, it is critical to study the biological functions of miR-200a by
using the established in vitro model for miR-196a, as reported in Paper IV.
Fourth, four miRNA196a target genes and six downstream genes are
regulated by silencing miR-196a, as shown in Paper IV. These data support a
further investigation of the network between target genes and downstream
genes. In addition, exploring their biological functions might clarify whether
some of them, such as genomic DNA, transcribed RNA, or encoded proteins,
might be developed as diagnostic, prognostic or therapeutic tools. Because
38
miR-196a target gene HOXB7 is able to regulate BMP4 and ETS1 genes that
are involved in controlling cell migration in melanoma cells [86], it is
important to further investigate whether the HOXB7 gene also regulates cell
migration in NET cells. In addition, RSPO2 and LRP4 genes impact on the
WNT signaling pathway [87,88]. They regulate four downstream genes,
CTNNB1, FZD5, LRP5 and LRP6, genes which are also involved in WNT
signaling regulation. It is therefore critical to expand the study of WNT
signaling roles in NETs.
In conclusion, the short term goal is to clarify the biological functions of
miR-196a target genes and to use this established in vitro model to
investigate the biological functions of the other eight miRNAs. The long
term goal is to develop a modern chip array to detect miRNA expression by
using body fluids, such as serum samples, and to understand the specific
roles that miRNAs might play in NET tumorigenesis and tumor progression.
39
Acknowledgements
This work was carried out at the Department of Medical Sciences, Endocrine
Oncology, Uppsala University, Uppsala, Sweden. The work was supported
financially by grants from the Lions Foundation for Cancer Research, the Tore
Nilsson Foundation for Medical Research, and the Erik, Karin and Gösta Selander
Foundation.
I would like to express my sincere gratitude to all the people who have been
involved in and have supported this work and especially to:
Assoc. Prof. Valeria Giandomenico, my main supervisor. Thank you for introducing
me to a rare disease full of scientific challenges to explore. You have been the best
supervisor during my Master and PhD programs. You are also my best friend in
Sweden and I will never forget what we have done during our scientific and private
lives.
Prof. Kjell Öberg, my co-supervisor. Thank you for all your support since I joined
the group. You remind me not only to work hard but also to spend some time on
leisure activities. I am impressed that although you spend so much time in clinics or
travelling around the word for meetings, you remain active with skiing or playing
other sports. I appreciate your ability to release stress and thus improve fitness.
Dr. Tao Cui, my best colleague. Many thanks for your company in the same group
since Master program time. You came to the laboratory earlier than me, and always
kindly provided me with critical information and support for my experiments. I also
thank Fan and you for spending some free time together.
Hao Shi is a new and young Master student in our group. Although you only
recently came to the lab, you are of great help in our group.
Prof. Magnus Essand. I thank you for your scientific working support at Rudbeck.
You have always been supportive when I performed experiments in your lab there.
Thanks to your band for providing joy on different occasions.
Dr. Di Yu. Thank you for our scientific discussions and your professional help in
teaching me how to use viral transduction in the microRNA functional study. I also
thank Chuan and you for spending time with me in our personal lives.
Åsa Forsberg. Thank you for your daily care of all the problems in the laboratory
during the first three years of my PhD program. I am grateful.
40
Dr. Malin Grönberg. Thank you for ordering reagents for the lab, and for chatting
with me during “fika” time. I was impressed to see your paintings, which prove your
broader talent in life. Indeed, I would like to see more of your art in the future.
Assoc. Prof. Thomas Lind. Thank you for spending some of your time with me
during “fika” and lunch breaks at the lab, and also for being a friend in private life
with my husband and son. Furthermore, I appreciate your expertise in your field,
because I have never studied bones.
Dr. Cécile Martijn. Thank you for collaborating on the first two papers and for
providing strong bioinformatics support. I also thank you for translating all sorts of
documents from Swedish to English to help in my personal life.
All the international collaborators, but especially Prof. Jean-Baptiste Demoulin, Dr.
Ahmed Essaghir, Prof. Justo P Castaño, Dr. Raul M. Luque, Prof. Ricardo Lloyd,
Prof. Tim Meyer, Prof. Martyn Caplin and Dr. Mohid Khan. Although I have not yet
had the chance to meet you, I would like to thank all of you for having provided a
variety of support to help me complete my scientific work.
All my national colleagues, especially at FOA2 and FOA3, Uppsala University
Hospital, and the IGP group at Rudbeck.
I am thankful to European Neuroendocrine Tumor Society (ENETS) and North
American Neuroendocrine Tumor Society (NANETS). I have received several times
of travel grants to attend the conferences and also had the opportunities to present
my scientific work in the conferences that organized by the two societies.
I am grateful to Mandy Trickett to the professional American English editing and to
provide critical suggestions and commons to improve the English quality of my
papers and the thesis.
I also thank all my friends at Uppsala University, the Taiwanese Student
Association, and my classmates in my previous Master program. You have all
enriched my life in Sweden.
Thanks to all the members of my family, especially my Mom. I miss you all the time
and you are always in my mind.
Last but not the least, I would like to thank my beloved husband, Shang-Feng, and
my lovely son, Chia-Ho. Shang-Feng, thank you for your courage to experience a
new life with me in Sweden and for taking care of our family all the time.
41
References
1. Kloppel G (2007) Tumour biology and histopathology of neuroendocrine
tumours. Best Pract Res Clin Endocrinol Metab 21: 15-31.
2. Wiedenmann B, Franke WW, Kuhn C, Moll R, Gould VE (1986)
Synaptophysin: a marker protein for neuroendocrine cells and
neoplasms. Proc Natl Acad Sci U S A 83: 3500-3504.
3. Kvetnoy IM (1999) Extrapineal melatonin: location and role within
diffuse neuroendocrine system. Histochem J 31: 1-12.
4. Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, et al. (2008)
Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol 9:
61-72.
5. Schimmack S, Svejda B, Lawrence B, Kidd M, Modlin IM (2011) The
diversity and commonalities of gastroenteropancreatic neuroendocrine tumors. Langenbecks Arch Surg 396: 273-298.
6. Rindi G, Leiter AB, Kopin AS, Bordi C, Solcia E (2004) The "normal"
endocrine cell of the gut: changing concepts and new evidences.
Ann N Y Acad Sci 1014: 1-12.
7. Ahlman H, Nilsson (2001) The gut as the largest endocrine organ in the
body. Ann Oncol 12 Suppl 2: S63-68.
8. Oberg K, Castellano D (2011) Current knowledge on diagnosis and
staging of neuroendocrine tumors. Cancer Metastasis Rev 30 Suppl
1: 3-7.
9. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, et al. (2008) One
hundred years after "carcinoid": epidemiology of and prognostic
factors for neuroendocrine tumors in 35,825 cases in the United
States. J Clin Oncol 26: 3063-3072.
10. Bosman TF, Carneiro F, Hruban RH, Theise ND, editors (2010) WHO
Classification of Tumours of the Digestive System 4ed. Lyon IARC.
13 p.
11. Kloppel G, Perren A, Heitz PU (2004) The gastroenteropancreatic
neuroendocrine cell system and its tumors: the WHO classification.
Ann N Y Acad Sci 1014: 13-27.
12. Klimstra DS, Modlin IR, Coppola D, Lloyd RV, Suster S (2010) The
pathologic classification of neuroendocrine tumors: a review of
nomenclature, grading, and staging systems. Pancreas 39: 707-712.
13. Oberg KE (2010) Gastrointestinal neuroendocrine tumors. Ann Oncol 21
Suppl 7: vii72-80.
42
14. Rindi G, Kloppel G, Couvelard A, Komminoth P, Korner M, et al.
(2007) TNM staging of midgut and hindgut (neuro) endocrine
tumors: a consensus proposal including a grading system. Virchows
Arch 451: 757-762.
15. Eriksson B, Oberg K, Stridsberg M (2000) Tumor markers in
neuroendocrine tumors. Digestion 62 Suppl 1: 33-38.
16. Rindi G, Bordi C, La Rosa S, Solcia E, Delle Fave G (2011)
Gastroenteropancreatic (neuro)endocrine neoplasms: the histology
report. Dig Liver Dis 43 Suppl 4: S356-360.
17. Pape UF, Berndt U, Muller-Nordhorn J, Bohmig M, Roll S, et al. (2008)
Prognostic factors of long-term outcome in gastroenteropancreatic
neuroendocrine tumours. Endocr Relat Cancer 15: 1083-1097.
18. Ramage JK, Ahmed A, Ardill J, Bax N, Breen DJ, et al. (2012)
Guidelines for the management of gastroenteropancreatic
neuroendocrine (including carcinoid) tumours (NETs). Gut 61: 6-32.
19. Kvols LK, Moertel CG, O'Connell MJ, Schutt AJ, Rubin J, et al. (1986)
Treatment of the malignant carcinoid syndrome. Evaluation of a
long-acting somatostatin analogue. N Engl J Med 315: 663-666.
20. Oberg K (1992) Interferons in the management of neuroendocrine
tumors and their possible mechanism of action. Yale J Biol Med 65:
519-529; discussion 531-516.
21. Pavel ME, Hainsworth JD, Baudin E, Peeters M, Horsch D, et al. (2011)
Everolimus plus octreotide long-acting repeatable for the treatment
of advanced neuroendocrine tumours associated with carcinoid
syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3
study. Lancet 378: 2005-2012.
22. Kim S, Ding W, Zhang L, Tian W, Chen S (2014) Clinical response to
sunitinib as a multitargeted tyrosine-kinase inhibitor (TKI) in solid
cancers: a review of clinical trials. Onco Targets Ther 7: 719-728.
23. O'Farrell AM, Abrams TJ, Yuen HA, Ngai TJ, Louie SG, et al. (2003)
SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent
activity in vitro and in vivo. Blood 101: 3597-3605.
24. Forrer F, Valkema R, Kwekkeboom DJ, de Jong M, Krenning EP (2007)
Neuroendocrine tumors. Peptide receptor radionuclide therapy. Best
Pract Res Clin Endocrinol Metab 21: 111-129.
25. Brazeau P, Vale W, Burgus R, Ling N, Butcher M, et al. (1973)
Hypothalamic polypeptide that inhibits the secretion of
immunoreactive pituitary growth hormone. Science 179: 77-79.
26. Mandarino L, Stenner D, Blanchard W, Nissen S, Gerich J, et al. (1981)
Selective effects of somatostatin-14, -25 and -28 on in vitro insulin
and glucagon secretion. Nature 291: 76-77.
27. Reichlin S (1983) Somatostatin (second of two parts). N Engl J Med 309:
1556-1563.
43
28. Pradayrol L, Jornvall H, Mutt V, Ribet A (1980) N-terminally extended
somatostatin: the primary structure of somatostatin-28. FEBS Lett
109: 55-58.
29. Modlin IM, Pavel M, Kidd M, Gustafsson BI (2010) Review article:
somatostatin analogues in the treatment of gastroenteropancreatic
neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther 31:
169-188.
30. Schonbrunn A, Tashjian H, Jr. (1978) Characterization of functional
receptors for somatostatin in rat pituitary cells in culture. J Biol
Chem 253: 6473-6483.
31. Moller LN, Stidsen CE, Hartmann B, Holst JJ (2003) Somatostatin
receptors. Biochim Biophys Acta 1616: 1-84.
32. Bell GI, Reisine T (1993) Molecular biology of somatostatin receptors.
Trends Neurosci 16: 34-38.
33. Rosenthal W, Hescheler J, Hinsch KD, Spicher K, Trautwein W, et al.
(1988) Cyclic AMP-independent, dual regulation of voltagedependent Ca2+ currents by LHRH and somatostatin in a pituitary
cell line. EMBO J 7: 1627-1633.
34. Cattaneo MG, Amoroso D, Gussoni G, Sanguini AM, Vicentini LM
(1996) A somatostatin analogue inhibits MAP kinase activation and
cell proliferation in human neuroblastoma and in human small cell
lung carcinoma cell lines. FEBS Lett 397: 164-168.
35. Cervia D, Bagnoli P (2007) An update on somatostatin receptor signaling
in native systems and new insights on their pathophysiology.
Pharmacol Ther 116: 322-341.
36. Ferone D, Gatto F, Arvigo M, Resmini E, Boschetti M, et al. (2009) The
clinical-molecular interface of somatostatin, dopamine and their
receptors in pituitary pathophysiology. J Mol Endocrinol 42: 361370.
37. Oberg KE, Reubi JC, Kwekkeboom DJ, Krenning EP (2010) Role of
somatostatins in gastroenteropancreatic neuroendocrine tumor
development and therapy. Gastroenterology 139: 742-753, 753 e741.
38. Susini C, Buscail L (2006) Rationale for the use of somatostatin analogs
as antitumor agents. Ann Oncol 17: 1733-1742.
39. Bruns C, Weckbecker G, Raulf F, Kaupmann K, Schoeffter P, et al.
(1994) Molecular pharmacology of somatostatin-receptor subtypes.
Ann N Y Acad Sci 733: 138-146.
40. Rinke A, Muller HH, Schade-Brittinger C, Klose KJ, Barth P, et al.
(2009) Placebo-controlled, double-blind, prospective, randomized
study on the effect of octreotide LAR in the control of tumor growth
in patients with metastatic neuroendocrine midgut tumors: a report
from the PROMID Study Group. J Clin Oncol 27: 4656-4663.
41. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic
gene lin-4 encodes small RNAs with antisense complementarity to
lin-14. Cell 75: 843-854.
44
42. Borchert GM, Lanier W, Davidson BL (2006) RNA polymerase III
transcribes human microRNAs. Nat Struct Mol Biol 13: 1097-1101.
43. Lee Y, Kim M, Han J, Yeom KH, Lee S, et al. (2004) MicroRNA genes
are transcribed by RNA polymerase II. EMBO J 23: 4051-4060.
44. Lee Y, Ahn C, Han J, Choi H, Kim J, et al. (2003) The nuclear RNase III
Drosha initiates microRNA processing. Nature 425: 415-419.
45. Lee YS, Nakahara K, Pham JW, Kim K, He Z, et al. (2004) Distinct
roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA
silencing pathways. Cell 117: 69-81.
46. Yi R, Qin Y, Macara IG, Cullen BR (2003) Exportin-5 mediates the
nuclear export of pre-microRNAs and short hairpin RNAs. Genes
Dev 17: 3011-3016.
47. He L, Hannon GJ (2004) MicroRNAs: small RNAs with a big role in
gene regulation. Nat Rev Genet 5: 522-531.
48. Meister G, Landthaler M, Patkaniowska A, Dorsett Y, Teng G, et al.
(2004) Human Argonaute2 mediates RNA cleavage targeted by
miRNAs and siRNAs. Mol Cell 15: 185-197.
49. Chen CZ, Li L, Lodish HF, Bartel DP (2004) MicroRNAs modulate
hematopoietic lineage differentiation. Science 303: 83-86.
50. Cheng AM, Byrom MW, Shelton J, Ford LP (2005) Antisense inhibition
of human miRNAs and indications for an involvement of miRNA in
cell growth and apoptosis. Nucleic Acids Res 33: 1290-1297.
51. Garzon R, Fabbri M, Cimmino A, Calin GA, Croce CM (2006)
MicroRNA expression and function in cancer. Trends Mol Med 12:
580-587.
52. Karp X, Ambros V (2005) Developmental biology. Encountering
microRNAs in cell fate signaling. Science 310: 1288-1289.
53. Poy MN, Eliasson L, Krutzfeldt J, Kuwajima S, Ma X, et al. (2004) A
pancreatic islet-specific microRNA regulates insulin secretion.
Nature 432: 226-230.
54. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, et al. (2002)
Frequent deletions and down-regulation of micro- RNA genes
miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc
Natl Acad Sci U S A 99: 15524-15529.
55. He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, et al.
(2005) A microRNA polycistron as a potential human oncogene.
Nature 435: 828-833.
56. Calin GA, Croce CM (2006) MicroRNA-cancer connection: the
beginning of a new tale. Cancer Res 66: 7390-7394.
57. Cortez MA, Bueso-Ramos C, Ferdin J, Lopez-Berestein G, Sood AK, et
al. (2011) MicroRNAs in body fluids--the mix of hormones and
biomarkers. Nat Rev Clin Oncol 8: 467-477.
58. Chen X, Ba Y, Ma L, Cai X, Yin Y, et al. (2008) Characterization of
microRNAs in serum: a novel class of biomarkers for diagnosis of
cancer and other diseases. Cell Res 18: 997-1006.
45
59. Gilad S, Meiri E, Yogev Y, Benjamin S, Lebanony D, et al. (2008)
Serum microRNAs are promising novel biomarkers. PLoS One 3:
e3148.
60. Akinc A, Zumbuehl A, Goldberg M, Leshchiner ES, Busini V, et al.
(2008) A combinatorial library of lipid-like materials for delivery of
RNAi therapeutics. Nat Biotechnol 26: 561-569.
61. Bitko V, Musiyenko A, Shulyayeva O, Barik S (2005) Inhibition of
respiratory viruses by nasally administered siRNA. Nat Med 11: 5055.
62. Weiler J, Hunziker J, Hall J (2006) Anti-miRNA oligonucleotides
(AMOs): ammunition to target miRNAs implicated in human
disease? Gene Ther 13: 496-502.
63. Rhoades MW, Reinhart BJ, Lim LP, Burge CB, Bartel B, et al. (2002)
Prediction of plant microRNA targets. Cell 110: 513-520.
64. Barbato C, Arisi I, Frizzo ME, Brandi R, Da Sacco L, et al. (2009)
Computational challenges in miRNA target predictions: to be or not
to be a true target? J Biomed Biotechnol 2009: 803069.
65. Yoon S, De Micheli G (2006) Computational identification of
microRNAs and their targets. Birth Defects Res C Embryo Today
78: 118-128.
66. Kuhn DE, Martin MM, Feldman DS, Terry AV, Jr., Nuovo GJ, et al.
(2008) Experimental validation of miRNA targets. Methods 44: 4754.
67. Qiu R, Liu Y, Wu JY, Liu K, Mo W, et al. (2009) Misexpression of miR196a induces eye anomaly in Xenopus laevis. Brain Res Bull 79: 2631.
68. Chen S, Ni M, Yu B, Lv T, Lu M, et al. (2007) Construction and
identification of a human liver specific microRNA eukaryotic
expression vector. Cell Mol Immunol 4: 473-477.
69. Donsante A, Miller DG, Li Y, Vogler C, Brunt EM, et al. (2007) AAV
vector integration sites in mouse hepatocellular carcinoma. Science
317: 477.
70. Kato Y, Sawata SY, Inoue A (2010) A lentiviral vector encoding two
fluorescent proteins enables imaging of adenoviral infection via
adenovirus-encoded miRNAs in single living cells. J Biochem 147:
63-71.
71. Sekine S, Ogawa R, Ito R, Hiraoka N, McManus MT, et al. (2009)
Disruption of Dicer1 induces dysregulated fetal gene expression and
promotes hepatocarcinogenesis. Gastroenterology 136: 2304-2315
e2301-2304.
72. Meister G, Landthaler M, Dorsett Y, Tuschl T (2004) Sequence-specific
inhibition of microRNA- and siRNA-induced RNA silencing. RNA
10: 544-550.
46
73. Naguibneva I, Ameyar-Zazoua M, Nonne N, Polesskaya A, Ait-Si-Ali S,
et al. (2006) An LNA-based loss-of-function assay for micro-RNAs.
Biomed Pharmacother 60: 633-638.
74. Ebert MS, Neilson JR, Sharp PA (2007) MicroRNA sponges:
competitive inhibitors of small RNAs in mammalian cells. Nat
Methods 4: 721-726.
75. Loya CM, Lu CS, Van Vactor D, Fulga TA (2009) Transgenic
microRNA inhibition with spatiotemporal specificity in intact
organisms. Nat Methods 6: 897-903.
76. Xiao J, Yang B, Lin H, Lu Y, Luo X, et al. (2007) Novel approaches for
gene-specific interference via manipulating actions of microRNAs:
examination on the pacemaker channel genes HCN2 and HCN4. J
Cell Physiol 212: 285-292.
77. Dhasarathy A, Phadke D, Mav D, Shah RR, Wade PA (2011) The
transcription factors snail and slug activate the transforming growth
factor-Beta signaling pathway in breast cancer. PLoS ONE 6:
e26514.
78. Rodrigo JP, Garcia-Pedrero JM, Llorente JL, Fresno MF, Allonca E, et
al. (2011) Down-regulation of annexin A1 and A2 protein
expression in intestinal-type sinonasal adenocarcinomas. Hum
Pathol 42: 88-94.
79. Sethi G, Sung B, Aggarwal BB (2009) Therapeutic Potential of
VEGI/TL1A in Autoimmunity and Cancer. Adv Exp Med Biol 647:
207-215.
80. Zinovyeva MV, Monastyrskaya GS, Kopantzev EP, Vinogradova TV,
Kostina MB, et al. (2010) Identification of some human genes
oppositely regulated during esophageal squamous cell carcinoma
formation and human embryonic esophagus development. Dis
Esophagus 23: 260-270.
81. Joshi JP, Brown NE, Griner SE, Nahta R (2011) Growth differentiation
factor 15 (GDF15)-mediated HER2 phosphorylation reduces
trastuzumab sensitivity of HER2-overexpressing breast cancer cells.
Biochem Pharmacol 82: 1090-1099.
82. Kim TY, Vigil D, Der CJ, Juliano RL (2009) Role of DLC-1, a tumor
suppressor protein with RhoGAP activity, in regulation of the
cytoskeleton and cell motility. Cancer Metastasis Rev 28: 77-83.
83. Maeda M, Hasegawa H, Hyodo T, Ito S, Asano E, et al. (2011)
ARHGAP18, a GTPase-activating protein for RhoA, controls cell
shape, spreading, and motility. Mol Biol Cell 22: 3840-3852.
84. Zhang J, Cao W, Xu Q, Chen WT (2011) The expression of EMP1 is
downregulated in oral squamous cell carcinoma and possibly
associated with tumour metastasis. J Clin Pathol 64: 25-29.
85. Leja J, Essaghir A, Essand M, Wester K, Oberg K, et al. (2009) Novel
markers for enterochromaffin cells and gastrointestinal
neuroendocrine carcinomas. Mod Pathol 22: 261-272.
47
86. Braig S, Mueller DW, Rothhammer T, Bosserhoff AK (2010)
MicroRNA miR-196a is a central regulator of HOX-B7 and BMP4
expression in malignant melanoma. Cell Mol Life Sci 67: 35353548.
87. Nam JS, Turcotte TJ, Smith PF, Choi S, Yoon JK (2006) Mouse
cristin/R-spondin family proteins are novel ligands for the Frizzled 8
and LRP6 receptors and activate beta-catenin-dependent gene
expression. J Biol Chem 281: 13247-13257.
88. Clement-Lacroix P, Ai M, Morvan F, Roman-Roman S, Vayssiere B, et
al. (2005) Lrp5-independent activation of Wnt signaling by lithium
chloride increases bone formation and bone mass in mice. Proc Natl
Acad Sci U S A 102: 17406-17411.
89. Tredan O, Galmarini CM, Patel K, Tannock IF (2007) Drug resistance
and the solid tumor microenvironment. J Natl Cancer Inst 99: 14411454.
90. Bauskin AR, Brown DA, Kuffner T, Johnen H, Luo XW, et al. (2006)
Role of macrophage inhibitory cytokine-1 in tumorigenesis and
diagnosis of cancer. Cancer Res 66: 4983-4986.
91. Deng W, Gu X, Lu Y, Gu C, Zheng Y, et al. (2012) Down-modulation of
TNFSF15 in ovarian cancer by VEGF and MCP-1 is a pre-requisite
for tumor neovascularization. Angiogenesis 15: 71-85.
92. Tan AR, Alexe G, Reiss M (2009) Transforming growth factor-beta
signaling: emerging stem cell target in metastatic breast cancer?
Breast Cancer Res Treat 115: 453-495.
93. Yi M, Schnitzer JE (2009) Impaired tumor growth, metastasis,
angiogenesis and wound healing in annexin A1-null mice. Proc Natl
Acad Sci U S A 106: 17886-17891.
48
Acta Universitatis Upsaliensis
Digital Comprehensive Summaries of Uppsala Dissertations
from the Faculty of Medicine 1039
Editor: The Dean of the Faculty of Medicine
A doctoral dissertation from the Faculty of Medicine, Uppsala
University, is usually a summary of a number of papers. A few
copies of the complete dissertation are kept at major Swedish
research libraries, while the summary alone is distributed
internationally through the series Digital Comprehensive
Summaries of Uppsala Dissertations from the Faculty of
Medicine. (Prior to January, 2005, the series was published
under the title “Comprehensive Summaries of Uppsala
Dissertations from the Faculty of Medicine”.)
Distribution: publications.uu.se
urn:nbn:se:uu:diva-233207
ACTA
UNIVERSITATIS
UPSALIENSIS
UPPSALA
2014