Journal Pre-proof Wnt-regulating microRNAs role in gastric cancer malignancy
Milad Ashrafizadeh, Hossein Rafiei, Reza Mohammadinejad, Tahereh Farkhondeh, Saeed Samarghandian PII:
S0024-3205(20)30295-2
DOI:
https://doi.org/10.1016/j.lfs.2020.117547
Reference:
LFS 117547
To appear in:
Life Sciences
Received date:
11 January 2020
Revised date:
6 March 2020
Accepted date:
11 March 2020
Please cite this article as: M. Ashrafizadeh, H. Rafiei, R. Mohammadinejad, et al., Wntregulating microRNAs role in gastric cancer malignancy, Life Sciences (2020), https://doi.org/10.1016/j.lfs.2020.117547
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
© 2020 Published by Elsevier.
Journal Pre-proof Wnt-regulating microRNAs role in gastric cancer malignancy Milad Ashrafizadeh1, Hossein Rafiei2, Reza Mohammadinejad3, Tahereh Farkhondeh4, Saeed Samarghandian5* 1
Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz,
Iran 2
Department of Biology, Faculty of Sciences, Shiraz Branch, Islamic Azad University, Shiraz,
Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of
ro
3
of
Iran
Medical Sciences, Kerman, Iran
-p
Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
re
5
Correspondence: Saeed Samarghandian, Department of Basic Medical Sciences, Neyshabur
lP
University of Medical Sciences, Neyshabur, Iran, Email:
[email protected]
Abstract
na
4
ur
Gastric cancer (GC) is responsible for high morbidity and mortality worldwide. This cancer
Jo
claims fifth place among other cancers. There are a number of factors associated with GC development such as alcohol consumption and tobacco smoking. It seems that genetic factors play significant role in GC malignancy and progression. MicroRNAs (miRs) are short noncoding RNA molecules with negative impact on the expression of target genes. A variety of studies have elucidated the potential role of miRs in GC growth. Investigation of molecular pathways have revealed that miRs function as upstream modulators of Wnt signaling pathway. This signaling pathway involves in important biological processes such as cell proliferation and differentiation, and its dysregulation is associated with GC invasion. At the present review, we demonstrate that how miRs regulate Wnt signaling pathway in GC malignancy. Keywords: MicroRNAs, Gastric cancer, Cancer therapy, Wnt, Signaling pathway
Journal Pre-proof 1. Introduction Gastric cancer (GC) is one of the most malignant cancers with high morbidity and mortality. This cancer claims the fifth place among other cancers [1-4]. It seems that annually, more than 500,000 new cases are diagnosed with GC [5, 6]. Studies demonstrate that GC has high incidence rate in some areas such as Asia, Korea, Japan and China [7]. Based on the high prevalence of GC, it is considered as one of the most important health issues. A variety of factors contribute to the development of GC and Helicobacter pylori infection, alcohol consumption and tobacco smoking as well as inappropriate diet are among them [8, 9]. The World Health
of
Organization (WHO) has divided GC into five categories such as papillary, tubular, mucinous,
ro
signet-ring cell and mixed carcinoma [10]. Surgery and chemotherapy are common strategies in GC therapy. However, the metastasis and recurrence challenge these methods [11]. Over the past
-p
decades, an improvement has been made in diagnostic tools that has enhanced our understanding of genetic factors involved in GC malignancy and progression.
re
As short non-coding RNA molecules, microRNAs (miRs) have a length as low as 24 nucleotides
lP
that exert inhibitory effect on the expression of target genes [12-14]. MiRs affect the target genes at post-transcriptional level [15]. Accumulating data demonstrate that miRs play a significant role in various main biological processes including differentiation, apoptotic cell death,
na
proliferation and metabolism [16]. In respect to the modulatory impact of miRs on expression, they have improved our comprehension towards regulation of protein expression [17-20]. The
ur
mRNA degradation, mRNA destabilization and translation inhibition are the strategies that miRs
Jo
follow to negatively affect the expression of target genes [21]. It seems that miR dysregulation is associated with a number of pathological conditions, particularly cancer [22-26]. Hence, miR regulation is of importance in the field of cancer therapy. It has been reported that abnormal expression of miRs remarkably affects the viability, proliferation capability and metastasis of cancer cells [27-29]. Overall, miRs are divided into two characteristic categories in the field of cancer: A) oncogenic miRs that enhance the malignancy of tumor cells, and B) oncosuppressor miRs that are associated with cancer inhibition [30]. It is held that miRs are able to sensitize tumor cells to chemo- and radiotherapy showing the potential role of miRs in cancer therapy [3133].
2. Wnt signaling pathway
Journal Pre-proof Wnt signaling pathway includes 18 ligands and 10 receptors that have important rules in different processes including embryogenesis, homeostasis, cell growth, cell proliferation, cell migration and cell survival [2, 34-40]. Wnt1 gene is the first Wnt gene that was introduced in 1982 while working on Drosophila melanogaster [41]. Any impairment in Wnt signaling pathway is related to the generation of pathological conditions [42-44]. A number of studies have elucidated the role of aberrant Wnt signaling pathway in cancer progression and malignancy [45, 46]. So, targeting this molecular pathway is of importance in cancer therapy. Wnt signaling pathway is divided into two subsets including canonical or -catenin-dependent pathway, and
of
non-canonical or -catenin-independent pathway [47]. In canonical pathway, a complex known
ro
as “destruction complex” contributes to the degradation of -catenin upon physiological condition. “Destruction complex” contains adenomatous polyposis coli (APC), Dishevelled
-p
(Dvl), casein kinase 1 (CK1) and Axin1/Axin2 that phosphorylate -catenin leading to its
re
degradation by glycogen synthase kinase-3 (GSK-3) [48-50]. After the attachment of Wnt ligand to the membrane receptor, Frizzled (Fzd) and low-density lipoprotein receptor-associated
lP
protein 5/6 (LRP5/6) undergo induction to trigger Wnt signaling pathway. Then, a disruption occurs in the “destruction complex” by stimulation of Dvl and consequently, leads to the GSK-
na
3 inactivation. Next, high amount of -catenin accumulates in the cytoplasm and then, translocates to the nucleus to induce the expression of target genes by interacting with lymphoid
ur
enhancer binding factors (LEF) and T-cell factors (TCF) [51, 52]. The non-canonical pathway is divided into two classes including Wnt/Ca2+ pathway, and planar cell polarity (PCP) pathway
Jo
[53-55]. It seems that Wnt5a is the major ligand of non-canonical pathway. Wnt/Ca2+ pathway involves in migration, cell adhesion and cytoskeletal rearrangement. The Wnt/Ca2+ signaling pathway undergoes upregulation by calcium signaling stimulation through phospholipase C/protein kinase C (PKC)/Ca2+ and calmodulin-sensitive protein kinase II (CaMKII) resulting in induction of nuclear factor associated with T cells (NFAT) [56-58]. However, upon PCP signaling pathway, binding of Wnt ligand to Fzd receptor stimulates Dvl. Activated Dvl induces Rho/Rho-associated kinase and Rac/c-Jun N-terminal kinase (JNK) as well as actin polymerization. It appears that PCP pathway plays a remarkable role in cell polarization and motility [59-62]. It seems that Wnt signaling pathway dysregulation is one of the principal causes of cancer due to its critical role in important biological processes. The anti-tumor activity of some of the drugs is
Journal Pre-proof a consequence of their modulatory impacts on Wnt signaling pathway. Upregulation of Wnt signaling pathway improves the proliferation and malignancy of triple negative breast cancer (TNBC) cells. Oxymatrine remarkably inhibits the progression of TNBC cells by downregulation of Wnt signaling pathway [63]. The same story occurs in colorectal cancer. Guanylate-binding protein-1 (GBP-1) elevates the efficacy of chemotherapy by inhibition of Wnt signaling pathway [64]. Long non-coding RNAs (lncRNAs), miRs and signaling pathways such as Hedgehog function as upstream modulators of Wnt pathway to suppress the progression and invasion of cancer cells [65-68]. Receptor for activated C kinase 1 (RACK1) is suggested to be
of
influential in treatment of GC. Stimulation of RACK1 is associated with reduced malignancy of
ro
GC cells by Wnt inhibition [69]. Besides, family with sequence similarity 83, member D (FAM83D) promotes the growth of GC cells and is related to the poor prognosis of patients with
-p
GC due to enhancing the expression of Wnt//-catenin signaling pathway [70]. These studies highlight that Wnt pathway dysregulation occurs in cancer and this aberration is also of
lP
re
importance in GC therapy.
3. Interaction between microRNAs and Wnt pathway in cancer cells
na
There are a number of phases involved in miR biogenesis [71-73]. This biogenesis is started in nucleus by transcription of miR via RNA polymerase II leading to the formation of primary-miR
ur
(pri-miR) .[74] Then, double stranded RNA-binding protein DiGeorge syndrome critical region 8 (DGCR8) and RNase III endonuclease Drosha involve in synthesis of a shorter miR from pri-
Jo
miR known as precursor-miR (pre-miR) [75]. Exportin 5 (Exp5) contributes to the translocation of pre-miR from nucleus to cytoplasm [76]. Noteworthy, a mature miR with the length as low as 19-24 nucleotides is produced through the action of Dicer enzyme [77]. To be functionalized, mature miR is embedded into RNA-induced silencing complex [78] including Argonaute 2 (Ago2) protein, transactivating response RNA-binding protein (TRBP), protein kinase Ractivating protein (PACT) and Dicer [79]. Several studies have investigated the interaction between miRs and Wnt/-catenin signaling pathway in cancer cells. It appears that miRs affect Wnt pathway in following steps: A) affecting the translocation of -catenin from cytoplasm to nucleus [80]; B) down-regulation/upregulation of target genes of Wnt pathway [81], C) regulation of upstream modulators [40, 82, 83], and D) influencing the expression of -catenin and Wnt [84-86].
Journal Pre-proof
4. Wnt-regulating microRNAs 4.1 MiR-503 MiR-503 is considered as an oncosuppressor miR that reduces the progression of tumor cells. It seems that miR-503 down-regulation is associated with enhanced malignancy of non-small celllung cancer (NSCLC) cells [87]. Besides, miR-503 diminishes the viability of osteosarcoma cells via targeting L1CAM [88]. The same story occurs in hepatocellular carcinoma and miR-503 is able to modulate cancer invasion [89]. Accumulating data demonstrates that miR-503 can be
of
beneficial in treatment of GC [90, 91]. Investigation of molecular signaling pathways shows that
ro
miR-503 is able to significantly inhibit the viability and malignancy of GC cells by Wnt down-
-p
regulation through enhancing GSK-3 and phosphorylated (p)--catenin [92].
4.2 MiR-675
re
Paired-like homeodomain transcription factor 1 (PITX1) is considered as a tumor suppression
lP
transcription factor and its minimal expression is related to the poor prognosis of patients with cancer [93-95]. MiR-675 is responsible for high viability and proliferation of GC cells. It is
na
suggested that miR-675 reduces the expression of PTX1. The down-regulated PTX1 is associated with stimulation of Wnt/-catenin signaling pathway leading to the induction of
ur
epithelial-to-mesenchymal transition (EMT) and subsequently, an increase in invasion and migration. It was found that the expression of cyclin D1 and c-Myc undergo upreglation under
4.3 MiR-204
Jo
Wnt pathway activation [96].
Plant-derived chemicals are potential candidates in treatment of cancers [97-99]. It has been demonstrated that naturally occurring compounds are able to target various signaling pathways in cancer therapy [100-102]. Sinomenine (SIN) is a plant-derived chemical alkaloid with the capability of suppressing the proliferation and metastasis of cancers [103-108]. SIN is capable of reducing the migration and malignancy of GC cells by inhibition of Wnt signaling pathway through down-regulation of Wnt3a and -catenin. It seems that these anti-tumor effects of SIN are mediated through miR-204 upregulation [109].
Journal Pre-proof 4.4 MiR-17HG MiR-17HG is a pri-miR located in the 800-base-pair region of human chromose 13 that involves in production of six main miRs including miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1 and miR-92-1 [110]. A number of studies have revealed that miR-17HG contributes to the metastasis of cancer cells and its members undergo upregulation in various cancers such as colorectal cancer and pancreatic cancer [78, 111, 112]. On the other hand, IRF-1 plays a significant role in some biological processes such as proliferation and differentiation [113, 114]. IRF-1 dysregulation occurs in GC and leukemia [115]. IRF-1 binds to the transcriptional site at the
of
miR-17HG promoter to down-regulate miR-18a and miR-19a resulting in inhibition of Wnt
ro
signaling pathway and GC malignancy [116].
-p
4.5 MiR-876-5p
MiR-876-5p is a potential oncosuppressor miR that involves in sensitizing tumor cells to
re
chemotherapy, inhibition of EMT and suppressing the proliferation of cancer cells [117-122].
lP
Investigation of molecular signaling pathways in GC has shown that miR-876-5p is able to bind to the 3/ untranslated region (3/ UTR) of Wnt5a resulting in an increase in apoptotic cell death
4.6 MiR-188-5p
na
and a decrease in cell viability and progression [123].
ur
Another important oncosuppressor factor is miR-188-5p that has demonstrated great potential in
Jo
inhibition of NSCLC, glioma and so on [124-127]. In contrast, it seems that miR-188-5p is associated with poor prognosis of patients with GC. MiR-188-5p phosphorylates GSK-3 at serin9 (Ser9) leading to the invasion and metastasis of GC both in vitro and in vivo [128].
4.7 MiR-15a-3p Zinc proteins play a remarkable role in cell growth, cell proliferation, cell differentiation and synthesis of genetic material [129, 130]. Zinc influx (SLC39/ZIP) and zinc efflux (SLC30/ZnT) are two important zinc proteins in mammals [131]. SLC39A7 is a member of zinc proteins [132]. Accumulating data demonstrates that SLC39A7 mediates the resistance of tumor cells to chemotherapy and is associated with proliferation and malignancy of cancer cells. MiR-15a-3p exerts inhibitory impact on the metastasis and invasion of GC cells by suppressing the nuclear
Journal Pre-proof translocation of -catenin. SLC39A7 undergoes inhibition under Wnt down-regulation leading to the good prognosis of patients with GC [133].
4.8 MiR-216a-3p BRD4 is a bromodomain and extra-terminal domain (BET) that has emerged as a therapeutic target in cancer therapy [134]. It seems that BRD4 overexpression is related to the EMT induction and tumor malignancy [135]. In GC, BRD4 seems to be involvement in GC progression by inhibition of Wnt3a. It has been reported that this inhibitory impact on the Wnt3a
ro
of
is mediated through miR-216a-3p upregulation [136].
4.9 MiR-23b-3p and miR-130a-5p
-p
Cannabinoid receptor 1 (CB1R) is a G-protein-coupled receptor and undergoes upregulation under the stimulation by endogenous ligands, anandmide and 2-arachidonoglycerol. It has been
re
demonstrated that a plant-derived compound known as Cannabis sativa induces CB1R through
lP
its ligand delta-9-tetrahydrocannabinol [137, 138]. It seems that the abnormal expression of CB1R occurs in a number of tumors [139]. In GC cells, the expressions of oncosuppressor miRs,
na
miR-23b-3p and miR-130a-5p significantly reduce resulting in upregulation of CB1R. The activated CB1R enhances the proliferation and malignancy of GC cells by stimulation of Wnt
ur
signaling pathway via upregulation of -catenin, c-Myc and cyclin D1 [140].
Jo
4.10 MiR-29c-3p
KIAA1199 responsible for nonsyndromic hair loss is related to the proliferation and viability of cancer cells, and poor prognosis of patients with GC through EMT induction [141-146]. This stimulatory effect has been shown on the malignancy of GC cells [147]. Activation of KIAA1199 occurs in GC cells leading to the high progression, lymph node metastasis and inhibition of apoptosis in GC cells. Investigation of molecular signaling pathways exhibits that KIAA1199 enhances the malignancy of GC cells by upregulation of Wnt/-catenin signaling pathway through EGFR. It seems that activation of KIAA1199 occurs by down-regulation of miR-29c-3p [148].
4.11 MiR-141-3p
Journal Pre-proof Signal transducer and activator of transcription 4 (STAT4) is suggested to be involvement in invasion and metastasis of cancer cells [89]. A same story occurs in GC. It appears that STAT4 undergoes upregulation in GC leading to the induction of Wnt/-catenin signaling pathway via on of oncosuppressor miR to reduce the malignancy of GC cells and induce apoptotic cell death by STAT4 inhibition [149].
4.12 MiR-125b Although miRs function as upstream modulators of Wnt signaling pathway in tumor cells, it has
of
been reported there are epigenetic factors regulating the function of miRs. KDM4 is a subfamily
ro
of dimethylases and accounts for controlling cell growth [150]. Accumulating data demonstrates that KDM4 upregulation considerably promotes the viability and proliferation of tumor cells
-p
through genomic instability [151]. More importantly, KDM4B increases the expression of miR125b. The induced miR-125b activates Wnt signaling pathway by upregulation of -catenin
lP
re
nuclear translocation [152].
4.13 MiR-375-3p
na
As a oncosuppressor miR, the down-regulation of miR-375-3p occurs during cancer to ensure the progression of tumor cells [153, 154]. Noteworthy, YWHAZ seems to be a potential prognostic marker of various cancers and has a negative relationship with miR-375-3p [155-158]. YWHAZ
ur
stimulates Wnt signaling pathway by nuclear translocation of -catenin. MiR-375-3p induces
Jo
apoptotic cell death in GC cells by inhibition of YWHAZ/Wnt axis [159].
4.14 MiR-142-5p
MiR-142 contains two major subsets including miR-142-3p as guide strand and miR-142-5p as passenger strand [160]. It seems that miR-142-5p suppresses apoptosis in cancer cells [161]. Besides, miR-142-5p has shown to enhance the proliferation of tumor cells via inhibition of TGF- signaling pathway [162]. These studies highlight the function of miR-142-5p as an oncogenesis miR. However, the strategy of miR-142-5p is a little different in GC, so that inhibition of miR-142-5p expression is associated with high proliferation capability of cancer cells. The inhibitory impact of this miR on GC cells is a result of Wnt pathway inhibition
Journal Pre-proof through CYR61. It seems that CYR61 enhances the nuclear translocation of -catenin and miR142-5p reduces the expression of CYR61 and subsequently, Wnt pathway [92].
4.15 MiR-106a-3p LncRNAs are RNA polymerase II transcripts with the size of 200 nucleotides [163]. During the past decades, much attention has been directed towards lncRNAs due to their critical roles in important biological processes. Aberration expression of lncRNAs is associated with development of pathological conditions, particularly cancer [164, 165]. LINCO1133 negatively
of
affects the proliferation of GC cells. It has been demonstrated that LINCO1133 down-regulates
ro
the expression of miR-106a-3p resulting in inhibition of EMT and stimulation of apoptosis. It is suggested that by miR-106a-3p inhibition, LINCO1133 exerts inhibitory effect on the nuclear
-p
translocation of -catenin leading to the inactivation of Wnt/-catenin signaling pathway and
re
reduced malignancy of cancer cells [166].
lP
4.16 MiR-361-5p
Studies show that EMT plays a significant role in tumorigenesis by transformation of epithelial
na
phenotype into mesenchymal one [167-169]. A number of molecular alterations occur during EMT induction. The expression of epithelial markers such as E-cadherin undergo down-
ur
regulation, while mesenchymal markers show high expression [170, 171]. Inhibition of EMT is a great strategy in cancer therapy. MiR-361-5p significantly diminishes the EMT by enhancing E-
Jo
cadherin levels while reducing N-cadherin. It appears that miR-361-5p inhibits EMT by decreasing the expression of TCF4, cyclin D1 and c-Myc as targets of Wnt signaling pathway [172].
5. Wnt and miRs interactions It seems that miRs have a great potential in regulation of Wnt signaling pathway. This regulation occurs in several stages as following: A) affecting GSK-3; B) upregulation/down-regulation of -catenin nuclear translocation; C) influencing the expression of -catenin and target genes such as cyclin D1 and c-Myc; D) affecting Wnt ligands such as Wnt1, Wnt3a and Wnt5a; and E) regulation of upstream modulators of Wnt signaling pathway such as Smad4, NKD1, TRIM24 and TRIMP29 (Figure 1).
Journal Pre-proof
6. Conclusion and remarks Accumulating data demonstrates that Wnt signaling aberration is associated with a number of pathological conditions, particularly cancer. Much attempt has been made in regulation of this important signaling pathway. In respect to the potential role of miRs in various important biological processes such as cell proliferation, cell growth and cell differentiation as well as apoptotic cell death, we concluded that Wnt signaling pathway may be a target of miRs. It was found that miRs based on their roles as being oncosuppressor or oncogenesis dually affect Wnt
of
signaling pathway to ensure the proliferation and malignancy of GC cells, while oncosuppressor
ro
miRs negatively affect Wnt signaling pathway to suppress GC invasion. However, more studies are needed to identify other miRs with their modulatory impact on Wnt signaling pathway.
-p
Competing interests: The authors declare no conflict of interest. Funding: Not applicable
re
Ethical approval and consent to participate: Not applicable
lP
Availability of supporting data: Not applicable
Authors' contributions: Study conception and design: S. S. and M.A. Acquisition of data: T.F., H.R. and R. M. Drafting of the manuscript: M. A., H.R. and R.M. Critical revision: S. S.
ur
Abbreviations:
na
Acknowledgements: Not applicable
Jo
GC, gastric cancer; WHO, World Health Organization; miR, microRNA; APC, adenomatous polyposiscoli; Dvl, dishevelled; CK1, casein kinase 1; GSK-3, glycogen synthase kinase-3; Fzd, Frizzled; LRP5/6, low-density lipoprotein receptor-associated protein 5/6; LEF, lymphoid enhancer binding factors; TCF, T-cell factors; PCP, planar cell polarity; PKC, protein kinase C; CaMKII, calmodulin-sensitive protein kinase II; NFAT, nuclear factor associated with T cells; JNK, c-Jun N-terminal kinase; TNBC, triple negative breast cancer; GBP-1, guanylate-binding protein-1; lncRNAs, long non-coding RNAs; RACK1, receptor for activated C kinase 1; FAM83D, family with sequence similarity 83, member D; pri-miR, primary-miR; DGCR8, DiGeorge syndrome critical region 8; pre-miR, precursor-miR; Exp5, Exportin 5; Ago2, Argonaute 2; TRBP, transactivating response RNA-binding protein; PACT, protein kinase Ractivating protein; NSCLC, non-small cell lung cancer; PITX1, paired-like homeodomain
Journal Pre-proof transcription factor 1; EMT, epithelial-to-mesenchymal transition; SIN, sinomenine; 3/ UTR, 3/ untranslated region; Ser9, Serin9; CB1R, cannabinoid receptor 1; STAT4, signal transducer and activator of transcription 4.
References: Biagioni, A., et al., Update on gastric cancer treatments and gene therapies. Cancer Metastasis
of
1.
Rev, 2019.
Malsin, E.S., et al., Macrophages as a Source and Recipient of Wnt Signals. Frontiers in
ro
2.
3.
-p
immunology, 2019. 10: p. 1813-1813.
Smyth, E.C. and M. Moehler, Late-line treatment in metastatic gastric cancer: today and
re
tomorrow. Therapeutic advances in medical oncology, 2019. 11: p. 17588359198675221758835919867522.
Smyth, E.C. and M. Moehler, Late-line treatment in metastatic gastric cancer: today and
lP
4.
tomorrow. Therapeutic Advances in Medical Oncology, 2019. 11: p. 1758835919867522. Charalampakis, N., et al., Medical management of gastric cancer: a 2017 update. Cancer Med, 2018. 7(1): p. 123-133.
Torre, L.A., et al., Global cancer statistics, 2012. CA: A Cancer Journal for Clinicians, 2015. 65(2): p. 87-108.
Bray, F., et al., Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality
Jo
7.
ur
6.
na
5.
worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018. 68(6): p. 394-424. 8.
Ishaq, S. and L. Nunn, Helicobacter pylori and gastric cancer: a state of the art review. Gastroenterology and hepatology from bed to bench, 2015. 8(Suppl 1): p. S6-S14.
9.
Kim, J., et al., Gene-diet interactions in gastric cancer risk: a systematic review. World journal of gastroenterology, 2014. 20(28): p. 9600-9610.
10.
Berlth, F., et al., Pathohistological classification systems in gastric cancer: diagnostic relevance and prognostic value. World J Gastroenterol, 2014. 20(19): p. 5679-84.
11.
Smyth, E.C., et al., Gastric cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol, 2016. 27(suppl 5): p. v38-v49.
Journal Pre-proof 12.
Moody, L., et al., The Efficacy of miR-20a as a Diagnostic and Prognostic Biomarker for Colorectal Cancer: A Systematic Review and Meta-Analysis. Cancers, 2019. 11(8): p. 1111.
13.
Ravegnini, G., et al., Prognostic Role of miR-221 and miR-222 Expression in Cancer Patients: A Systematic Review and Meta-Analysis. Cancers, 2019. 11(7): p. 970.
14.
Malhotra, P., G.H. Read, and J.B. Weidhaas, Breast Cancer and miR-SNPs: The Importance of miR Germ-Line Genetics. Non-coding RNA, 2019. 5(1): p. 27.
15.
Cui, M., et al., Interactive functions of microRNAs in the miR-23a-27a-24-2 cluster and the potential for targeted therapy in cancer. J Cell Physiol, 2019. Calin, G.A. and C.M. Croce, MicroRNA signatures in human cancers. Nature Reviews Cancer,
of
16.
2006. 6(11): p. 857-866.
Ha, M. and V.N. Kim, Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol, 2014. 15(8): p.
ro
17.
18.
-p
509-24.
Di Leva, G., M. Garofalo, and C.M. Croce, MicroRNAs in cancer. Annu Rev Pathol, 2014. 9: p. 287-
19.
re
314.
Garzon, R., G.A. Calin, and C.M. Croce, MicroRNAs in Cancer. Annu Rev Med, 2009. 60: p. 167-
Choi, J.D. and J.S. Lee, Interplay between Epigenetics and Genetics in Cancer. Genomics Inform, 2013. 11(4): p. 164-73.
na
20.
lP
79.
Gu, S. and M.A. Kay, How do miRNAs mediate translational repression? Silence, 2010. 1(1): p. 11.
22.
Menbari, M.N., et al., miR-483-3p suppresses the proliferation and progression of human triple
ur
21.
negative breast cancer cells by targeting the HDAC8 oncogene. J Cell Physiol, 2019. Stella di Stadio, C., et al., GKN1 expression in gastric cancer cells is negatively regulated by miR-
Jo
23.
544a. Biochimie, 2019. 24.
Yang, Y., et al., Reciprocal regulation of miR-206 and IL-6/STAT3 pathway mediates IL6-induced gefitinib resistance in EGFR-mutant lung cancer cells. J Cell Mol Med, 2019.
25.
Yu, X., et al., The miR-195 Axis Regulates Chemoresistance through TUBB and Lung Cancer Progression through BIRC5. Mol Ther Oncolytics, 2019. 14: p. 288-298.
26.
Li, L., et al., miR-21 modulates prostaglandin signaling and promotes gastric tumorigenesis by targeting 15-PGDH. Biochemical and biophysical research communications, 2018. 495(1): p. 928934.
27.
Cao, C., et al., LINC01303 functions as a competing endogenous RNA to regulate EZH2 expression by sponging miR-101-3p in gastric cancer. J Cell Mol Med, 2019.
Journal Pre-proof 28.
Jia, C., et al., miR-200a-3p plays tumor suppressor roles in gastric cancer cells by targeting KLF12. Artif Cells Nanomed Biotechnol, 2019. 47(1): p. 3697-3703.
29.
Liu, S., et al., miR-425-5p suppresses tumorigenesis and DDP resistance in human-prostate cancer by targeting GSK3beta and inactivating the Wnt/beta-catenin signaling pathway. J Biosci, 2019. 44(4).
30.
Svoronos, A.A., D.M. Engelman, and F.J. Slack, OncomiR or Tumor Suppressor? The Duplicity of MicroRNAs in Cancer. Cancer Res, 2016. 76(13): p. 3666-70.
31.
Zheng, S., et al., miR-183-5p enhances the radioresistance of colorectal cancer by directly
32.
of
targeting ATG5. J Biosci, 2019. 44(4). Zhou, Y., et al., MiR-1307 influences the chemotherapeutic sensitivity in ovarian cancer cells
Ding, F.N., et al., miR-122-5p modulates the radiosensitivity of cervical cancer cells by regulating
-p
33.
ro
through the regulation of the CIC transcriptional repressor. Pathol Res Pract, 2019: p. 152606.
cell division cycle 25CDC25A. FEBS Open Bio, 2019.
Mukherjee, T. and K.N. Balaji, The WNT Framework in Shaping Immune Cell Responses During
re
34.
Bacterial Infections. Frontiers in immunology, 2019. 10: p. 1985-1985. Mukherjee, T. and K.N. Balaji, The WNT Framework in Shaping Immune Cell Responses During
lP
35.
Bacterial Infections. Frontiers in Immunology, 2019. 10: p. 1985. Ram Makena, M., et al., Wnt/beta-Catenin Signaling: The Culprit in Pancreatic Carcinogenesis
na
36.
and Therapeutic Resistance. Int J Mol Sci, 2019. 20(17). Lang, C.M.R., et al., Wnt Signaling Pathways in Keratinocyte Carcinomas. Cancers (Basel), 2019. 11(9).
Kolluri, A. and M. Ho, The Role of Glypican-3 in Regulating Wnt, YAP, and Hedgehog in Liver
Jo
38.
ur
37.
Cancer. Frontiers in oncology, 2019. 9: p. 708-708. 39.
Raisch, J., A. Côté-Biron, and N. Rivard, A Role for the WNT Co-Receptor LRP6 in Pathogenesis and Therapy of Epithelial Cancers. Cancers, 2019. 11(8): p. 1162.
40.
Tang, K.L., et al., MiR-638 suppresses the progression of oral squamous cell carcinoma through wnt/beta-catenin pathway by targeting phospholipase D1. Artif Cells Nanomed Biotechnol, 2019. 47(1): p. 3278-3285.
41.
Clevers, H. and R. Nusse, Wnt/beta-catenin signaling and disease. Cell, 2012. 149(6): p. 1192205.
42.
Cui, J., et al., MicroRNA-545 targets ZEB2 to inhibit the development of non-small cell lung cancer by inactivating Wnt/beta-catenin pathway. Oncol Lett, 2019. 18(3): p. 2931-2938.
Journal Pre-proof 43.
Sun, Q., et al., Emetine exhibits anticancer activity in breast cancer cells as an antagonist of Wnt/betacatenin signaling. Oncol Rep, 2019.
44.
Sun, Y., et al., Inhibitor of DNA binding 1 (Id1) mediates stemness of colorectal cancer cells through the Id1-c-Myc-PLAC8 axis via the Wnt/beta-catenin and Shh signaling pathways. Cancer Manag Res, 2019. 11: p. 6855-6869.
45.
Jiang, S., et al., WNT5B governs the phenotype of basal-like breast cancer by activating WNT signaling. Cell Commun Signal, 2019. 17(1): p. 109.
46.
Hsu, C.C., et al., The differential distributions of ASPM isoforms and their roles in Wnt signaling,
47.
of
cell cycle progression, and pancreatic cancer prognosis. J Pathol, 2019. Galluzzi, L., et al., WNT Signaling in Cancer Immunosurveillance. Trends Cell Biol, 2019. 29(1): p.
Stamos, J.L. and W.I. Weis, The beta-catenin destruction complex. Cold Spring Harb Perspect
-p
48.
ro
44-65.
Biol, 2013. 5(1): p. a007898.
Behrens, J., et al., Functional interaction of an axin homolog, conductin, with beta-catenin, APC,
re
49.
and GSK3beta. Science, 1998. 280(5363): p. 596-9.
Staal, F.J. and H. Clevers, Tcf/Lef transcription factors during T-cell development: unique and
lP
50.
overlapping functions. Hematol J, 2000. 1(1): p. 3-6. Bastakoty, D. and P.P. Young, Wnt/beta-catenin pathway in tissue injury: roles in pathology and
na
51.
therapeutic opportunities for regeneration. Faseb j, 2016. 30(10): p. 3271-3284. Jeong, W.J., E.J. Ro, and K.Y. Choi, Interaction between Wnt/beta-catenin and RAS-ERK pathways
ur
52.
and an anti-cancer strategy via degradations of beta-catenin and RAS by targeting the
53.
Jo
Wnt/beta-catenin pathway. NPJ Precis Oncol, 2018. 2(1): p. 5. Veeman, M.T., J.D. Axelrod, and R.T. Moon, A second canon. Functions and mechanisms of betacatenin-independent Wnt signaling. Dev Cell, 2003. 5(3): p. 367-77. 54.
Kuhl, M., et al., The Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes shape. Trends Genet, 2000. 16(7): p. 279-83.
55.
Katoh, M., WNT/PCP signaling pathway and human cancer (review). Oncol Rep, 2005. 14(6): p. 1583-8.
56.
Ahumada, A., et al., Signaling of rat Frizzled-2 through phosphodiesterase and cyclic GMP. Science, 2002. 298(5600): p. 2006-10.
57.
Sheldahl, L.C., et al., Dishevelled activates Ca2+ flux, PKC, and CamKII in vertebrate embryos. J Cell Biol, 2003. 161(4): p. 769-77.
Journal Pre-proof 58.
Slusarski, D.C., V.G. Corces, and R.T. Moon, Interaction of Wnt and a Frizzled homologue triggers G-protein-linked phosphatidylinositol signalling. Nature, 1997. 390(6658): p. 410-3.
59.
Blumenthal, A., et al., The Wingless homolog WNT5A and its receptor Frizzled-5 regulate inflammatory responses of human mononuclear cells induced by microbial stimulation. Blood, 2006. 108(3): p. 965-73.
60.
Pashirzad, M., et al., Role of Wnt5a in the Pathogenesis of Inflammatory Diseases. J Cell Physiol, 2017. 232(7): p. 1611-1616.
61.
Pereira, C., et al., Wnt5A/CaMKII signaling contributes to the inflammatory response of
of
macrophages and is a target for the antiinflammatory action of activated protein C and interleukin-10. Arterioscler Thromb Vasc Biol, 2008. 28(3): p. 504-10.
Undi, R.B., et al., Wnt Signaling: Role in Regulation of Haematopoiesis. Indian J Hematol Blood
ro
62.
63.
-p
Transfus, 2016. 32(2): p. 123-34.
Xie, W., et al., Oxymatrine enhanced anti-tumor effects of Bevacizumab against triple-negative
re
breast cancer via abating Wnt/beta-Catenin signaling pathway. Am J Cancer Res, 2019. 9(8): p. 1796-1814.
Wang, J., et al., Guanylate-binding protein-2 inhibits colorectal cancer cell growth and increases
lP
64.
the sensitivity to paclitaxel of paclitaxel-resistant colorectal cancer cells by interfering Wnt
65.
na
signaling. J Cell Biochem, 2019.
Luo, N.Q., et al., Long non-coding RNA ENST00000434223 inhibits the progression of renal
p. 6868-6877.
Wang, H., et al., LncRNA MIR4435-2HG targets desmoplakin and promotes growth and
Jo
66.
ur
cancer through Wnt/hygro-catenin signaling pathway. Eur Rev Med Pharmacol Sci, 2019. 23(16):
metastasis of gastric cancer by activating Wnt/beta-catenin signaling. Aging (Albany NY), 2019. 11. 67.
Deng, Q., et al., Activation of Hedgehog signaling in mesenchymal stem cells induces cartilage and bone tumor formation via Wnt/beta-Catenin. Elife, 2019. 8.
68.
Sun, Q.X., et al., Dysregulation of miR-204-3p Driven by the Viability and Motility of Retinoblastoma via Wnt/beta-catenin Pathway In Vitro and In Vivo. Pathol Oncol Res, 2019.
69.
Zhu, L., et al., Upregulated RACK1 attenuates gastric cancer cell growth and epithelialmesenchymal transition via suppressing Wnt/beta-catenin signaling. Onco Targets Ther, 2019. 12: p. 4795-4805.
Journal Pre-proof 70.
Wang, F., et al., Upregulation of family with sequence similarity 83 member D expression enhances cell proliferation and motility via activation of Wnt/beta-catenin signaling and predicts poor prognosis in gastric cancer. Cancer Manag Res, 2019. 11: p. 6775-6791.
71.
Deng, P.-c., et al., LncRNA SNHG14 potentiates pancreatic cancer progression via modulation of annexin A2 expression by acting as a competing endogenous RNA for miR-613. Journal of Cellular and Molecular Medicine. 0(0).
72.
Lange, S., et al., miR-486 is modulated by stretch and increases ventricular growth. JCI Insight, 2019. Mohajeri, M., et al., MicroRNAs: Novel molecular targets and response modulators of statin therapy. Trends in pharmacological sciences, 2018.
Borchert, G.M., W. Lanier, and B.L. Davidson, RNA polymerase III transcribes human microRNAs.
ro
74.
of
73.
75.
-p
Nature structural & molecular biology, 2006. 13(12): p. 1097.
Berezikov, E., V. Guryev, and E. Cuppen, Exploring conservation of transcription factor binding
76.
re
sites with CONREAL, in Comparative Genomics. 2007, Springer. p. 437-448. MacRae, I.J., K. Zhou, and J.A. Doudna, Structural determinants of RNA recognition and cleavage
77.
Khvorova, A., A. Reynolds, and S.D. Jayasena, Functional siRNAs and miRNAs exhibit strand bias.
na
Cell, 2003. 115(2): p. 209-216. 78.
lP
by Dicer. Nature structural & molecular biology, 2007. 14(10): p. 934.
Quattrochi, B., et al., MicroRNAs of the mir-17~ 92 cluster regulate multiple aspects of
79.
ur
pancreatic tumor development and progression. Oncotarget, 2017. 8(22): p. 35902. Soleimani, A., et al., Role of TGF‐β signaling regulatory microRNAs in the pathogenesis of
80.
Jo
colorectal cancer. Journal of cellular physiology, 2019. Yue, X., F. Lan, and T. Xia, Hypoxic Glioma Cell-Secreted Exosomal miR-301a Activates Wnt/betacatenin Signaling and Promotes Radiation Resistance by Targeting TCEAL7. Mol Ther, 2019. 81.
Ye, K., C. Xu, and T. Hui, MiR-34b inhibits the proliferation and promotes apoptosis in colon cancer cells by targeting Wnt/beta-catenin signaling pathway. Biosci Rep, 2019.
82.
Liu, Q., et al., miR-504 suppresses mesenchymal phenotype of glioblastoma by directly targeting the FZD7-mediated Wnt-beta-catenin pathway. J Exp Clin Cancer Res, 2019. 38(1): p. 358.
83.
Zhao, G., Y. Yin, and B. Zhao, miR-140-5p is negatively correlated with proliferation, invasion, and tumorigenesis in malignant melanoma by targeting SOX4 via the Wnt/beta-catenin and NFkappaB cascades. J Cell Physiol, 2019.
Journal Pre-proof 84.
Che, Q.Q., et al., Effect of miR-124 on neuronal apoptosis in rats with cerebral infarction through Wnt/beta-catenin signaling pathway. Eur Rev Med Pharmacol Sci, 2019. 23(15): p. 6657-6664.
85.
Zhao, D.L. and Q.L. Wu, Effect of inhibition to Yes-related proteins-mediated Wnt/beta-catenin signaling pathway through miR-195-5p on apoptosis of gastric cancer cells. Eur Rev Med Pharmacol Sci, 2019. 23(15): p. 6486-6496.
86.
Liu, Q.P., et al., MiR-140-3p overexpression activates the Wnt signaling pathway to promote fracture healing. Eur Rev Med Pharmacol Sci, 2019. 23(14): p. 6011-6017.
87.
Liu, L., W. Qu, and Z. Zhong, Down-regulation of miR-503 expression predicate advanced
of
mythological features and poor prognosis in patients with NSCLC. International journal of clinical and experimental pathology, 2015. 8(5): p. 5609.
Chong, Y., et al., MicroRNA-503 acts as a tumor suppressor in osteosarcoma by targeting L1CAM.
ro
88.
89.
-p
PloS one, 2014. 9(12): p. e114585.
Zhou, X., et al., Down-regulation of miR-141 induced by helicobacter pylori promotes the
re
invasion of gastric cancer by targeting STAT4. Cellular Physiology and Biochemistry, 2014. 33(4): p. 1003-1012.
Peng, Y., et al., microRNA‑503 inhibits gastric cancer cell growth and epithelial ‑to‑mesenchymal
lP
90.
transition. Oncology letters, 2014. 7(4): p. 1233-1238. Wang, T., et al., MiR-503 regulates cisplatin resistance of human gastric cancer cell lines by
na
91.
targetingIGF1RandBCL2. Chinese medical journal, 2014. 127(12): p. 2357-2362. Yan, J., et al., Downregulation of miR-142-5p promotes tumor metastasis through directly
ur
92.
regulating CYR61 expression in gastric cancer. Gastric Cancer, 2019. 22(2): p. 302-313. Lamonerie, T., et al., Ptx1, a bicoid-related homeo box transcription factor involved in
Jo
93.
transcription of the pro-opiomelanocortin gene. Genes & development, 1996. 10(10): p. 12841295. 94.
Qiao, F., et al., Downregulated PITX1 modulated by MiR-19a-3p promotes cell malignancy and predicts a poor prognosis of gastric cancer by affecting transcriptionally activated PDCD5. Cellular Physiology and Biochemistry, 2018. 46(6): p. 2215-2231.
95.
Chen, Y.-N., et al., Expression of pituitary homeobox 1 gene in human gastric carcinogenesis and its clinicopathological significance. World journal of gastroenterology: WJG, 2008. 14(2): p. 292.
96.
Liu, L., et al., MiR-675 is frequently overexpressed in gastric cancer and enhances cell proliferation and invasion via targeting a potent anti-tumor gene PITX1. Cellular signalling, 2019. 62: p. 109352.
Journal Pre-proof 97.
Ahmadi, Z., R. Mohammadinejad, and M. Ashrafizadeh, Drug delivery systems for resveratrol, a non-flavonoid polyphenol: Emerging evidence in last decades. Journal of Drug Delivery Science and Technology, 2019.
98.
Ashrafizadeh, M., et al., Autophagy, anoikis, ferroptosis, necroptosis, and endoplasmic reticulum stress: Potential applications in melanoma therapy. Journal of cellular physiology, 2019.
99.
Mohammadinejad, R., et al., Berberine as a potential autophagy modulator. Journal of cellular physiology, 2019.
100.
Ahmadi, Z. and M. Ashrafizadeh, Melatonin as a potential modulator of Nrf2. Fundamental &
101.
of
clinical pharmacology, 2019. Ahmadi, Z., et al., The Targeting of Autophagy and Endoplasmic Reticulum Stress Mechanisms by
Ashrafizadeh, M., et al., Monoterpenes modulating autophagy: A review study. Basic & Clinical
-p
102.
ro
Honokiol Therapy. Reviews in Clinical Medicine, 2019. 6(2): p. 66-73.
Pharmacology & Toxicology.
Wang, W., et al., Sinomenine attenuates angiotensin II-induced autophagy via inhibition of P47-
re
103.
phox translocation to the membrane and influences reactive oxygen species generation in
104.
lP
podocytes. Kidney and Blood Pressure Research, 2016. 41(2): p. 158-167. LIN, X.-j., X.-y. CAI, and J.-h. YE, The clinical observition on Sinomenine for Rheumatoid Arthritis
105.
na
[J]. Journal of Traditional Chinese Medicine University of Hunan, 2009. 8. Zhou, L., et al., Activation of PI3K/Akt and ERK signaling pathways antagonized sinomenine-
106.
ur
induced lung cancer cell apoptosis. Molecular medicine reports, 2012. 5(5): p. 1256-1260. Deng, F., et al., The pro-apoptosis effect of sinomenine in renal carcinoma via inducing
Jo
autophagy through inactivating PI3K/AKT/mTOR pathway. Biomedicine & Pharmacotherapy, 2018. 97: p. 1269-1274. 107.
Song, L., et al., Sinomenine inhibits breast cancer cell invasion and migration by suppressing NFκB activation mediated by IL-4/miR-324-5p/CUEDC2 axis. Biochemical and biophysical research communications, 2015. 464(3): p. 705-710.
108.
Jiang, S., et al., Sinomenine inhibits A549 human lung cancer cell invasion by mediating the STAT3 signaling pathway. Oncology letters, 2016. 12(2): p. 1380-1386.
109.
Yuan, H., et al., Sinomenine exerts anti‐tumor effect in gastric cancer cells via enhancement of miR‐204 expression. Basic & Clinical Pharmacology & Toxicology.
110.
Mendell, J.T., miRiad roles for the miR-17-92 cluster in development and disease. Cell, 2008. 133(2): p. 217-222.
Journal Pre-proof 111.
Jepsen, R.K., et al., Early metastatic colorectal cancers show increased tissue expression of miR17/92 cluster members in the invasive tumor front. Human pathology, 2018. 80: p. 231-238.
112.
Kandalam, M.M., et al., Oncogenic microRNA 17-92 cluster is regulated by epithelial cell adhesion molecule and could be a potential therapeutic target in retinoblastoma. Molecular vision, 2012. 18: p. 2279.
113.
Chen, F.F., et al., Function and mechanism by which interferon regulatory factor ‑1 inhibits oncogenesis. Oncology letters, 2013. 5(2): p. 417-423.
114.
Kröger, A., et al., Activities of IRF-1. Journal of interferon & cytokine research, 2002. 22(1): p. 5-
115.
of
14. Nozawa, H., et al., Functionally inactivating point mutation in the tumor‐suppressor IRF‐1 gene
Yuan, J., et al., MIR17HG-miR-18a/19a axis, regulated by interferon regulatory factor-1,
-p
116.
ro
identified in human gastric cancer. International journal of cancer, 1998. 77(4): p. 522-527.
promotes gastric cancer metastasis via Wnt/β-catenin signalling. Cell death & disease, 2019.
117.
re
10(6): p. 454-454.
Bao, L., et al., MiR-876-5p suppresses epithelial–mesenchymal transition of lung cancer by
lP
directly down-regulating bone morphogenetic protein 4. Journal of biosciences, 2017. 42(4): p. 671-681.
Dong, Y., et al., MiR-876-5p modulates head and neck squamous cell carcinoma metastasis and
na
118.
invasion by targeting vimentin. Cancer cell international, 2018. 18(1): p. 121. Wang, Y., et al., MiR-876-5p acts as an inhibitor in hepatocellular carcinoma progression by
ur
119.
targeting DNMT3A. Pathology-Research and Practice, 2018. 214(7): p. 1024-1030. Xie, W., et al., MicroRNA‐876‐5p inhibits cell proliferation, migration and invasion by targeting
Jo
120.
c‐Met in osteosarcoma. Journal of cellular and molecular medicine, 2019. 23(5): p. 3293-3301. 121.
Xu, Q., et al., MicroRNA-876-5p inhibits epithelial-mesenchymal transition and metastasis of hepatocellular carcinoma by targeting BCL6 corepressor like 1. Biomedicine & Pharmacotherapy, 2018. 103: p. 645-652.
122.
Zhi, Y., et al., FOXM1-Mediated LINC-ROR Regulates the Proliferation and Sensitivity to Sorafenib in Hepatocellular Carcinoma. Molecular Therapy-Nucleic Acids, 2019. 16: p. 576-588.
123.
Xu, Z., et al., MiR-876-5p regulates gastric cancer cell proliferation, apoptosis and migration through targeting WNT5A and MITF. Bioscience reports, 2019. 39(6): p. BSR20190066.
124.
Fang, F., et al., MicroRNA-188-5p suppresses tumor cell proliferation and metastasis by directly targeting FGF5 in hepatocellular carcinoma. Journal of hepatology, 2015. 63(4): p. 874-885.
Journal Pre-proof 125.
Li, N., et al., miR-188 inhibits glioma cell proliferation and cell cycle progression through targeting β-catenin. Oncology Research Featuring Preclinical and Clinical Cancer Therapeutics, 2018. 26(5): p. 785-794.
126.
Zhang, H., et al., miR-188-5p inhibits tumour growth and metastasis in prostate cancer by repressing LAPTM4B expression. Oncotarget, 2015. 6(8): p. 6092.
127.
Zhao, L., et al., MiroRNA-188 acts as tumor suppressor in non-small-cell lung Cancer by targeting MAP3K3. Molecular pharmaceutics, 2018. 15(4): p. 1682-1689.
128.
Li, Y., et al., Aberrantly expressed miR-188-5p promotes gastric cancer metastasis by activating
129.
of
Wnt/β-catenin signaling. BMC cancer, 2019. 19(1): p. 505-505. Franklin, R.B. and L.C. Costello, The important role of the apoptotic effects of zinc in the
Yan, G., et al., Slc39a7/zip7 plays a critical role in development and zinc homeostasis in zebrafish.
-p
130.
ro
development of cancers. Journal of cellular biochemistry, 2009. 106(5): p. 750-757.
PloS one, 2012. 7(8): p. e42939.
Cousins, R.J., J.P. Liuzzi, and L.A. Lichten, Mammalian zinc transport, trafficking, and signals.
re
131.
Journal of Biological Chemistry, 2006. 281(34): p. 24085-24089. Taylor, K.M., et al., Structure-function analysis of HKE4, a member of the new LIV-1 subfamily of
lP
132.
zinc transporters. Biochemical Journal, 2004. 377(1): p. 131-139. Cui, Y., et al., miR-15a-3p Suppresses Prostate Cancer Cell Proliferation and Invasion by Targeting
na
133.
SLC39A7 Via Downregulating Wnt/β-Catenin Signaling Pathway. Cancer biotherapy &
134.
ur
radiopharmaceuticals, 2019.
Jin, X., et al., DUB3 promotes BET inhibitor resistance and cancer progression by deubiquitinating
135.
Jo
BRD4. Molecular cell, 2018. 71(4): p. 592-605. e4. Zhang, P., et al., BRD4 promotes tumor growth and epithelial-mesenchymal transition in hepatocellular carcinoma. International journal of immunopathology and pharmacology, 2015. 28(1): p. 36-44. 136.
Song, H., et al., BRD4 promotes the stemness of gastric cancer cells via attenuating miR-216a-3pmediated inhibition of Wnt/β-catenin signaling. European journal of pharmacology, 2019. 852: p. 189-197.
137.
Gerard, C.M., et al., Molecular cloning of a human cannabinoid receptor which is also expressed in testis. Biochemical Journal, 1991. 279(1): p. 129-134.
138.
Matsuda, L.A., et al., Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature, 1990. 346(6284): p. 561.
Journal Pre-proof 139.
Chakravarti, B., J. Ravi, and R.K. Ganju, Cannabinoids as therapeutic agents in cancer: current status and future implications. Oncotarget, 2014. 5(15): p. 5852.
140.
Xian, X., et al., miR-23b-3p and miR-130a-5p affect cell growth, migration and invasion by targeting CB1R via the Wnt/β-catenin signaling pathway in gastric carcinoma. OncoTargets and therapy, 2018. 11: p. 7503-7512.
141.
Abe, S., S.-i. Usami, and Y. Nakamura, Mutations in the gene encoding KIAA1199 protein, an inner-ear protein expressed in Deiters' cells and the fibrocytes, as the cause of nonsyndromic hearing loss. Journal of human genetics, 2003. 48(11): p. 564. Nagase, T., et al., Prediction of the coding sequences of unidentified human genes. XV. The
of
142.
complete sequences of 100 new cDNA clones from brain which code for large proteins in vitro.
Li, L., et al., Central role of CEMIP in tumorigenesis and its potential as therapeutic target. Journal of Cancer, 2017. 8(12): p. 2238.
Evensen, N.A., et al., Unraveling the role of KIAA1199, a novel endoplasmic reticulum protein, in
re
144.
-p
143.
ro
Dna Research, 1999. 6(5): p. 337-345.
cancer cell migration. Journal of the National Cancer Institute, 2013. 105(18): p. 1402-1416. Michishita, E., et al., Upregulation of the KIAA1199 gene is associated with cellular mortality.
lP
145.
Cancer letters, 2006. 239(1): p. 71-77.
Yoshida, H., et al., KIAA1199, a deafness gene of unknown function, is a new hyaluronan binding
na
146.
protein involved in hyaluronan depolymerization. Proceedings of the National Academy of
147.
ur
Sciences, 2013. 110(14): p. 5612-5617. Matsuzaki, S., et al., Clinicopathologic significance of KIAA1199 overexpression in human gastric
148.
Jo
cancer. Annals of surgical oncology, 2009. 16(7): p. 2042-2051. Wang, L., et al., The miR-29c-KIAA1199 axis regulates gastric cancer migration by binding with WBP11 and PTP4A3. Oncogene, 2019. 38(17): p. 3134. 149.
Zhou, Y., et al., MiR-141-3p suppresses gastric cancer induced transition of normal fibroblast and BMSC to cancer-associated fibroblasts via targeting STAT4. Experimental and molecular pathology, 2019. 107: p. 85-94.
150.
Young, L.C., D.W. McDonald, and M.J. Hendzel, Kdm4b histone demethylase is a DNA damage response protein and confers a survival advantage following γ-irradiation. Journal of Biological Chemistry, 2013. 288(29): p. 21376-21388.
151.
Berry, W.L., T.-D. Kim, and R. Janknecht, Stimulation of β-catenin and colon cancer cell growth by the KDM4B histone demethylase. International journal of oncology, 2014. 44(4): p. 1341-1348.
Journal Pre-proof 152.
Jing, J.C., et al., KDM4B promotes gastric cancer metastasis by regulating miR‐125b‐mediated activation of Wnt signaling. Journal of cellular biochemistry, 2019. 120(5): p. 7897-7906.
153.
Kang, W., et al., miR-375 is involved in Hippo pathway by targeting YAP1/TEAD4-CTGF axis in gastric carcinogenesis. Cell death & disease, 2018. 9(2): p. 92.
154.
Yan, J.W., J.S. Lin, and X.X. He, The emerging role of miR‐375 in cancer. International journal of cancer, 2014. 135(5): p. 1011-1018.
155.
Nishimura, Y., et al., Overexpression of YWHAZ relates to tumor cell proliferation and malignant outcome of gastric carcinoma. British journal of cancer, 2013. 108(6): p. 1324. Fan, T., et al., Up-regulation of 14-3-3ζ in lung cancer and its implication as prognostic and
of
156.
therapeutic target. Cancer research, 2007. 67(16): p. 7901-7906.
Neal, C.L., et al., 14-3-3ζ overexpression defines high risk for breast cancer recurrence and
ro
157.
158.
-p
promotes cancer cell survival. Cancer research, 2009. 69(8): p. 3425-3432. Ralhan, R., et al., Prognostic significance of head-and-neck cancer biomarkers discovered using
re
isobaric mass tagging and multidimensional liquid chromatography-tandem mass spectrometry. 2008, AACR.
Guo, F., et al., miR-375-3p/YWHAZ/beta-catenin axis regulates migration, invasion, EMT in
lP
159.
gastric cancer cells. Clin Exp Pharmacol Physiol, 2019. 46(2): p. 144-152. Shrestha, A., et al., MicroRNA‐142 is a multifaceted regulator in organogenesis, homeostasis,
na
160.
and disease. Developmental Dynamics, 2017. 246(4): p. 285-290. Liu, S., et al., miR-142-5p promotes development of colorectal cancer through targeting SDHB
ur
161.
and facilitating generation of aerobic glycolysis. Biomedicine & Pharmacotherapy, 2017. 92: p.
162.
Jo
1119-1127.
Ma, Z., et al., MicroRNA regulatory pathway analysis identifies miR-142-5p as a negative regulator of TGF-β pathway via targeting SMAD3. Oncotarget, 2016. 7(44): p. 71504.
163.
Sun, G.-L., et al., miR-324-3p promotes gastric cancer development by activating Smad4mediated Wnt/beta-catenin signaling pathway. Journal of gastroenterology, 2018. 53(6): p. 725739.
164.
Bracken, C.P., H.S. Scott, and G.J. Goodall, A network-biology perspective of microRNA function and dysfunction in cancer. Nature Reviews Genetics, 2016. 17(12): p. 719.
165.
Xue, X., et al., LncRNA HOTAIR enhances ER signaling and confers tamoxifen resistance in breast cancer. Oncogene, 2016. 35(21): p. 2746.
Journal Pre-proof 166.
Yang, X.-Z., et al., LINC01133 as ceRNA inhibits gastric cancer progression by sponging miR-106a3p to regulate APC expression and the Wnt/β-catenin pathway. Molecular cancer, 2018. 17(1): p. 126.
167.
Kang, Y. and J. Massagué, Epithelial-mesenchymal transitions: twist in development and metastasis. Cell, 2004. 118(3): p. 277-279.
168.
Hu, S., et al., miR-532 promoted gastric cancer migration and invasion by targeting NKD1. Life sciences, 2017. 177: p. 15-19.
169.
Zha, L., et al., HMGA2 elicits EMT by activating the Wnt/β-catenin pathway in gastric cancer.
170.
of
Digestive diseases and sciences, 2013. 58(3): p. 724-733. Hsu, Y.-M., et al., KCl cotransporter-3 down-regulates E-cadherin/β-catenin complex to promote
Gloushankova, N.A., S.N. Rubtsova, and I.Y. Zhitnyak, Cadherin-mediated cell-cell interactions in
-p
171.
ro
epithelial-mesenchymal transition. Cancer research, 2007. 67(22): p. 11064-11073.
normal and cancer cells. Tissue barriers, 2017. 5(3): p. e1356900. Tian, L., et al., MiR-361-5p inhibits the mobility of gastric cancer cells through suppressing
re
172.
epithelial-mesenchymal transition via the Wnt/β-catenin pathway. Gene, 2018. 675: p. 102-109. Liu, L., et al., MiR-675 is frequently overexpressed in gastric cancer and enhances cell
lP
173.
109352. 174.
na
proliferation and invasion via targeting a potent anti-tumor gene PITX1. Cell Signal, 2019. 62: p.
Yuan, H., et al., Sinomenine exerts antitumour effect in gastric cancer cells via enhancement of
175.
ur
miR-204 expression. Basic Clin Pharmacol Toxicol, 2019. Yuan, J., et al., MIR17HG-miR-18a/19a axis, regulated by interferon regulatory factor-1,
p. 454. 176.
Jo
promotes gastric cancer metastasis via Wnt/beta-catenin signalling. Cell Death Dis, 2019. 10(6):
Li, Y., et al., Aberrantly expressed miR-188-5p promotes gastric cancer metastasis by activating Wnt/beta-catenin signaling. BMC Cancer, 2019. 19(1): p. 505.
177.
Zhao, D. and Q. Wu, Effect of inhibition to Yes-related proteins-mediated Wnt/β-catenin signaling pathway through miR-195-5p on apoptosis of gastric cancer cells. European review for medical and pharmacological sciences, 2019. 23(15): p. 6486-6496.
178.
Li, H., et al., Targeting of GSK-3β by miR-214 to facilitate gastric cancer cell proliferation and decrease of cell apoptosis. Eur Rev Med Pharmacol Sci, 2018. 22(1): p. 127-134.
179.
Huang, J., et al., miR-302b inhibits tumorigenesis by targeting EphA2 via Wnt/β-catenin/EMT signaling cascade in gastric cancer. BMC cancer, 2017. 17(1): p. 886.
Journal Pre-proof 180.
Zhang, X., et al., SMG‐1 inhibition by miR‐192/‐215 causes epithelial‐mesenchymal transition in gastric carcinogenesis via activation of Wnt signaling. Cancer medicine, 2018. 7(1): p. 146-156.
181.
Huang, T., et al., SRGAP1, a crucial target of miR-340 and miR-124, functions as a potential oncogene in gastric tumorigenesis. Oncogene, 2018. 37(9): p. 1159.
182.
Zhang, L., et al., SLC34A2 regulates miR‐25‐Gsk3β signaling pathway to affect tumor progression in gastric cancer stem cell‐like cells. Molecular carcinogenesis, 2018. 57(3): p. 440-450.
183.
Cheng, C., et al., Knockdown of long non-coding RNA HOTAIR inhibits cisplatin resistance of gastric cancer cells through inhibiting the PI3K/Akt and Wnt/β-catenin signaling pathways by up-
of
regulating miR-34a. International journal of biological macromolecules, 2018. 107: p. 26202629.
Yue, H., et al., MIR-519d suppresses the gastric cancer epithelial-mesenchymal transition via
ro
184.
-p
Twist1 and inhibits Wnt/β-catenin signaling pathway. American journal of translational research, 2017. 9(8): p. 3654.
Yang, D., D. Zhao, and X. Chen, MiR-133b inhibits proliferation and invasion of gastric cancer
re
185.
cells by up-regulating FBN1 expression. Cancer Biomarkers, 2017. 19(4): p. 425-436. Fang, Z., et al., Regulation of TRIM24 by miR-511 modulates cell proliferation in gastric cancer.
lP
186.
Journal of Experimental & Clinical Cancer Research, 2017. 36(1): p. 17. Fan, D., et al., Upregulation of miR-501-5p activates the wnt/β-catenin signaling pathway and
na
187.
enhances stem cell-like phenotype in gastric cancer. Journal of Experimental & Clinical Cancer
188.
ur
Research, 2016. 35(1): p. 177.
Cao, D., et al., 18beta-glycyrrhetinic acid suppresses gastric cancer by activation of miR-149-3p-
189.
Jo
Wnt-1 signaling. Oncotarget, 2016. 7(44): p. 71960-71973. Yanaka, Y., et al., miR-544a induces epithelial-mesenchymal transition through the activation of WNT signaling pathway in gastric cancer. Carcinogenesis, 2015. 36(11): p. 1363-71. 190.
Qiu, F., et al., TRIM29 functions as an oncogene in gastric cancer and is regulated by miR-185. Int J Clin Exp Pathol, 2015. 8(5): p. 5053-61.
191.
Tang, H., et al., Diallyl disulfide suppresses proliferation and induces apoptosis in human gastric cancer through Wnt-1 signaling pathway by up-regulation of miR-200b and miR-22. Cancer Lett, 2013. 340(1): p. 72-81.
Table 1: Wnt-regulating miRs in gastric cancer malignancy. MiR
Cell line
Effect
on Interaction
Major outcomes
Refs
Journal Pre-proof Wnt pathway
MiR876-5p
MiR-188-5p
Inhibition
Induction
Inhibition
Jo
MiR-195-5p
GC cells
MiR-216a3p
Gastric cancer cell lines Inhibition AGS, BGC-823, MKN-45, MGC-803, SCG-7901 GC cell lines, BGC823, Inhibition MGC803, SGC7901, AGS and N87
MiR-142-5p
MiR-106a3p
of
ro
Induction
ur
MiR-15a-3p
Human GC cell lines (MKN45, AGS and SGC7901) Human GC cell lines (MGC803, MKN-45, MKN-28) Human GC cell lines (MKN74, AGS, KATOIII, NUGC3, MGC803, MKN45, and HGC27) Tumor xenografts Human PCa cell lines, including PC3, DU145, LNCaP, and 22Rv1
-p
MiR-17HG
Inhibition
Human GC cell lines Induction (SUN-216, BGC-823, AGS, BGC-803, NUGC4,
DownSuppressing the migration and regulation of invasion of cancer cells Wnt3a and catenin Expression of Enhancing the malignancy of Wnt cancer cells by stimulation of Wnt signaling pathway DownInhibition of EMT and regulation of reducing the proliferation and Wnt5a migration of cancer cells Enhancing the Enhancing the migration and level of p-GSK- invasion of cancer cells 3
re
MiR-204
Enhancing the Promoting the progression and [173] nuclear induction of EMT translocation of -catenin
lP
MiR-675
Human GC cell lines Inhibition MKN-45, BGC-823, SGC7901, MKN-28, and AGS Xenograft tumor model Human gastric normal Induction epithelial mucosa cell line GES-1 and GC cell lines (MGC-803, SGC-7901 and AGS) GC cells of AGS, MKN28, Inhibition MKN45 and SGC‐7901
na
MiR-503
between miR and Wnt pathway Elevating GSK- Suppressing the proliferation [152] 3 and p-- and invasion of cancer cells catenin
Downregulation nuclear catenin translocation Inhibition of catenin expression Inhibition Wnt3a
[174]
[175]
[123]
[176]
Inhibition of Wnt pathway [133] of results in down-regulation of - SLC39A7 and subsequently, decreased migration and viability of cancer cells - Induction of apoptotic cell [177] death of Reducing the progression and [136] malignancy of cancer cells
Downregulation of nuclear catenin translocation Enhancing the nuclear translocation of
Induction of apoptotic cell [92] death and reducing the migration capability
Elevating EMT and inhibition [166] of apoptosis
Journal Pre-proof
MiR-630
Human gastric cancer cell Induction lines SGC-7901, BGC-823, MGC803
MiR-214
GC tissues
MiR-302b
Human gastric Inhibition adenocarcinoma cell line SGC-7901
lP
cell
lines Induction
Inhibition
Jo
ur
MiR-340 GC cell lines and miR124
-p
Inhibition
na
MiR-192 Human GC and miR- BGC‐823 215
MiR-25
Animal models patients with GC
and Induction
MiR-324-3p
GC cells and tissues
Induction
MiR-34a
GC cell lines
Inhibition
Inactivation of Wnt signaling [172] pathway suppresses EMT mechanism
Inhibition of growth, invasion [26] and migration of cancer cells
Suppressing the EMT and [178] stimulation of apoptosis
of
Human gastric carcinoma Inhibition cell line NCI-N87
ro
MiR-3200
Reducing the expression of targets genes such as cyclin D1 and c-Myc Downregulation of catenin, Wnt3a and Wnt5a Upregulation of -catenin, Wnt3a and Wnt5a Downregulation of GSK-3 Decreasing the expression of catenin
re
MiR-361-5p
-catenin
MKN74, MKN45, SGC7901, and HGC-27) GC cell lines (SGC-7901, Inhibition MGC-803, MKN-28, TMK-1)
Enhancing the expression of target genes such as cyclin D1 Downregulation of nuclear catenin translocation Downregulation of GSK-3
Smad4/Wnt signaling pathway stimulation Downregulation of catenin and Wnt1
Promoting the proliferation of [178] cancer cells and inhibition of apoptosis Prevention of EMT [179] mechanism and subsequently, inhibition of invasion and metastasis Induction of EMT and [180] promoting the viability of cancer cells
Down-regulation of signaling pathway suppressing SRGAP1
Wnt [181] by
SLC34A2 enhances the [182] malignancy of GC cells by down-regulation of miR-25 leading to the activation of Wnt signaling pathway Stimulation of an increase in [163] cell growth and migration, and a decrease in apoptosis HOTAIR induces [183] chemotherapy resistance in GC cancer cells by inhibition of miR-34a and subsequently, stimulation of Wnt signaling
Journal Pre-proof
Human GC cell lines Inhibition (SGC-7901, BGC-823, SGC-7901 and HGC-27) GC cell lines AGS, HGC- Inhibition 27, KATO III, HGC-27, 108 and NCI-N87 GC tissues GC tissues and cells Induction
MiR-149-3p
Transgenic mice
MiR-544a
GC cell line, MKN1
MiR-185
GC-derived MGC803
MiR-200b and miR-22
MGC-803 cells
Inhibition
of
MiR-501-5p
GC tissues Inhibition Human gastric cancer cell lines (AGS, BGC823, MGC803, HGC-27and SGC7901) GC cells and tissues Induction
na ur
line Inhibition
Jo
cell
Induction
Inhibition
Suppressing the proliferation [185] and invasion of cancer cells through FBN1/Wnt axis
MiR-532 exerts inhibitory [168] effect on the NKD1 to induce Wnt signaling pathway resulting in increased cancer progression DownInhibition of Wnt signaling [186] via TRIM24 regulation of - pathway catenin, cyclin inhibition leading to the decreased cancer malignancy D1 and c-Myc
lP
MiR-511
Downregulation GSK-3 Downregulation Wnt1
Promoting the malignancy of [187] of cancer cells through Wnt induction Attenuation of inflammatory [188] of microenvironment and induction of apoptosis by inhibition of Wnt signaling pathway Upregulation of Induction of EMT [189] nuclear catenin translocation DownMiR-185 inhibits TRIMP29 to [190] Wnt pathway regulation of - suppress resulting in decreased catenin, cyclin malignancy of cancer cells D1 and c-Myc DownStimulation of apoptosis and [191] regulation of inhibition of proliferation Wnt1
Figure 1. Regulation of Wnt signaling pathway by miRs in GC cells. MiR, microRNA; APC, adenomatous polyposiscoli; Dvl, Dishevelled; CK1, casein kinase 1; GSK-3, glycogen synthase kinase -3.
Graphical abstract
[184]
ro
MiR-532
Downregulation of catenin and Wnt1 Upregulation of nuclear catenin translocation
-p
MiR-133b
re
MiR-519d
pathway Upregulation of Inhibition of EMT mechanism p-GSK-3
Figure 1