AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (3.4 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

MiR-378a-3p acts as a tumor suppressor in gastric cancer via directly targeting RAB31 and inhibiting the Hedgehog pathway proteins GLI1/2

Xinxin Xu1,2,*Yang Li1,2,*Guoxiao Liu1,2Kai Li1,2Peng Chen3Yunhe Gao2Wenquan Liang2Hongqing Xi2Xinxin Wang2Bo Wei2Hongtao Li3 ( )Lin Chen2 ( )
Medical School of Chinese PLA, Beijing 100853, China
Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of People’s Liberation Army, Lanzhou 730050, China

*These authors contributed equally to this work.

Show Author Information

Abstract

Objective

To improve the prognosis of patients with gastric cancer (GC), more effective therapeutic targets are urgently needed. Increasing evidence indicates that miRNAs are involved in the progression of various tumors, and RAS-associated protein in the brain 31 (RAB31) is upregulated and promotes the progression of multiple malignant tumors. Here, we focused on identifying RAB31-targeted miRNAs and elucidating their potential mechanism in the progression of GC.

Methods

RAB31 and miR-378a-3p expression levels were detected in paired fresh GC tissues and GC cell lines. Bioinformatics analysis was used to predict the miRNAs targeting RAB31 and the relationships between RAB31 and other genes. Dual-luciferase reporter assays were applied to verify the targeted interaction relationship. CCK-8, colony formation, flow cytometry, wound healing, and Transwell assays were performed to assess the proliferation, apoptosis, migration, and invasion of GC cells. Tumorsphere formation assays were performed to assess the stemness of gastric cancer stem cells. Related proteins were detected by Western blot. Xenograft assays in nude mice were performed to explore the effect of miR-378a-3p in vivo.

Results

We report the first evidence that miR-378a-3p is downregulated in GC, whereas its overexpression inhibits proliferation, invasion, and migration as well as promotes apoptosis in GC cells. Mechanistically, miR-378a-3p inhibits the progression of GC by directly targeting RAB31. Restoring RAB31 expression partially offsets the inhibitory effect of miR-378a-3p. Further research revealed that miR-378a-3p inhibits GLI1/2 in the Hedgehog signaling pathway and attenuates the stemness of gastric cancer stem cells. Finally, xenograft assays showed that miR-378a-3p inhibits GC tumorigenesis in vivo.

Conclusions

MiR-378a-3p inhibits GC progression by directly targeting RAB31 and inhibiting the Hedgehog signaling pathway proteins GLI1/2.

Electronic Supplementary Material

Download File(s)
cbm-19-12-1662_ESM.pdf (59.9 KB)

References

1

Addario BJ. Lung cancer is a global epidemic and requires a global effort. Ann Transl Med. 2015; 3: 26-28.

2

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. Ca Cancer J Clin. 2016; 66: 115-32.

3

Chu DT, Zhang XR, Li JL, Qu FL, Sun Y. The treatment of advanced non-small cell lung cancer (NSCLC)-overseas updated and local experiences. Gan To Kagaku Ryoho. 2002; 29: 117-24.

4

Arriagada R, Bergman B, Dunant A, Le Chevalier T, Pignon JP, Vansteenkiste J, et al. Cisplatin-based adjuvant chemotherapy in patients with completely resected non-small-cell lung cancer. N Engl J Med. 2004; 350: 351-60.

5

Mikell JL, Gillespie TW, Hall WA, Nickleach DC, Liu Y, Lipscomb J, et al. Postoperative radiotherapy is associated with better survival in non-small cell lung cancer with involved N2 lymph nodes: results of an analysis of the National Cancer Data Base. J Thorac Oncol. 2015; 10: 462-71.

6

Krause M, Gurtner K, Deuse Y, Baumann M. Heterogeneity of tumour response to combined radiotherapy and EGFR inhibitors: differences between antibodies and TK inhibitors. Int J Radiat Biol. 2009; 85: 943-54.

7

Saintigny P, Burger JA. Recent advances in non-small cell lung cancer biology and clinical management. Discov Med. 2012; 13: 287-97.

8

Amé JC, Spenlehauer C, de Murcia G. The PARP superfamily. Bioessays. 2004; 26: 882-93.

9

Caldecott KW. Single-strand break repair and genetic disease. Nat Rev Genet. 2008; 9: 619-31.

10

Schreiber V, Dantzer F, Ame JC, de Murcia G. Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol. 2006; 7: 517-28.

11

Gadducci A, Guerrieri ME. PARP inhibitors in epithelial ovarian cancer: state of art and perspectives of clinical research. Anticancer Res. 2016; 36: 2055-64.

12

Lee JM, Ledermann JA, Kohn EC. PARP inhibitors for BRCA1/2 mutation-associated and BRCA-like malignancies. Ann Oncol. 2014; 25: 32-40.

13

Yap TA, Sandhu SK, Carden CP, de Bono JS. Poly(ADP-ribose) polymerase (PARP) inhibitors: exploiting a synthetic lethal strategy in the clinic. CA Cancer J Clin. 2011; 61: 31-49.

14

Mendes-Pereira AM, Martin SA, Brough R, McCarthy A, Taylor JR, Kim JS, et al. Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO Mol Med. 2009; 1: 315-22.

15

Sourisseau T, Maniotis D, McCarthy A, Tang C, Lord CJ, Ashworth A, et al. Aurora-A expressing tumour cells are deficient for homology-directed DNA double strand-break repair and sensitive to PARP inhibition. EMBO Mol Med. 2010; 2: 130-42.

16

Williamson CT, Muzik H, Turhan AG, Zamò A, O'Connor MJ, Bebb DG, et al. ATM deficiency sensitizes mantle cell lymphoma cells to poly(ADP-Ribose) polymerase-1 inhibitors. Mol Cancer Ther. 2010; 9: 347-57.

17

Shen Y, Rehman FL, Feng Y, Boshuizen J, Bajrami I, Elliott R, et al. BMN 673, a novel and highly potent PARP1/2 inhibitor for the treatment of human cancers with DNA repair deficiency. Clin Cancer Res. 2013; 19: 5003-15.

18

de Haan R, van Werkhoven E, van den Heuvel MM, Peulen HMU, Sonke GS, Elkhuizen P, et al. Study protocols of 3 parallel phase 1 trials combining radical radiotherapy with the PARP inhibitor olaparib. BMC Cancer. 2019; 19: 901-13.

19

Pommier Y, O'Connor MJ, de Bono J. Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action. Sci Transl Med. 2016; 8: 362-68.

20

Powell C, Mikropoulos C, Kaye SB, Nutting CM, Bhide SA, Newbold K, et al. Pre-clinical and clinical evaluation of PARP inhibitors as tumour-specific radiosensitisers. Cancer Treat Rev. 2010; 36: 566-75.

21

Chu WK, Hickson ID. RecQ helicases: multifunctional genome caretaker. Nat Rev Cancer. 2009; 9: 644-54.

22

Manthei KA, Keck JL. The BLM dissolvasome in DNA replication and repair. Cell Mol Life Sci. 2013; 70: 4067-84.

23

Van Brabant AJ, Ye T, Sanz M, German Ⅲ JL, Ellis NA, Holloman WK. Binding and melting of D-loops by the Bloom syndrome helicase. Biochemistry. 2000; 39: 14617-25.

24

Wu L, Hickson ID. The Bloom's syndrome helicase suppresses crossing over during homologous recombination. Nature. 2003; 426: 870-4.

25

Cunniff C, Bassetti JA, Ellis NA. Bloom's syndrome: clinical spectrum, molecular pathogenesis, and cancer predisposition. Mol Syndromol. 2017; 8: 4-23.

26

Shuai F, Wang B, Dong S. miR-522-3p Promotes tumorigenesis in human colorectal cancer via targeting Bloom syndrome protein. Oncol Res. 2018; 26: 1113-21.

27

Nguyen GH, Dexheimer TS, Rosenthal AS, Chu WK, Singh DK, Mosedale G, et al. A small molecule inhibitor of the BLM helicase modulates chromosome stability in human cells. Chem Biol. 2013; 20: 55-62.

28

Rosenthal AS, Dexheimer TS, Gileadi O, Nguyen GH, Chu WK, Hickson ID, et al. Synthesis and SAR studies of 5-(pyridin-4-yl)-1, 3, 4-thiadiazol-2-amine derivatives as potent inhibitors of Bloom helicase. Bioorg Med Chem Lett. 2013; 23: 5660-6.

29

Yin QK, Wang CX, Wang YQ, Guo QL, Zhang ZL, Ou TM, et al. Discovery of isaindigotone derivatives as novel Bloom's syndrome protein (BLM) helicase inhibitors that disrupt the BLM/DNA interactions and regulate the homologous recombination repair. J Med Chem. 2019; 62: 3147-62.

30

Wang CX, Zhang ZL, Yin QK, Tu JL, Wang JE, Xu YH, et al. Design, synthesis, and evaluation of new quinazolinone derivatives that inhibit Bloom syndrome protein (BLM) helicase, trigger DNA damage at the telomere region, and synergize with PARP inhibitors. J Med Chem. 2020; 63: 9752-72.

31

Khanna KK, Jackson SP. DNA double-strand breaks: signaling, repair and the cancer connection. Nat Genet. 2001; 27: 247-54.

32

Modesti M, Kanaar R. DNA repair: spot(light)s on chromatin. Curr Biol. 2001; 11: R229-R32.

33

Lees-Miller SP, Meek K. Repair of DNA double strand breaks by non-homologous end joining. Biochimie. 2003; 85: 1161-73.

34

van Gent DC, Hoeijmakers JH, Kanaar R. Chromosomal stability and the DNA double-stranded break connection. Nat Rev Genet. 2001; 2: 196-06.

35

Baumann P, West SC. Role of the human RAD51 protein in homologous recombination and double-stranded-break repair. Trends Biochem Sci. 1998; 23: 247-51.

36

Uematsu N, Weterings E, Yano K, Morotomi-Yano K, Jakob B, Taucher-Scholz G, et al. Autophosphorylation of DNA-PKCS regulates its dynamics at DNA double-strand breaks. J Cell Biol. 2007; 177: 219-29.

37

Liu T, Huang J. DNA end resection: facts and mechanisms. Genom Proteom Bioinf. 2016; 14: 126-30.

38

Sleeth KM, Sørensen CS, N Issaeva, Dziegielewski J, Bartek J, Helleday T. RPA mediates recombination repair during replication stress and is displaced from DNA by checkpoint signalling in human cells. J Mol Biol. 2007; 373: 38-47.

39

Rulten SL, Grundy GJ. Non-homologous end joining: common interaction sites and exchange of multiple factors in the DNA repair process. Bioessays. 2017; 39-50.

40

Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly (ADP-ribose) polymerase. Nature. 2007; 434: 913-7.

41

Reinhardt HC, Yaffe MB. Kinases that control the cell cycle in response to DNA damage: Chk1, Chk2, and MK2. Curr Opin Cell Biol. 2009; 21: 245-55.

42

LaGory EL, Sitailo LA, Denning MF. The protein kinase Cdelta catalytic fragment is critical for maintenance of the G2/M DNA damage checkpoint. J Biol Chem. 2010; 285: 1879-87.

43

Lees JA, Buchkovich KJ, Marshak DR, Anderson CW, Harlow E. The retinoblastoma protein is phosphorylated on multiple sites by human cdc2. EMBO J. 1991; 10: 4279-90.

44

Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown JP, et al. Requirement for p53 and p21 to sustain G2 arrest after DNA damage. Science. 1998; 282: 1497-501.

45

Higuchi T, Flies DB, Marjon NA, Mantia-Smaldone G, Ronner L, Gimotty PA, et al. CTLA-4 blockade synergizes therapeutically with PARP inhibition in BRCA1-deficient ovarian cancer. Cancer Immunol Res. 2015; 3: 1257-68.

46

Tangutoori S, Baldwin P, Sridhar S. PARP inhibitors: a new era of targeted therapy. Maturitas. 2015; 81: 5-9.

47

Parsels LA, Engelke CG, Parsels J, Flanagan SA, Zhang Q, Tanska D, et al. Combinatorial efficacy of olaparib with radiation and ATR Inhibitor requires PARP1 protein in homologous recombination-proficient pancreatic cancer. Mol Cancer Ther. 2021; 20: 263-73.

48

Senra JM, Telfer BA, Cherry KE, McCrudden CM, Hirst DG, O'Connor MJ, et al. Inhibition of PARP-1 by olaparib (AZD2281) increases the radiosensitivity of a lung tumor xenograft. Mol Cancer Ther. 2011; 10: 1949-58.

49

Kötter A, Cornils K, Borgmann K, Dahm-Daphi J, Petersen C, Dikomey E, et al. Inhibition of PARP1-dependent end-joining contributes to olaparib-mediated radiosensitization in tumor cells. Mol Oncol. 2014; 8: 1616-25.

50

Wang H, Li S, Zhang H, Wang Y, Hao S, Wu X. BLM prevents instability of structure-forming DNA sequences at common fragile sites. PLOS Genet. 2018; 14: e1007816-38.

51

Datta A, Dhar S, Awate S, Brosh RM. Synthetic lethal interactions of RECQ helicases. Trends Cancer. 2021; 7: 146-61.

52

Chen X, Ali YI, Fisher CE, Arribas-Bosacoma R, Rajasekaran MB, Williams G, et al. Uncovering an allosteric mode of action for a selective inhibitor of human Bloom syndrome protein. Elife. 2021; 10: e65339-70.

53

Gottipati P, Vischioni B, Schultz N, Solomons J, Bryant HE, Djureinovic T, et al. Poly (ADP-ribose) polymerase is hyperactivated in homologous recombination-defective cells. Cancer Res. 2010; 70: 5389-98.

54

Bridges KA, Toniatti C, Buser CA, Liu H, Buchholz TA, Meyn RE. Niraparib (MK-4827), a novel poly (ADP-Ribose) polymerase inhibitor, radiosensitizes human lung and breast cancer cells. Oncotarget. 2014; 5: 5076-86.

55

Otsuki M, Seki M, Inoue E, Abe T, Narita Y, Yoshimura A, et al. Analyses of functional interaction between RECQL1, RECQL5, and BLM which physically interact with DNA topoisomerase Ⅲalpha. Biochim Biophys Acta. 2008; 1782: 75-81.

56

Otsuki M, Seki M, Inoue E, Yoshimura A, Kato G, Yamanouchi S, et al. Functional interactions between BLM and XRCC3 in the cell. J Cell Biol. 2007; 179: 53-63.

57

Patel DS, Misenko SM, Her J, Bunting SF. BLM helicase regulates DNA repair by counteracting RAD51 loading at DNA double-strand break sites. J Cell Biol. 2017; 216: 3521-34.

58

Gravel S, Chapman JR, Magill C, Jackson SP. DNA helicases Sgs1 and BLM promote DNA double-strand break resection. Genes Dev. 2008; 22: 2767-72.

59

Nimonkar AV, Ozsoy AZ, Genschel J, Modrich P, Kowalczykowski SC. Human exonuclease 1 and BLM helicase interact to resect DNA and initiate DNA repair. Proc Natl Acad Sci U S A. 2008; 105: 16906-11.

60

Wang W, Duan B, Zeng L. Effect and mechanism of radiosensitization of poly (ADP-Ribose) polymerase inhibitor in Lewis cells and xenografts. Zhongguo Fei Ai Za Zhi. 2016; 19: 16-23.

61

Hirai T, Saito S, Fujimori H, Matsushita K, Nishio T, Okayasu R, et al. Radiosensitization by PARP inhibition to proton beam irradiation in cancer cells. Biochem Biophys Res Commun. 2016; 478: 234-40.

62

Bischof O, Kim SH, Irving J, Ellis NA, Campisi J. Regulation and localization of the Bloom syndrome protein in response to DNA damage. J Cell Biol. 2001; 153: 367-80.

63

Dutertre S, Ababou M, Onclercq R, Delic J, Chatton B, Jaulin C, et al. Cell cycle regulation of the endogenous wild type Bloom's syndrome DNA helicase. Oncogene. 2000; 19: 2731-8.

64

Chapman JR, Sossick AJ, Boulton SJ, Jackson SP. BRCA1-associated exclusion of 53BP1 from DNA damage sites underlies temporal control of DNA repair. J Cell Sci. 2012; 125: 3529-34.

65

Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005; 434: 917-21.

66

Hilton JF, Hadfield MJ, Tran MT, Shapiro GI. Poly (ADP-ribose) polymerase inhibitors as cancer therapy. Front Biosci (Landmark Ed). 2013; 18: 1392-406.

67

Shaheen M, Allen C, Nickoloff JA, Hromas R. Synthetic lethality: exploiting the addiction of cancer to DNA repair. Blood. 2011; 117: 6074-82.

Cancer Biology & Medicine
Pages 1662-1682
Cite this article:
Xu X, Li Y, Liu G, et al. MiR-378a-3p acts as a tumor suppressor in gastric cancer via directly targeting RAB31 and inhibiting the Hedgehog pathway proteins GLI1/2. Cancer Biology & Medicine, 2022, 19(12): 1662-1682. https://doi.org/10.20892/j.issn.2095-3941.2022.0337

131

Views

1

Downloads

3

Crossref

8

Web of Science

7

Scopus

Altmetrics

Received: 17 June 2022
Accepted: 22 September 2022
Published: 12 December 2022
©2022 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return