Chonnam Med J.  2017 Sep;53(3):178-186. 10.4068/cmj.2017.53.3.178.

Role of Recepteur D'origine Nantais on Gastric Cancer Development and Progression

Affiliations
  • 1Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea. ydjung@chonnam.ac.kr

Abstract

Recepteur d'origine nantais (RON) is a receptor tyrosine kinase belonging to the subfamily of which c-MET is the prototype. Large epidemiologic studies have confirmed the strong association between RON and gastric cancer development. Constitutive activation of RON signaling directly correlates with tumorigenic phenotypes of gastric cancer and a poor survival rate in advanced gastric cancer patients. In this review, we focus on recent evidence of the aberrant expression and activation of RON in gastric cancer tumors and provide insights into the mechanism of RON signaling associated with gastric cancer progression and metastasis. Current therapeutics against RON in gastric cancer are summarized.

Keyword

Humans; Stomach Neoplasms; MET protein, human; Proto-Oncogene Proteins c-met; Phenotype

MeSH Terms

Epidemiologic Studies
Humans
Neoplasm Metastasis
Phenotype
Protein-Tyrosine Kinases
Proto-Oncogene Proteins c-met
Stomach Neoplasms*
Survival Rate
Protein-Tyrosine Kinases
Proto-Oncogene Proteins c-met

Figure

  • FIG. 1 Comparison of human RON and c-MET. Sema: semaphoring, PSI: plexin semaphorin-integrin, IPT: immunoglobulin-plexin-transcription factor, TM: transmembrane, TK: tyrosine kinase catalytic sites.

  • FIG. 2 Alternative splicing and various isoforms of RON. E: Exon.

  • FIG. 3 Receptor activation of RON. SOS: son of sevenless, GRB2: growth factor receptor-bound protein 2, CBL: casitas B-lineage lymphome.

  • FIG. 4 Downstream signaling of RON activation. GRB2: growth factor receptor-bound protein 2, MAPK: mitogen-activated protein kinases, ERK1/2: extracellular signal-regulated kinase 1/2, IκBα: nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha, EMT: epithelial mesenchymal transition, TGF-β: transforming growth factor beta, NF-κB: nuclear factor kappa light chain enhancer of activated B cells, JAK: janus kinase, STAT3: signal transducer and activator of transcription 3, mTOR: mamalian target of rapamycin, HIF1α: hypoxia inducible factor 1-alplha, P70S6K: ribosomal protein S6 kinase, GSK3β: Glycogen synthase kinase 3 beta.

  • FIG. 5 Intracellular signals of RON activation for the growth and invasion in gastric cancer cells. CDKI: Cycline-dependent kinase inhibitor, CDK4: Cycline-dependent kinase 4, MAPK: Mitogen-activated protein kinases, Erk1/2: extracellular signal-regulated kinase 1/2, JNK: c-Jun N-terminal kinases, AP-1: Activator protein-1, NF-kB: nuclear factor kappa-light-chain-enhancer of activated B cells, UPAR: Urokinase receptor.

  • FIG. 6 Intracellular signals of RON activation for the cell apoptosis in gastric cancer cells. XIAP: X-linked inhibitor of apoptosis protein, Bcl-2: B-cell lymphoma 2, Bax: Bcl-2-associated X protein, Bik: BCL2 Interacting Killer, Bad: Bcl-2-associated death promoter, Bid: BH3 interacting domain death agonist, BimSL: Bcl-2-like protein 11, PTEN: Phosphatase and tensin homolog.


Reference

1. Ronsin C, Muscatelli F, Mattei MG, Breathnach R. A novel putative receptor protein tyrosine kinase of the met family. Oncogene. 1993; 8:1195–1202.
2. Gaudino G, Follenzi A, Naldini L, Collesi C, Santoro M, Gallo KA, et al. RON is a heterodimeric tyrosine kinase receptor activated by the HGF homologue MSP. EMBO J. 1994; 13:3524–3532.
Article
3. Zarei O, Benvenuti S, Ustun-Alkan F, Hamzeh-Mivehroud M, Dastmalchi S. Strategies of targeting the extracellular domain of RON tyrosine kinase receptor for cancer therapy and drug delivery. J Cancer Res Clin Oncol. 2016; 142:2429–2446.
Article
4. Wang MH, Padhye SS, Guin S, Ma Q, Zhou YQ. Potential therapeutics specific to c-MET/RON receptor tyrosine kinases for molecular targeting in cancer therapy. Acta Pharmacol Sin. 2010; 31:1181–1188.
Article
5. Quantin B, Schuhbaur B, Gesnel MC, Doll'e P, Breathnach R. Restricted expression of the ron gene encoding the macrophage stimulating protein receptor during mouse development. Dev Dyn. 1995; 204:383–390.
Article
6. Del Gatto F, Gilbert E, Ronsin C, Breathnach R. Structure of the promoter for the human macrophage stimulating protein receptor gene. Biochim Biophys Acta. 1995; 1263:93–95.
Article
7. Angeloni D, Danilkovitch-Miagkova A, Ivanov SV, Breathnach R, Johnson BE, Leonard EJ, et al. Gene structure of the human receptor tyrosine kinase RON and mutation analysis in lung cancer samples. Genes Chromosomes Cancer. 2000; 29:147–156.
Article
8. Bardella C, Costa B, Maggiora P, Patane' S, Olivero M, Ranzani GN, et al. Truncated RON tyrosine kinase drives tumor cell progression and abrogates cell-cell adhesion through E-cadherin transcriptional repression. Cancer Res. 2004; 64:5154–5161.
Article
9. Angeloni D, Danilkovitch-Miagkova A, Ivanova T, Braga E, Zabarovsky E, Lerman MI. Hypermethylation of Ron proximal promoter associates with lack of full-length Ron and transcription of oncogenic short-Ron from an internal promoter. Oncogene. 2007; 26:4499–4512.
Article
10. Collesi C, Santoro MM, Gaudino G, Comoglio PM. A splicing variant of the RON transcript induces constitutive tyrosine kinase activity and an invasive phenotype. Mol Cell Biol. 1996; 16:5518–5526.
Article
11. Santoro MM, Collesi C, Grisendi S, Gaudino G, Comoglio PM. Constitutive activation of the RON gene promotes invasive growth but not transformation. Mol Cell Biol. 1996; 16:7072–7083.
Article
12. Zhou YQ, He C, Chen YQ, Wang D, Wang MH. Altered expression of the RON receptor tyrosine kinase in primary human colorectal adenocarcinomas: generation of different splicing RON variants and their oncogenic potential. Oncogene. 2003; 22:186–197.
Article
13. Liu X, Zhao L, Derose YS, Lin YC, Bieniasz M, Eyob H, et al. Short-form ron promotes spontaneous breast cancer metastasis through interaction with phosphoinositide 3-kinase. Genes Cancer. 2011; 2:753–762.
Article
14. Wang MH, Kurtz AL, Chen Y. Identification of a novel splicing product of the RON receptor tyrosine kinase in human colorectal carcinoma cells. Carcinogenesis. 2000; 21:1507–1512.
Article
15. Lu Y, Yao HP, Wang MH. Multiple variants of the RON receptor tyrosine kinase: biochemical properties, tumorigenic activities, and potential drug targets. Cancer Lett. 2007; 257:157–164.
Article
16. Zhang K, Zhou YQ, Yao HP, Wang MH. Alterations in a defined extracellular region of the RON receptor tyrosine kinase promote RON-mediated motile and invasive phenotypes in epithelial cells. Int J Oncol. 2010; 36:255–264.
Article
17. Ghigna C, De Toledo M, Bonomi S, Valacca C, Gallo S, Apicella M, et al. Pro-metastatic splicing of Ron proto-oncogene mRNA can be reversed: therapeutic potential of bifunctional oligonucleotides and indole derivatives. RNA Biol. 2010; 7:495–503.
Article
18. Yao HP, Zhou YQ, Zhang R, Wang MH. MSP-RON signalling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 2013; 13:466–481.
Article
19. Faham N, Welm AL. RON signaling is a key mediator of tumor progression in many human cancers. Cold Spring Harb Symp Quant Biol. 2016; 81:177–188.
Article
20. Shimamoto A, Kimura T, Matsumoto K, Nakamura T. Hepatocyte growth factor-like protein is identical to macrophage stimulating protein. FEBS Lett. 1993; 333:61–66.
Article
21. Wang MH, Ronsin C, Gesnel MC, Coupey L, Skeel A, Leonard EJ, et al. Identification of the ron gene product as the receptor for the human macrophage stimulating protein. Science. 1994; 266:117–119.
Article
22. Danilkovitch A, Miller M, Leonard EJ. Interaction of macrophage-stimulating protein with its receptor. Residues critical for beta chain binding and evidence for independent alpha chain binding. J Biol Chem. 1999; 274:29937–29943.
23. Ponzetto C, Bardelli A, Zhen Z, Maina F, dalla Zonca P, Giordano S, et al. A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell. 1994; 77:261–271.
Article
24. Xiao ZQ, Chen YQ, Wang MH. Requirement of both tyrosine residues 1330 and 1337 in the C-terminal tail of the RON receptor tyrosine kinase for epithelial cell scattering and migration. Biochem Biophys Res Commun. 2000; 267:669–675.
Article
25. Iwama A, Yamaguchi N, Suda T. STK/RON receptor tyrosine kinase mediates both apoptotic and growth signals via the multifunctional docking site conserved among the HGF receptor family. EMBO J. 1996; 15:5866–5875.
Article
26. Santoro MM, Penengo L, Minetto M, Orecchia S, Cilli M, Gaudino G. Point mutations in the tyrosine kinase domain release the oncogenic and metastatic potential of the Ron receptor. Oncogene. 1998; 17:741–749.
Article
27. McClaine RJ, Marshall AM, Wagh PK, Waltz SE. Ron receptor tyrosine kinase activation confers resistance to tamoxifen in breast cancer cell lines. Neoplasia. 2010; 12:650–658.
Article
28. Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, et al. Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res. 2010; 70:1130–1140.
Article
29. Takeuchi K, Ito F. Receptor tyrosine kinases and targeted cancer therapeutics. Biol Pharm Bull. 2011; 34:1774–1780.
Article
30. O'Toole JM, Rabenau KE, Burns K, Lu D, Mangalampalli V, Balderes P, et al. Therapeutic implications of a human neutralizing antibody to the macrophage-stimulating protein receptor tyrosine kinase (RON), a c-MET family member. Cancer Res. 2006; 66:9162–9170.
31. Zhou D, Pan G, Zheng C, Zheng J, Yian L, Teng X. Expression of the RON receptor tyrosine kinase and its association with gastric carcinoma versus normal gastric tissues. BMC Cancer. 2008; 8:353.
Article
32. Catenacci DV, Cervantes G, Yala S, Nelson EA, El-Hashani E, Kanteti R, et al. RON (MST1R) is a novel prognostic marker and therapeutic target for gastroesophageal adenocarcinoma. Cancer Biol Ther. 2011; 12:9–46.
Article
33. Song YA, Park YL, Kim KY, Myung E, Chung CY, Cho SB, et al. RON is associated with tumor progression via the inhibition of apoptosis and cell cycle arrest in human gastric cancer. Pathol Int. 2012; 62:127–136.
Article
34. Lee J, Kang WK, Park JO, Park SH, Park YS, Lim HY, et al. Expression of activated signal transducer and activator of transcription 3 predicts poor clinical outcome in gastric adenocarcinoma. APMIS. 2009; 117:598–606.
Article
35. Kim DY, Cha ST, Ahn DH, Kang HY, Kwon CI, Ko KH, et al. STAT3 expression in gastric cancer indicates a poor prognosis. J Gastroenterol Hepatol. 2009; 24:646–651.
Article
36. Deng JY, Sun D, Liu XY, Pan Y, Liang H. STAT-3 correlates with lymph node metastasis and cell survival in gastric cancer. World J Gastroenterol. 2010; 16:5380–5387.
Article
37. Lee KE, Park JS, Khoi PN, Joo YE, Lee YH, Jung YD. Upregulation of recepteur d'origine nantais tyrosine kinase and cell invasiveness via early growth response-1 in gastric cancer cells. J Cell Biochem. 2012; 113:1217–1223.
Article
38. Park JS, Park JH, Khoi PN, Joo YE, Jung YD. MSP-induced RON activation upregulates uPAR expression and cell invasiveness via MAPK, AP-1 and NF-κB signals in gastric cancer cells. Carcinogenesis. 2011; 32:175–181.
Article
39. Park JS, Park JH, Lee S, Joo YE, Jung YD. Small interfering RNA targeting of Recepteur d'Origine Nantais induces apoptosis via modulation of nuclear factor-kappaB and Bcl-2 family in gastric cancer cells. Oncol Rep. 2010; 24:709–714.
40. Kang CM, Babicky ML, Lowy AM. The RON receptor tyrosine kinase in pancreatic cancer pathogenesis and its potential implications for future targeted therapies. Pancreas. 2014; 43:183–189.
Article
41. Kretschmann KL, Eyob H, Buys SS, Welm AL. The macrophage stimulating protein/Ron pathway as a potential therapeutic target to impede multiple mechanisms involved in breast cancer progression. Curr Drug Targets. 2010; 11:1157–1168.
Article
42. Zhao S, Cao L, Freeman JW. Knockdown of RON receptor kinase delays but does not prevent tumor progression while enhancing HGF/MET signaling in pancreatic cancer cell lines. Oncogenesis. 2013; 2:e76.
Article
43. Kataoka Y, Mukohara T, Tomioka H, Funakoshi Y, Kiyota N, Fujiwara Y, et al. Foretinib (GSK1363089), a multi-kinase inhibitor of MET and VEGFRs, inhibits growth of gastric cancer cell lines by blocking inter-receptor tyrosine kinase networks. Invest New Drugs. 2012; 30:1352–1360.
Article
44. Shah MA, Wainberg ZA, Catenacci DV, Hochster HS, Ford J, Kunz P, et al. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One. 2013; 8:e54014.
Article
45. Awazu Y, Nakamura K, Mizutani A, Kakoi Y, Iwata H, Yamasaki S, et al. A novel inhibitor of c-Met and VEGF receptor tyrosine kinases with a broad spectrum of in vivo antitumor activities. Mol Cancer Ther. 2013; 12:913–924.
Article
46. Toiyama Y, Yasuda H, Saigusa S, Matushita K, Fujikawa H, Tanaka K, et al. Co-expression of hepatocyte growth factor and c-Met predicts peritoneal dissemination established by autocrine hepatocyte growth factor/c-Met signaling in gastric cancer. Int J Cancer. 2012; 130:2912–2921.
Article
47. Yashiro M, Nishii T, Hasegawa T, Matsuzaki T, Morisaki T, Fukuoka T, et al. A c-Met inhibitor increases the chemosensitivity of cancer stem cells to the irinotecan in gastric carcinoma. Br J Cancer. 2013; 109:2619–2628.
Article
48. Isaacs JS, Xu W, Neckers L. Heat shock protein 90 as a molecular target for cancer therapeutics. Cancer Cell. 2003; 3:213–217.
Article
49. Neckers L. Hsp90 inhibitors as novel cancer chemotherapeutic agents. Trends Mol Med. 2002; 8:4 Suppl. S55–S61.
Article
50. Zhang H, Burrows F. Targeting multiple signal transduction pathways through inhibition of Hsp90. J Mol Med (Berl). 2004; 82:488–499.
Article
51. Germano S, Barberis D, Santoro MM, Penengo L, Citri A, Yarden Y, et al. Geldanamycins trigger a novel Ron degradative pathway, hampering oncogenic signaling. J Biol Chem. 2006; 281:21710–21719.
Article
52. Moser C, Lang SA, Hackl C, Zhang H, Lundgren K, Hong V, et al. Oncogenic MST1R activity in pancreatic and gastric cancer represents a valid target of HSP90 inhibitors. Anticancer Res. 2012; 32:427–437.
53. Jung YD, Kim MS, Shin BA, Chay KO, Ahn BW, Liu W, et al. EGCG, a major component of green tea, inhibits tumour growth by inhibiting VEGF induction in human colon carcinoma cells. Br J Cancer. 2001; 84:844–850.
Article
54. Park JS, Khoi PN, Joo YE, Lee YH, Lang SA, Stoeltzing O, et al. EGCG inhibits recepteur d'origine nantais expression by suppressing Egr-1 in gastric cancer cells. Int J Oncol. 2013; 42:1120–1126.
Article
55. Xia Y, Lian S, Khoi PN, Yoon HJ, Han JY, Chay KO, et al. Chrysin inhibits cell invasion by inhibition of Recepteur d'origine Nantais via suppressing early growth response-1 and NF-κB transcription factor activities in gastric cancer cells. Int J Oncol. 2015; 46:1835–1843.
Article
56. Lian S, Park JS, Xia Y, Nguyen TT, Joo YE, Kim KK, et al. Micro-RNA-375 functions as a tumor-suppressor gene in gastric cancer by targeting recepteur d'Origine Nantais. Int J Mol Sci. 2016; 17:E1633.
Article
Full Text Links
  • CMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr