The emerging role of RUNX3 in cancer metastasis (Review)
- Authors:
- Feifei Chen
- Xin Liu
- Jin Bai
- Dongsheng Pei
- Junnian Zheng
-
Affiliations: Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China - Published online on: December 24, 2015 https://doi.org/10.3892/or.2015.4515
- Pages: 1227-1236
This article is mentioned in:
Abstract
Gupta GP and Massagué J: Cancer metastasis: Building a framework. Cell. 127:679–695. 2006. View Article : Google Scholar : PubMed/NCBI | |
Steeg PS: Tumor metastasis: Mechanistic insights and clinical challenges. Nat Med. 12:895–904. 2006. View Article : Google Scholar : PubMed/NCBI | |
Al-Mehdi AB, Tozawa K, Fisher AB, Shientag L, Lee A and Muschel RJ: Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: A new model for metastasis. Nat Med. 6:100–102. 2000. View Article : Google Scholar | |
Fidler IJ: The pathogenesis of cancer metastasis: The 'seed and soil' hypothesis revisited. Nat Rev Cancer. 3:453–458. 2003. View Article : Google Scholar : PubMed/NCBI | |
Fukushima-Nakase Y, Naoe Y, Taniuchi I, Hosoi H, Sugimoto T and Okuda T: Shared and distinct roles mediated through C-terminal subdomains of acute myeloid leukemia/Runt-related transcription factor molecules in murine development. Blood. 105:4298–4307. 2005. View Article : Google Scholar : PubMed/NCBI | |
Wotton S, Terry A, Kilbey A, Jenkins A, Herzyk P, Cameron E and Neil JC: Gene array analysis reveals a common Runx transcriptional programme controlling cell adhesion and survival. Oncogene. 27:5856–5866. 2008. View Article : Google Scholar : PubMed/NCBI | |
Brady G and Farrell PJ: RUNX3-mediated repression of RUNX1 in B cells. J Cell Physiol. 221:283–287. 2009. View Article : Google Scholar : PubMed/NCBI | |
Fukamachi H and Ito K: Growth regulation of gastric epithelial cells by Runx3. Oncogene. 23:4330–4335. 2004. View Article : Google Scholar : PubMed/NCBI | |
Levanon D, Bettoun D, Harris-Cerruti C, Woolf E, Negreanu V, Eilam R, Bernstein Y, Goldenberg D, Xiao C, Fliegauf M, et al: The Runx3 transcription factor regulates development and survival of TrkC dorsal root ganglia neurons. EMBO J. 21:3454–3463. 2002. View Article : Google Scholar : PubMed/NCBI | |
Woolf E, Brenner O, Goldenberg D, Levanon D and Groner Y: Runx3 regulates dendritic epidermal T cell development. Dev Biol. 303:703–714. 2007. View Article : Google Scholar : PubMed/NCBI | |
Bagchi A and Mills AA: The quest for the 1p36 tumor suppressor. Cancer Res. 68:2551–2556. 2008. View Article : Google Scholar : PubMed/NCBI | |
Subramaniam MM, Chan JY, Yeoh KG, Quek T, Ito K and Salto-Tellez M: Molecular pathology of RUNX3 in human carcinogenesis. Biochim Biophys Acta. 1796:315–331. 2009.PubMed/NCBI | |
Oshimo Y, Oue N, Mitani Y, Nakayama H, Kitadai Y, Yoshida K, Ito Y, Chayama K and Yasui W: Frequent loss of RUNX3 expression by promoter hypermethylation in gastric carcinoma. Pathobiology. 71:137–143. 2004. View Article : Google Scholar : PubMed/NCBI | |
Lau QC, Raja E, Salto-Tellez M, Liu Q, Ito K, Inoue M, Putti TC, Loh M, Ko TK, Huang C, et al: RUNX3 is frequently inactivated by dual mechanisms of protein mislocalization and promoter hypermethylation in breast cancer. Cancer Res. 66:6512–6520. 2006. View Article : Google Scholar : PubMed/NCBI | |
Ahlquist T, Lind GE, Costa VL, Meling GI, Vatn M, Hoff GS, Rognum TO, Skotheim RI, Thiis-Evensen E and Lothe RA: Gene methylation profiles of normal mucosa, and benign and malignant colorectal tumors identify early onset markers. Mol Cancer. 7:942008. View Article : Google Scholar | |
Ito K, Liu Q, Salto-Tellez M, Yano T, Tada K, Ida H, Huang C, Shah N, Inoue M, Rajnakova A, et al: RUNX3, a novel tumor suppressor, is frequently inactivated in gastric cancer by protein mislocalization. Cancer Res. 65:7743–7750. 2005.PubMed/NCBI | |
Voon DC, Wang H, Koo JK, Nguyen TA, Hor YT, Chu YS, Ito K, Fukamachi H, Chan SL, Thiery JP, et al: Runx3 protects gastric epithelial cells against epithelial-mesenchymal transition-induced cellular plasticity and tumorigenicity. Stem Cells. 30:2088–2099. 2012. View Article : Google Scholar : PubMed/NCBI | |
Chen F, Bai J, Li W, Mei P, Liu H, Li L, Pan Z, Wu Y and Zheng J: RUNX3 suppresses migration, invasion and angiogenesis of human renal cell carcinoma. PLoS One. 8:e562412013. View Article : Google Scholar : PubMed/NCBI | |
Sakakura C, Hasegawa K, Miyagawa K, Nakashima S, Yoshikawa T, Kin S, Nakase Y, Yazumi S, Yamagishi H, Okanoue T, et al: Possible involvement of RUNX3 silencing in the peritoneal metastases of gastric cancers. Clin Cancer Res. 11:6479–6488. 2005. View Article : Google Scholar : PubMed/NCBI | |
Yano T, Ito K, Fukamachi H, Chi XZ, Wee HJ, Inoue K, Ida H, Bouillet P, Strasser A, Bae SC, et al: The RUNX3 tumor suppressor upregulates Bim in gastric epithelial cells undergoing transforming growth factor beta-induced apoptosis. Mol Cell Biol. 26:4474–4488. 2006. View Article : Google Scholar : PubMed/NCBI | |
Peng Z, Wei D, Wang L, Tang H, Zhang J, Le X, Jia Z, Li Q and Xie K: RUNX3 inhibits the expression of vascular endothelial growth factor and reduces the angiogenesis, growth, and metastasis of human gastric cancer. Clin Cancer Res. 12:6386–6394. 2006. View Article : Google Scholar : PubMed/NCBI | |
Mei PJ, Bai J, Liu H, Li C, Wu YP, Yu ZQ and Zheng JN: RUNX3 expression is lost in glioma and its restoration causes drastic suppression of tumor invasion and migration. J Cancer Res Clin Oncol. 137:1823–1830. 2011. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Chen G, Cheng Y, Martinka M and Li G: Prognostic significance of RUNX3 expression in human melanoma. Cancer. 117:2719–2727. 2011. View Article : Google Scholar : PubMed/NCBI | |
Chen F, Wang M, Bai J, Liu Q, Xi Y, Li W and Zheng J: Role of RUNX3 in suppressing metastasis and angiogenesis of human prostate cancer. PLoS One. 9:e869172014. View Article : Google Scholar : PubMed/NCBI | |
Chen W, Salto-Tellez M, Palanisamy N, Ganesan K, Hou Q, Tan LK, Sii LH, Ito K, Tan B, Wu J, et al: Targets of genome copy number reduction in primary breast cancers identified by integrative genomics. Genes Chromosomes Cancer. 46:288–301. 2007. View Article : Google Scholar | |
Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, Chen H, Omeroglu G, Meterissian S, Omeroglu A, et al: Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 14:518–527. 2008. View Article : Google Scholar : PubMed/NCBI | |
Sakakura C, Miyagawa K, Fukuda KI, Nakashima S, Yoshikawa T, Kin S, Nakase Y, Ida H, Yazumi S, Yamagishi H, et al: Frequent silencing of RUNX3 in esophageal squamous cell carcinomas is associated with radioresistance and poor prognosis. Oncogene. 26:5927–5938. 2007. View Article : Google Scholar : PubMed/NCBI | |
Chuang LS and Ito Y: RUNX3 is multifunctional in carcinogenesis of multiple solid tumors. Oncogene. 29:2605–2615. 2010. View Article : Google Scholar : PubMed/NCBI | |
Yu YY, Chen C, Kong FF and Zhang W: Clinicopathological significance and potential drug target of RUNX3 in breast cancer. Drug Des Devel Ther. 8:2423–2430. 2014.PubMed/NCBI | |
Wang D, Cui W, Wu X, Qu Y, Wang N, Shi B and Hou P: RUNX3 site-specific hypermethylation predicts papillary thyroid cancer recurrence. Am J Cancer Res. 4:725–737. 2014.PubMed/NCBI | |
Katayama Y, Takahashi M and Kuwayama H: Helicobacter pylori causes runx3 gene methylation and its loss of expression in gastric epithelial cells, which is mediated by nitric oxide produced by macrophages. Biochem Biophys Res Commun. 388:496–500. 2009. View Article : Google Scholar : PubMed/NCBI | |
Cheng AS, Culhane AC, Chan MW, Venkataramu CR, Ehrich M, Nasir A, Rodriguez BA, Liu J, Yan PS, Quackenbush J, et al: Epithelial progeny of estrogen-exposed breast progenitor cells display a cancer-like methylome. Cancer Res. 68:1786–1796. 2008. View Article : Google Scholar : PubMed/NCBI | |
Fujii S, Ito K, Ito Y and Ochiai A: Enhancer of zeste homologue 2 (EZH2) down-regulates RUNX3 by increasing histone H3 methylation. J Biol Chem. 283:17324–17332. 2008. View Article : Google Scholar : PubMed/NCBI | |
Lee SH, Kim J, Kim WH and Lee YM: Hypoxic silencing of tumor suppressor RUNX3 by histone modification in gastric cancer cells. Oncogene. 28:184–194. 2009. View Article : Google Scholar | |
Varambally S, Cao Q, Mani RS, Shankar S, Wang X, Ateeq B, Laxman B, Cao X, Jing X, Ramnarayanan K, et al: Genomic loss of microRNA-101 leads to overexpression of histone methyl-transferase EZH2 in cancer. Science. 322:1695–1699. 2008. View Article : Google Scholar : PubMed/NCBI | |
Lai KW, Koh KX, Loh M, Tada K, Subramaniam MM, Lim XY, Vaithilingam A, Salto-Tellez M, Iacopetta B, Ito Y, et al Singapore Gastric Cancer Consortium: MicroRNA-130b regulates the tumour suppressor RUNX3 in gastric cancer. Eur J Cancer. 46:1456–1463. 2010. View Article : Google Scholar : PubMed/NCBI | |
Kitago M, Martinez SR, Nakamura T, Sim MS and Hoon DS: Regulation of RUNX3 tumor suppressor gene expression in cutaneous melanoma. Clin Cancer Res. 15:2988–2994. 2009. View Article : Google Scholar : PubMed/NCBI | |
Goh YM, Cinghu S, Hong ET, Lee YS, Kim JH, Jang JW, Li YH, Chi XZ, Lee KS, Wee H, et al: Src kinase phosphorylates RUNX3 at tyrosine residues and localizes the protein in the cytoplasm. J Biol Chem. 285:10122–10129. 2010. View Article : Google Scholar : PubMed/NCBI | |
Kim HR, Oh BC, Choi JK and Bae SC: Pim-1 kinase phosphorylates and stabilizes RUNX3 and alters its subcellular localization. J Cell Biochem. 105:1048–1058. 2008. View Article : Google Scholar : PubMed/NCBI | |
Kim JH, Choi JK, Cinghu S, Jang JW, Lee YS, Li YH, Goh YM, Chi XZ, Lee KS, Wee H, et al: Jab1/CSN5 induces the cytoplasmic localization and degradation of RUNX3. J Cell Biochem. 107:557–565. 2009. View Article : Google Scholar : PubMed/NCBI | |
Sheen YY, Kim MJ, Park SA, Park SY and Nam JS: Targeting the transforming growth factor-β signaling in cancer therapy. Biomol Ther. 21:323–331. 2013. View Article : Google Scholar | |
Ito Y and Miyazono K: RUNX transcription factors as key targets of TGF-beta superfamily signaling. Curr Opin Genet Dev. 13:43–47. 2003. View Article : Google Scholar : PubMed/NCBI | |
Chi XZ, Yang JO, Lee KY, Ito K, Sakakura C, Li QL, Kim HR, Cha EJ, Lee YH, Kaneda A, et al: RUNX3 suppresses gastric epithelial cell growth by inducing p21WAF1/Cip1 expression in cooperation with transforming growth factor β-activated SMAD. Mol Cell Biol. 25:8097–8107. 2005. View Article : Google Scholar : PubMed/NCBI | |
Yamamura Y, Lee WL, Inoue K, Ida H and Ito Y: RUNX3 cooperates with FoxO3a to induce apoptosis in gastric cancer cells. J Biol Chem. 281:5267–5276. 2006. View Article : Google Scholar | |
Yao H, Ashihara E and Maekawa T: Targeting the Wnt/β-catenin signaling pathway in human cancers. Expert Opin Ther Targets. 15:873–887. 2011. View Article : Google Scholar : PubMed/NCBI | |
Tanaka S, Shiraha H, Nakanishi Y, Nishina S, Matsubara M, Horiguchi S, Takaoka N, Iwamuro M, Kataoka J, Kuwaki K, et al: Runt-related transcription factor 3 reverses epithelial-mesenchymal transition in hepatocellular carcinoma. Int J Cancer. 131:2537–2546. 2012. View Article : Google Scholar : PubMed/NCBI | |
Ito K, Lim AC, Salto-Tellez M, Motoda L, Osato M, Chuang LS, Lee CW, Voon DC, Koo JK, Wang H, et al: RUNX3 attenuates beta-catenin/T cell factors in intestinal tumorigenesis. Cancer Cell. 14:226–237. 2008. View Article : Google Scholar : PubMed/NCBI | |
Ju X, Ishikawa TO, Naka K, Ito K, Ito Y and Oshima M: Context-dependent activation of Wnt signaling by tumor suppressor RUNX3 in gastric cancer cells. Cancer Sci. 105:418–424. 2014. View Article : Google Scholar : PubMed/NCBI | |
Palmero I, Pantoja C and Serrano M: p19ARF links the tumour suppressor p53 to Ras. Nature. 395:125–126. 1998. View Article : Google Scholar : PubMed/NCBI | |
Lee KS, Lee YS, Lee JM, Ito K, Cinghu S, Kim JH, Jang JW, Li YH, Goh YM, Chi XZ, et al: Runx3 is required for the differentiation of lung epithelial cells and suppression of lung cancer. Oncogene. 29:3349–3361. 2010. View Article : Google Scholar : PubMed/NCBI | |
Kruse JP and Gu W: Modes of p53 regulation. Cell. 137:609–622. 2009. View Article : Google Scholar : PubMed/NCBI | |
Chi XZ, Kim J, Lee YH, Lee JW, Lee KS, Wee H, Kim WJ, Park WY, Oh BC, Stein GS, et al: Runt-related transcription factor RUNX3 is a target of MDM2-mediated ubiquitination. Cancer Res. 69:8111–8119. 2009. View Article : Google Scholar : PubMed/NCBI | |
Efeyan A and Serrano M: p53: Guardian of the genome and policeman of the oncogenes. Cell Cycle. 6:1006–1010. 2007. View Article : Google Scholar : PubMed/NCBI | |
Lee YS, Lee JW, Jang JW, Chi XZ, Kim JH, Li YH, Kim MK, Kim DM, Choi BS, Kim EG, et al: Runx3 inactivation is a crucial early event in the development of lung adenocarcinoma. Cancer Cell. 24:603–616. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hnilicová J, Hozeifi S, Stejskalová E, Dušková E, Poser I, Humpolíčková J, Hof M and Staněk D: The C-terminal domain of Brd2 is important for chromatin interaction and regulation of transcription and alternative splicing. Mol Biol Cell. 24:3557–3568. 2013. View Article : Google Scholar : PubMed/NCBI | |
Jin YH, Jeon EJ, Li QL, Lee YH, Choi JK, Kim WJ, Lee KY and Bae SC: Transforming growth factor-beta stimulates p300-dependent RUNX3 acetylation, which inhibits ubiquitination-mediated degradation. J Biol Chem. 279:29409–29417. 2004. View Article : Google Scholar : PubMed/NCBI | |
Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI | |
Padua D and Massagué J: Roles of TGFbeta in metastasis. Cell Res. 19:89–102. 2009. View Article : Google Scholar | |
Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI | |
Lee JM, Shin JO, Cho KW, Hosoya A, Cho SW, Lee YS, Ryoo HM, Bae SC and Jung HS: Runx3 is a crucial regulator of alveolar differentiation and lung tumorigenesis in mice. Differentiation. 81:261–268. 2011. View Article : Google Scholar : PubMed/NCBI | |
Chang CJ, Chao CH, Xia W, Yang JY, Xiong Y, Li CW, Yu WH, Rehman SK, Hsu JL, Lee HH, et al: p53 regulates epithelial-mesenchymal transition and stem cell properties through modulating miRNAs. Nat Cell Biol. 13:317–323. 2011. View Article : Google Scholar : PubMed/NCBI | |
Kumarswamy R, Mudduluru G, Ceppi P, Muppala S, Kozlowski M, Niklinski J, Papotti M and Allgayer H: MicroRNA-30a inhibits epithelial-to-mesenchymal transition by targeting Snai1 and is downregulated in non-small cell lung cancer. Int J Cancer. 130:2044–2053. 2012. View Article : Google Scholar | |
Yamasaki T, Seki N, Yamada Y, Yoshino H, Hidaka H, Chiyomaru T, Nohata N, Kinoshita T, Nakagawa M and Enokida H: Tumor suppressive microRNA-138 contributes to cell migration and invasion through its targeting of vimentin in renal cell carcinoma. Int J Oncol. 41:805–817. 2012.PubMed/NCBI | |
Cheng CW, Wang HW, Chang CW, Chu HW, Chen CY, Yu JC, Chao JI, Liu HF, Ding SL and Shen CY: MicroRNA-30a inhibits cell migration and invasion by downregulating vimentin expression and is a potential prognostic marker in breast cancer. Breast Cancer Res Treat. 134:1081–1093. 2012. View Article : Google Scholar : PubMed/NCBI | |
Liu Z, Chen L, Zhang X, Xu X, Xing H, Zhang Y, Li W, Yu H, Zeng J and Jia J: RUNX3 regulates vimentin expression via miR-30a during epithelial-mesenchymal transition in gastric cancer cells. J Cell Mol Med. 18:610–623. 2014. View Article : Google Scholar : PubMed/NCBI | |
Folkman J: Tumor angiogenesis: Therapeutic implications. N Engl J Med. 285:1182–1186. 1971. View Article : Google Scholar : PubMed/NCBI | |
Nishida N, Yano H, Nishida T, Kamura T and Kojiro M: Angiogenesis in cancer. Vasc Health Risk Manag. 2:213–219. 2006. View Article : Google Scholar | |
Hanahan D and Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 86:353–364. 1996. View Article : Google Scholar : PubMed/NCBI | |
Jain RK: Antiangiogenesis strategies revisited: From starving tumors to alleviating hypoxia. Cancer Cell. 26:605–622. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yang M, Kuang X, Pan Y, Tan M, Lu B, Lu J, Cheng Q and Li J: Clinicopathological characteristics of vascular endothelial growth factor expression in uveal melanoma: A meta-analysis. Mol Clin Oncol. 2:363–368. 2014.PubMed/NCBI | |
Lee JM, Lee DJ, Bae SC and Jung HS: Abnormal liver differentiation and excessive angiogenesis in mice lacking Runx3. Histochem Cell Biol. 139:751–758. 2013. View Article : Google Scholar : PubMed/NCBI | |
Lee JM, Kwon HJ, Lai WF and Jung HS: Requirement of Runx3 in pulmonary vasculogenesis. Cell Tissue Res. 356:445–449. 2014. View Article : Google Scholar : PubMed/NCBI | |
Semenza GL: Angiogenesis in ischemic and neoplastic disorders. Annu Rev Med. 54:17–28. 2003. View Article : Google Scholar | |
Harris AL: Hypoxia - a key regulatory factor in tumour growth. Nat Rev Cancer. 2:38–47. 2002. View Article : Google Scholar : PubMed/NCBI | |
Lee SH, Bae SC, Kim KW and Lee YM: RUNX3 inhibits hypoxia-inducible factor-1α protein stability by interacting with prolyl hydroxylases in gastric cancer cells. Oncogene. 33:1458–1467. 2014. View Article : Google Scholar | |
Meng S, Cao J, Zhang X, Fan Y, Fang L, Wang C, Lv Z, Fu D and Li Y: Downregulation of microRNA-130a contributes to endothelial progenitor cell dysfunction in diabetic patients via its target Runx3. PLoS One. 8:e686112013. View Article : Google Scholar : PubMed/NCBI | |
Luzzi KJ, MacDonald IC, Schmidt EE, Kerkvliet N, Morris VL, Chambers AF and Groom AC: Multistep nature of metastatic inefficiency: Dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 153:865–873. 1998. View Article : Google Scholar : PubMed/NCBI | |
Nagasaka A, Kawane K, Yoshida H and Nagata S: Apaf-1-independent programmed cell death in mouse development. Cell Death Differ. 17:931–941. 2010. View Article : Google Scholar | |
Burgess DJ: Apoptosis: Refined and lethal. Nat Rev Cancer. 13:792013. View Article : Google Scholar : PubMed/NCBI | |
Nakanishi Y, Shiraha H, Nishina S, Tanaka S, Matsubara M, Horiguchi S, Iwamuro M, Takaoka N, Uemura M, Kuwaki K, et al: Loss of runt-related transcription factor 3 expression leads hepatocellular carcinoma cells to escape apoptosis. BMC Cancer. 11:32011. View Article : Google Scholar : PubMed/NCBI | |
Zhai FX, Liu XF, Fan RF, Long ZJ, Fang ZG, Lu Y, Zheng YJ and Lin DJ: RUNX3 is involved in caspase-3-dependent apoptosis induced by a combination of 5-aza-CdR and TSA in leukaemia cell lines. J Cancer Res Clin Oncol. 138:439–449. 2012. View Article : Google Scholar | |
Liu Z, Zhang X, Xu X, Chen L, Li W, Yu H, Sun Y, Zeng J and Jia J: RUNX3 inhibits survivin expression and induces cell apoptosis in gastric cancer. Eur J Cell Biol. 93:118–126. 2014. View Article : Google Scholar : PubMed/NCBI | |
Meng X, Franklin DA, Dong J and Zhang Y: MDM2-p53 pathway in hepatocellular carcinoma. Cancer Res. 74:7161–7167. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yamada C, Ozaki T, Ando K, Suenaga Y, Inoue K, Ito Y, Okoshi R, Kageyama H, Kimura H, Miyazaki M, et al: RUNX3 modulates DNA damage-mediated phosphorylation of tumor suppressor p53 at Ser-15 and acts as a co-activator for p53. J Biol Chem. 285:16693–16703. 2010. View Article : Google Scholar : PubMed/NCBI | |
Lai CB and Mager DL: Role of runt-related transcription factor 3 (RUNX3) in transcription regulation of natural cytotoxicity receptor 1 (NCR1/NKp46), an activating natural killer (NK) cell receptor. J Biol Chem. 287:7324–7334. 2012. View Article : Google Scholar : PubMed/NCBI | |
Levanon D, Negreanu V, Lotem J, Bone KR, Brenner O, Leshkowitz D and Groner Y: Transcription factor Runx3 regulates interleukin-15-dependent natural killer cell activation. Mol Cell Biol. 34:1158–1169. 2014. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Lu Q and Cai X: MicroRNA-106a induces multidrug resistance in gastric cancer by targeting RUNX3. FEBS Lett. 587:3069–3075. 2013. View Article : Google Scholar : PubMed/NCBI | |
Li Q, Wang JX, He YQ, Feng C, Zhang XJ, Sheng JQ and Li PF: MicroRNA-185 regulates chemotherapeutic sensitivity in gastric cancer by targeting apoptosis repressor with caspase recruitment domain. Cell Death Dis. 5:e11972014. View Article : Google Scholar : PubMed/NCBI | |
Boya P, Reggiori F and Codogno P: Emerging regulation and functions of autophagy. Nat Cell Biol. 15:713–720. 2013. View Article : Google Scholar : PubMed/NCBI | |
Kenific CM, Thorburn A and Debnath J: Autophagy and metastasis: Another double-edged sword. Curr Opin Cell Biol. 22:241–245. 2010. View Article : Google Scholar : | |
Xu Q, Meng S, Liu B, Li MQ, Li Y, Fang L and Li YG: MicroRNA-130a regulates autophagy of endothelial progenitor cells through Runx3. Clin Exp Pharmacol Physiol. 41:351–357. 2014. View Article : Google Scholar : PubMed/NCBI | |
Denhardt DT, Lopez CA, Rollo EE, Hwang SM, An XR and Walther SE: Osteopontin-induced modifications of cellular functions. Ann NY Acad Sci. 760:127–142. 1995. View Article : Google Scholar : PubMed/NCBI | |
Anborgh PH, Mutrie JC, Tuck AB and Chambers AF: Role of the metastasis-promoting protein osteopontin in the tumour microenvironment. J Cell Mol Med. 14:2037–2044. 2010. View Article : Google Scholar : PubMed/NCBI | |
Fong YC, Liu SC, Huang CY, Li TM, Hsu SF, Kao ST, Tsai FJ, Chen WC, Chen CY and Tang CH: Osteopontin increases lung cancer cells migration via activation of the alphavbeta3 integrin/FAK/Akt and NF-kappaB-dependent pathway. Lung Cancer. 64:263–270. 2009. View Article : Google Scholar | |
Wang Y, Yan W, Lu X, Qian C, Zhang J, Li P, Shi L, Zhao P, Fu Z, Pu P, et al: Overexpression of osteopontin induces angiogenesis of endothelial progenitor cells via the avβ3/PI3K/AKT/eNOS/NO signaling pathway in glioma cells. Eur J Cell Biol. 90:642–648. 2011. View Article : Google Scholar : PubMed/NCBI | |
Singhal H, Bautista DS, Tonkin KS, O'Malley FP, Tuck AB, Chambers AF and Harris JF: Elevated plasma osteopontin in metastatic breast cancer associated with increased tumor burden and decreased survival. Clin Cancer Res. 3:605–611. 1997.PubMed/NCBI | |
Schneider S, Yochim J, Brabender J, Uchida K, Danenberg KD, Metzger R, Schneider PM, Salonga D, Hölscher AH and Danenberg PV: Osteopontin but not osteonectin messenger RNA expression is a prognostic marker in curatively resected non-small cell lung cancer. Clin Cancer Res. 10:1588–1596. 2004. View Article : Google Scholar : PubMed/NCBI | |
Ue T, Yokozaki H, Kitadai Y, Yamamoto S, Yasui W, Ishikawa T and Tahara E: Co-expression of osteopontin and CD44v9 in gastric cancer. Int J Cancer. 79:127–132. 1998. View Article : Google Scholar : PubMed/NCBI | |
Cheng HC, Liu YP, Shan YS, Huang CY, Lin FC, Lin LC, Lee L, Tsai CH, Hsiao M and Lu PJ: Loss of RUNX3 increases osteopontin expression and promotes cell migration in gastric cancer. Carcinogenesis. 34:2452–2459. 2013. View Article : Google Scholar : PubMed/NCBI | |
Min KW, Kim DH, Do SI, Kim K, Lee HJ, Chae SW, Sohn JH, Pyo JS, Oh YH, Kim WS, et al: Expression patterns of stromal MMP-2 and tumoural MMP-2 and -9 are significant prognostic factors in invasive ductal carcinoma of the breast. APMIS. 122:1196–1206. 2014. View Article : Google Scholar : PubMed/NCBI | |
Stellas D and Patsavoudi E: Inhibiting matrix metalloproteinases, an old story with new potentials for cancer treatment. Anticancer Agents Med Chem. 12:707–717. 2012. View Article : Google Scholar : PubMed/NCBI | |
Galis ZS and Khatri JJ: Matrix metalloproteinases in vascular remodeling and atherogenesis: The good, the bad, and the ugly. Circ Res. 90:251–262. 2002.PubMed/NCBI | |
Zuo JH, Zhu W, Li MY, Li XH, Yi H, Zeng GQ, Wan XX, He QY, Li JH, Qu JQ, et al: Activation of EGFR promotes squamous carcinoma SCC10A cell migration and invasion via inducing EMT-like phenotype change and MMP-9-mediated degradation of E-cadherin. J Cell Biochem. 112:2508–2517. 2011. View Article : Google Scholar : PubMed/NCBI | |
Murphy DA and Courtneidge SA: The 'ins' and 'outs' of podosomes and invadopodia: Characteristics, formation and function. Nat Rev Mol Cell Biol. 12:413–426. 2011. View Article : Google Scholar : PubMed/NCBI | |
Bai ZK, Guo B, Tian XC, Li DD, Wang ST, Cao H, Wang QY and Yue ZP: Expression and regulation of Runx3 in mouse uterus during the peri-implantation period. J Mol Histol. 44:519–526. 2013. View Article : Google Scholar : PubMed/NCBI | |
Jinga DC, Blidaru A, Condrea I, Ardeleanu C, Dragomir C, Szegli G, Stefanescu M and Matache C: MMP-9 and MMP-2 gelatinases and TIMP-1 and TIMP-2 inhibitors in breast cancer: Correlations with prognostic factors. J Cell Mol Med. 10:499–510. 2006. View Article : Google Scholar : PubMed/NCBI |