Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

miR-148b-3p inhibits gastric cancer metastasis by inhibiting the Dock6/Rac1/Cdc42 axis

Authors: Xiaowei Li, Mingzuo Jiang, Di Chen, Bing Xu, Rui Wang, Yi Chu, Weijie Wang, Lin Zhou, Zhijie Lei, Yongzhan Nie, Daiming Fan, Yulong Shang, Kaichun Wu, Jie Liang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Our previous work showed that some Rho GTPases, including Rho, Rac1 and Cdc42, play critical roles in gastric cancer (GC); however, how they are regulated in GC remains largely unknown. In this study, we aimed to investigate the roles and molecular mechanisms of Dock6, an atypical Rho guanine nucleotide exchange factor (GEF), in GC metastasis.

Methods

The expression levels of Dock6 and miR-148b-3p in GC tissues and paired nontumor tissues were determined by immunohistochemistry (IHC) and in situ hybridization (ISH), respectively. The correlation between Dock6/miR-148b-3p expression and the overall survival of GC patients was calculated by the Kaplan-Meier method and log-rank test. The roles of Dock6 and miR-148b-3p in GC were investigated by in vitro and in vivo functional studies. Rac1 and Cdc42 activation was investigated by GST pull-down assays. The inhibition of Dock6 transcription by miR-148b-3p was determined by luciferase reporter assays.

Results

A significant increase in Dock6 expression was found in GC tissues compared with nontumor tissues, and its positive expression was associated with lymph node metastasis and a higher TNM stage. Patients with positive Dock6 expression exhibited shorter overall survival periods than patients with negative Dock6 expression. Dock6 promoted GC migration and invasion by increasing the activation of Rac1 and Cdc42. miR-148b-3p expression was negatively correlated with Dock6 expression in GC, and it decreased the motility of GC cells by inhibiting the Dock6/Rac1/Cdc42 axis.

Conclusions

Dock6 was over-expressed in GC tissues, and its positive expression was associated with GC metastasis and indicated poor prognosis of GC patients. Targeting of Dock6 by miR-148b-3p could activate Rac1 and Cdc42, directly affecting the motility of GC cells. Targeting the Dock6-Rac1/Cdc42 axis could serve as a new therapeutic strategy for GC treatment.
Appendix
Available only for authorised users
Literature
1.
go back to reference Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66:115–32.CrossRefPubMed Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66:115–32.CrossRefPubMed
3.
go back to reference Tie J, Pan Y, Zhao L, Wu K, Liu J, Sun S, Guo X, Wang B, Gang Y, Zhang Y, et al. MiR-218 inhibits invasion and metastasis of gastric cancer by targeting the Robo1 receptor. PLoS Genet. 2010;6:e1000879.CrossRefPubMedPubMedCentral Tie J, Pan Y, Zhao L, Wu K, Liu J, Sun S, Guo X, Wang B, Gang Y, Zhang Y, et al. MiR-218 inhibits invasion and metastasis of gastric cancer by targeting the Robo1 receptor. PLoS Genet. 2010;6:e1000879.CrossRefPubMedPubMedCentral
4.
go back to reference Wang SM, Tie J, Wang WL, Hu SJ, Yin JP, Yi XF, Tian ZH, Zhang XY, Li MB, Li ZS, et al. POU2F2-oriented network promotes human gastric cancer metastasis. Gut. 2016;65:1427–38.CrossRefPubMed Wang SM, Tie J, Wang WL, Hu SJ, Yin JP, Yi XF, Tian ZH, Zhang XY, Li MB, Li ZS, et al. POU2F2-oriented network promotes human gastric cancer metastasis. Gut. 2016;65:1427–38.CrossRefPubMed
5.
go back to reference Jansen S, Gosens R, Wieland T, Schmidt M. Paving the rho in cancer metastasis: rho GTPases and beyond. Pharmacol Ther. 2018;183:1-21. Jansen S, Gosens R, Wieland T, Schmidt M. Paving the rho in cancer metastasis: rho GTPases and beyond. Pharmacol Ther. 2018;183:1-21.
6.
go back to reference Kazanietz MG, Caloca MJ. The Rac GTPase in Cancer: from old concepts to new paradigms. Cancer Res. 2017;77:5445–51.CrossRefPubMed Kazanietz MG, Caloca MJ. The Rac GTPase in Cancer: from old concepts to new paradigms. Cancer Res. 2017;77:5445–51.CrossRefPubMed
8.
go back to reference Kong R, Yi F, Wen P, Liu J, Chen X, Ren J, Li X, Shang Y, Nie Y, Wu K, et al. Myo9b is a key player in SLIT/ROBO-mediated lung tumor suppression. J Clin Invest. 2015;125:4407–20.CrossRefPubMedPubMedCentral Kong R, Yi F, Wen P, Liu J, Chen X, Ren J, Li X, Shang Y, Nie Y, Wu K, et al. Myo9b is a key player in SLIT/ROBO-mediated lung tumor suppression. J Clin Invest. 2015;125:4407–20.CrossRefPubMedPubMedCentral
10.
go back to reference Li K, Lu Y, Liang J, Luo G, Ren G, Wang X, Fan D. RhoE enhances multidrug resistance of gastric cancer cells by suppressing Bax. Biochem Biophys Res Commun. 2009;379:212–6.CrossRefPubMed Li K, Lu Y, Liang J, Luo G, Ren G, Wang X, Fan D. RhoE enhances multidrug resistance of gastric cancer cells by suppressing Bax. Biochem Biophys Res Commun. 2009;379:212–6.CrossRefPubMed
11.
go back to reference Liu N, Zhang G, Bi F, Pan Y, Xue Y, Shi Y, Yao L, Zhao L, Zheng Y, Fan D. RhoC is essential for the metastasis of gastric cancer. J Mol Med (Berl). 2007;85:1149–56.CrossRef Liu N, Zhang G, Bi F, Pan Y, Xue Y, Shi Y, Yao L, Zhao L, Zheng Y, Fan D. RhoC is essential for the metastasis of gastric cancer. J Mol Med (Berl). 2007;85:1149–56.CrossRef
12.
go back to reference Pan Y, Bi F, Liu N, Xue Y, Yao X, Zheng Y, Fan D. Expression of seven main rho family members in gastric carcinoma. Biochem Biophys Res Commun. 2004;315:686–91.CrossRefPubMed Pan Y, Bi F, Liu N, Xue Y, Yao X, Zheng Y, Fan D. Expression of seven main rho family members in gastric carcinoma. Biochem Biophys Res Commun. 2004;315:686–91.CrossRefPubMed
13.
go back to reference Xue Y, Bi F, Zhang X, Pan Y, Liu N, Zheng Y, Fan D. Inhibition of endothelial cell proliferation by targeting Rac1 GTPase with small interference RNA in tumor cells. Biochem Biophys Res Commun. 2004;320:1309–15.CrossRefPubMed Xue Y, Bi F, Zhang X, Pan Y, Liu N, Zheng Y, Fan D. Inhibition of endothelial cell proliferation by targeting Rac1 GTPase with small interference RNA in tumor cells. Biochem Biophys Res Commun. 2004;320:1309–15.CrossRefPubMed
14.
go back to reference Xue Y, Bi F, Zhang X, Zhang S, Pan Y, Liu N, Shi Y, Yao X, Zheng Y, Fan D. Role of Rac1 and Cdc42 in hypoxia induced p53 and von Hippel-Lindau suppression and HIF1alpha activation. Int J Cancer. 2006;118:2965–72.CrossRefPubMed Xue Y, Bi F, Zhang X, Zhang S, Pan Y, Liu N, Shi Y, Yao X, Zheng Y, Fan D. Role of Rac1 and Cdc42 in hypoxia induced p53 and von Hippel-Lindau suppression and HIF1alpha activation. Int J Cancer. 2006;118:2965–72.CrossRefPubMed
15.
go back to reference Gadea G, Blangy A. Dock-family exchange factors in cell migration and disease. Eur J Cell Biol. 2014;93:466–77.CrossRefPubMed Gadea G, Blangy A. Dock-family exchange factors in cell migration and disease. Eur J Cell Biol. 2014;93:466–77.CrossRefPubMed
17.
go back to reference Namekata K, Kimura A, Kawamura K, Harada C, Harada T. Dock GEFs and their therapeutic potential: neuroprotection and axon regeneration. Prog Retin Eye Res. 2014;43:1–16.CrossRefPubMed Namekata K, Kimura A, Kawamura K, Harada C, Harada T. Dock GEFs and their therapeutic potential: neuroprotection and axon regeneration. Prog Retin Eye Res. 2014;43:1–16.CrossRefPubMed
18.
go back to reference Nishikimi A, Kukimoto-Niino M, Yokoyama S, Fukui Y. Immune regulatory functions of DOCK family proteins in health and disease. Exp Cell Res. 2013;319:2343–9.CrossRefPubMed Nishikimi A, Kukimoto-Niino M, Yokoyama S, Fukui Y. Immune regulatory functions of DOCK family proteins in health and disease. Exp Cell Res. 2013;319:2343–9.CrossRefPubMed
19.
go back to reference Premkumar L, Bobkov AA, Patel M, Jaroszewski L, Bankston LA, Stec B, Vuori K, Cote JF, Liddington RC. Structural basis of membrane targeting by the Dock180 family of rho family guanine exchange factors (rho-GEFs). J Biol Chem. 2010;285:13211–22.CrossRefPubMedPubMedCentral Premkumar L, Bobkov AA, Patel M, Jaroszewski L, Bankston LA, Stec B, Vuori K, Cote JF, Liddington RC. Structural basis of membrane targeting by the Dock180 family of rho family guanine exchange factors (rho-GEFs). J Biol Chem. 2010;285:13211–22.CrossRefPubMedPubMedCentral
21.
go back to reference Sanz-Moreno V, Gadea G, Ahn J, Paterson H, Marra P, Pinner S, Sahai E, Marshall CJ. Rac activation and inactivation control plasticity of tumor cell movement. Cell. 2008;135:510–23.CrossRefPubMed Sanz-Moreno V, Gadea G, Ahn J, Paterson H, Marra P, Pinner S, Sahai E, Marshall CJ. Rac activation and inactivation control plasticity of tumor cell movement. Cell. 2008;135:510–23.CrossRefPubMed
23.
go back to reference Ruiz-Lafuente N, Alcaraz-Garcia MJ, Garcia-Serna AM, Sebastian-Ruiz S, Moya-Quiles MR, Garcia-Alonso AM, Parrado A. Dock10, a Cdc42 and Rac1 GEF, induces loss of elongation, filopodia, and ruffles in cervical cancer epithelial HeLa cells. Biol Open. 2015;4:627–35.CrossRefPubMedPubMedCentral Ruiz-Lafuente N, Alcaraz-Garcia MJ, Garcia-Serna AM, Sebastian-Ruiz S, Moya-Quiles MR, Garcia-Alonso AM, Parrado A. Dock10, a Cdc42 and Rac1 GEF, induces loss of elongation, filopodia, and ruffles in cervical cancer epithelial HeLa cells. Biol Open. 2015;4:627–35.CrossRefPubMedPubMedCentral
24.
go back to reference Yelo E, Bernardo MV, Gimeno L, Alcaraz-Garcia MJ, Majado MJ, Parrado A. Dock10, a novel CZH protein selectively induced by interleukin-4 in human B lymphocytes. Mol Immunol. 2008;45:3411–8.CrossRefPubMed Yelo E, Bernardo MV, Gimeno L, Alcaraz-Garcia MJ, Majado MJ, Parrado A. Dock10, a novel CZH protein selectively induced by interleukin-4 in human B lymphocytes. Mol Immunol. 2008;45:3411–8.CrossRefPubMed
25.
go back to reference Murray DW, Didier S, Chan A, Paulino V, Van Aelst L, Ruggieri R, Tran NL, Byrne AT, Symons M. Guanine nucleotide exchange factor Dock7 mediates HGF-induced glioblastoma cell invasion via Rac activation. Br J Cancer. 2014;110:1307–15.CrossRefPubMedPubMedCentral Murray DW, Didier S, Chan A, Paulino V, Van Aelst L, Ruggieri R, Tran NL, Byrne AT, Symons M. Guanine nucleotide exchange factor Dock7 mediates HGF-induced glioblastoma cell invasion via Rac activation. Br J Cancer. 2014;110:1307–15.CrossRefPubMedPubMedCentral
26.
go back to reference Hernandez-Garcia R, Iruela-Arispe ML, Reyes-Cruz G, Vazquez-Prado J. Endothelial RhoGEFs: a systematic analysis of their expression profiles in VEGF-stimulated and tumor endothelial cells. Vasc Pharmacol. 2015;74:60–72.CrossRef Hernandez-Garcia R, Iruela-Arispe ML, Reyes-Cruz G, Vazquez-Prado J. Endothelial RhoGEFs: a systematic analysis of their expression profiles in VEGF-stimulated and tumor endothelial cells. Vasc Pharmacol. 2015;74:60–72.CrossRef
27.
go back to reference Yuan X, Wang X, Gu B, Ma Y, Liu Y, Sun M, Kong J, Sun W, Wang H, Zhou F, Gao S. Directional migration in esophageal squamous cell carcinoma (ESCC) is epigenetically regulated by SET nuclear oncogene, a member of the inhibitor of histone Acetyltransferase complex. Neoplasia. 2017;19:868–84.CrossRefPubMedPubMedCentral Yuan X, Wang X, Gu B, Ma Y, Liu Y, Sun M, Kong J, Sun W, Wang H, Zhou F, Gao S. Directional migration in esophageal squamous cell carcinoma (ESCC) is epigenetically regulated by SET nuclear oncogene, a member of the inhibitor of histone Acetyltransferase complex. Neoplasia. 2017;19:868–84.CrossRefPubMedPubMedCentral
28.
go back to reference Miyamoto Y, Torii T, Yamamori N, Ogata T, Tanoue A, Yamauchi J. Akt and PP2A reciprocally regulate the guanine nucleotide exchange factor Dock6 to control axon growth of sensory neurons. Sci Signal. 2013;6:ra15.CrossRefPubMed Miyamoto Y, Torii T, Yamamori N, Ogata T, Tanoue A, Yamauchi J. Akt and PP2A reciprocally regulate the guanine nucleotide exchange factor Dock6 to control axon growth of sensory neurons. Sci Signal. 2013;6:ra15.CrossRefPubMed
29.
go back to reference Ruusala A, Aspenstrom P. Isolation and characterisation of DOCK8, a member of the DOCK180-related regulators of cell morphology. FEBS Lett. 2004;572:159–66.CrossRefPubMed Ruusala A, Aspenstrom P. Isolation and characterisation of DOCK8, a member of the DOCK180-related regulators of cell morphology. FEBS Lett. 2004;572:159–66.CrossRefPubMed
30.
go back to reference Bao W, Fu HJ, Xie QS, Wang L, Zhang R, Guo ZY, Zhao J, Meng YL, Ren XL, Wang T, et al. HER2 interacts with CD44 to up-regulate CXCR4 via epigenetic silencing of microRNA-139 in gastric cancer cells. Gastroenterology. 2011;141:2076–87. e2076CrossRefPubMed Bao W, Fu HJ, Xie QS, Wang L, Zhang R, Guo ZY, Zhao J, Meng YL, Ren XL, Wang T, et al. HER2 interacts with CD44 to up-regulate CXCR4 via epigenetic silencing of microRNA-139 in gastric cancer cells. Gastroenterology. 2011;141:2076–87. e2076CrossRefPubMed
31.
go back to reference Wu M, Ye X, Wang S, Li Q, Lai Y, Yi Y. MicroRNA-148b suppresses proliferation, migration, and invasion of nasopharyngeal carcinoma cells by targeting metastasis-associated gene 2. Onco Targets Ther. 2017;10:2815–22.CrossRefPubMedPubMedCentral Wu M, Ye X, Wang S, Li Q, Lai Y, Yi Y. MicroRNA-148b suppresses proliferation, migration, and invasion of nasopharyngeal carcinoma cells by targeting metastasis-associated gene 2. Onco Targets Ther. 2017;10:2815–22.CrossRefPubMedPubMedCentral
32.
go back to reference Zhou Z, Su Y, Fa X. Restoration of BRG1 inhibits proliferation and metastasis of lung cancer by regulating tumor suppressor miR-148b. Onco Targets Ther. 2015;8:3603–12.PubMedPubMedCentral Zhou Z, Su Y, Fa X. Restoration of BRG1 inhibits proliferation and metastasis of lung cancer by regulating tumor suppressor miR-148b. Onco Targets Ther. 2015;8:3603–12.PubMedPubMedCentral
33.
go back to reference Chen X, Bo L, Lu W, Zhou G, Chen Q. MicroRNA-148b targets rho-associated protein kinase 1 to inhibit cell proliferation, migration and invasion in hepatocellular carcinoma. Mol Med Rep. 2016;13:477–82.CrossRefPubMed Chen X, Bo L, Lu W, Zhou G, Chen Q. MicroRNA-148b targets rho-associated protein kinase 1 to inhibit cell proliferation, migration and invasion in hepatocellular carcinoma. Mol Med Rep. 2016;13:477–82.CrossRefPubMed
34.
go back to reference Zhao G, Zhang JG, Liu Y, Qin Q, Wang B, Tian K, Liu L, Li X, Niu Y, Deng SC, Wang CY. miR-148b functions as a tumor suppressor in pancreatic cancer by targeting AMPKalpha1. Mol Cancer Ther. 2013;12:83–93.CrossRefPubMed Zhao G, Zhang JG, Liu Y, Qin Q, Wang B, Tian K, Liu L, Li X, Niu Y, Deng SC, Wang CY. miR-148b functions as a tumor suppressor in pancreatic cancer by targeting AMPKalpha1. Mol Cancer Ther. 2013;12:83–93.CrossRefPubMed
35.
go back to reference Chen X, Wang G, Lu X, Gao P, Song Y, Sun J, Li A, Xu Y, Xu H, Wang Z. Polymorphisms and haplotypes of the miR-148/152 family are associated with the risk and clinicopathological features of gastric cancer in a northern Chinese population. Mutagenesis. 2014;29:401–7.CrossRefPubMed Chen X, Wang G, Lu X, Gao P, Song Y, Sun J, Li A, Xu Y, Xu H, Wang Z. Polymorphisms and haplotypes of the miR-148/152 family are associated with the risk and clinicopathological features of gastric cancer in a northern Chinese population. Mutagenesis. 2014;29:401–7.CrossRefPubMed
36.
37.
go back to reference Song YX, Yue ZY, Wang ZN, Xu YY, Luo Y, Xu HM, Zhang X, Jiang L, Xing CZ, Zhang Y. MicroRNA-148b is frequently down-regulated in gastric cancer and acts as a tumor suppressor by inhibiting cell proliferation. Mol Cancer. 2011;10(1):1-13. Song YX, Yue ZY, Wang ZN, Xu YY, Luo Y, Xu HM, Zhang X, Jiang L, Xing CZ, Zhang Y. MicroRNA-148b is frequently down-regulated in gastric cancer and acts as a tumor suppressor by inhibiting cell proliferation. Mol Cancer. 2011;10(1):1-13.
38.
go back to reference Dong J, Wang R, Ren G, Li X, Wang J, Sun Y, Liang J, Nie Y, Wu K, Feng B, et al. HMGA2-FOXL2 Axis regulates metastases and epithelial-to-Mesenchymal transition of Chemoresistant gastric Cancer. Clin Cancer Res. 2017;;23(13):3461-73. Dong J, Wang R, Ren G, Li X, Wang J, Sun Y, Liang J, Nie Y, Wu K, Feng B, et al. HMGA2-FOXL2 Axis regulates metastases and epithelial-to-Mesenchymal transition of Chemoresistant gastric Cancer. Clin Cancer Res. 2017;;23(13):3461-73.
39.
go back to reference Huang W, Chen Z, Shang X, Tian D, Wang D, Wu K, Fan D, Xia L. Sox12, a direct target of FoxQ1, promotes hepatocellular carcinoma metastasis through up-regulating Twist1 and FGFBP1. Hepatology. 2015;61:1920–33.CrossRefPubMed Huang W, Chen Z, Shang X, Tian D, Wang D, Wu K, Fan D, Xia L. Sox12, a direct target of FoxQ1, promotes hepatocellular carcinoma metastasis through up-regulating Twist1 and FGFBP1. Hepatology. 2015;61:1920–33.CrossRefPubMed
40.
go back to reference Szasz AM, Lanczky A, Nagy A, Forster S, Hark K, Green JE, Boussioutas A, Busuttil R, Szabo A, Gyorffy B. Cross-validation of survival associated biomarkers in gastric cancer using transcriptomic data of 1,065 patients. Oncotarget. 2016;7:49322–33.CrossRefPubMedPubMedCentral Szasz AM, Lanczky A, Nagy A, Forster S, Hark K, Green JE, Boussioutas A, Busuttil R, Szabo A, Gyorffy B. Cross-validation of survival associated biomarkers in gastric cancer using transcriptomic data of 1,065 patients. Oncotarget. 2016;7:49322–33.CrossRefPubMedPubMedCentral
41.
go back to reference Miyamoto Y, Yamauchi J, Sanbe A, Tanoue A. Dock6, a Dock-C subfamily guanine nucleotide exchanger, has the dual specificity for Rac1 and Cdc42 and regulates neurite outgrowth. Exp Cell Res. 2007;313:791–804.CrossRefPubMed Miyamoto Y, Yamauchi J, Sanbe A, Tanoue A. Dock6, a Dock-C subfamily guanine nucleotide exchanger, has the dual specificity for Rac1 and Cdc42 and regulates neurite outgrowth. Exp Cell Res. 2007;313:791–804.CrossRefPubMed
42.
go back to reference Kim K, Yang DK, Kim S, Kang H. miR-142-3p is a regulator of the TGFbeta-mediated vascular smooth muscle cell phenotype. J Cell Biochem. 2015;116:2325–33.CrossRefPubMed Kim K, Yang DK, Kim S, Kang H. miR-142-3p is a regulator of the TGFbeta-mediated vascular smooth muscle cell phenotype. J Cell Biochem. 2015;116:2325–33.CrossRefPubMed
43.
go back to reference Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA. The cBio Cancer genomics portal: an open platform for exploring multidimensional Cancer genomics data (vol 2, pg 401, 2012). Cancer Discov. 2012;2:960.CrossRef Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA. The cBio Cancer genomics portal: an open platform for exploring multidimensional Cancer genomics data (vol 2, pg 401, 2012). Cancer Discov. 2012;2:960.CrossRef
45.
go back to reference Cerikan B, Shaheen R, Colo GP, Glasser C, Hata S, Knobeloch KP, Alkuraya FS, Fassler R, Schiebel E. Cell-intrinsic adaptation arising from chronic ablation of a key rho GTPase regulator. Dev Cell. 2016;39:28–43.CrossRefPubMed Cerikan B, Shaheen R, Colo GP, Glasser C, Hata S, Knobeloch KP, Alkuraya FS, Fassler R, Schiebel E. Cell-intrinsic adaptation arising from chronic ablation of a key rho GTPase regulator. Dev Cell. 2016;39:28–43.CrossRefPubMed
46.
go back to reference McGarry DJ, Olson MF. Coping with loss: cell adaptation to cytoskeleton disruption. Dev Cell. 2016;39:3–4.CrossRefPubMed McGarry DJ, Olson MF. Coping with loss: cell adaptation to cytoskeleton disruption. Dev Cell. 2016;39:3–4.CrossRefPubMed
47.
go back to reference Cerikan B, Schiebel E. DOCK6 inactivation highlights ISGylation as RHO-GTPase balancer. Cell Cycle. 2017;16:304–5.CrossRefPubMed Cerikan B, Schiebel E. DOCK6 inactivation highlights ISGylation as RHO-GTPase balancer. Cell Cycle. 2017;16:304–5.CrossRefPubMed
48.
go back to reference Cerikan B, Schiebel E. Mechanism of cell-intrinsic adaptation to Adams-Oliver syndrome gene DOCK6 disruption highlights ubiquitin-like modifier ISG15 as a regulator of RHO GTPases. Small GTPases. 2017;23:1–8. Cerikan B, Schiebel E. Mechanism of cell-intrinsic adaptation to Adams-Oliver syndrome gene DOCK6 disruption highlights ubiquitin-like modifier ISG15 as a regulator of RHO GTPases. Small GTPases. 2017;23:1–8.
Metadata
Title
miR-148b-3p inhibits gastric cancer metastasis by inhibiting the Dock6/Rac1/Cdc42 axis
Authors
Xiaowei Li
Mingzuo Jiang
Di Chen
Bing Xu
Rui Wang
Yi Chu
Weijie Wang
Lin Zhou
Zhijie Lei
Yongzhan Nie
Daiming Fan
Yulong Shang
Kaichun Wu
Jie Liang
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0729-z

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine