Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01-12-2019 | Hepatocellular Carcinoma | Research

Cx32 exerts anti-apoptotic and pro-tumor effects via the epidermal growth factor receptor pathway in hepatocellular carcinoma

Authors: Yuke Xiang, Qin Wang, Yunquan Guo, Hui Ge, Yile Fu, Xiyan Wang, Liang Tao

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Abnormal expression or distribution of connexin 32 (Cx32) is associated with hepatocarcinogenesis, but the role of Cx32 and the underlying mechanisms are still unclear.

Methods

The expression level of Cx32 in 96 hepatocellular carcinoma (HCC) specimens was determined using western blotting and immunohistochemistry. The correlation between Cx32 expression and clinicopathological parameters was analyzed. The cell apoptosis rate was examined using flow cytometry and western blotting. The role of Cx32 in the Src kinase and epidermal growth factor receptor (EGFR) signaling pathways was measured by quantitative real-time PCR, western blotting and coimmunoprecipitation (CO-IP). The effect of Cx32 overexpression on the streptonigrin (SN)-induced tumor growth suppression and apoptosis was assessed in nude mice.

Results

Our study showed that overexpressed Cx32 accumulated in the cytoplasm and that Cx32-containing gap junctions (GJs) were nearly absent in HCC specimens. Upregulated Cx32 expression was highly correlated with advanced tumor-node-metastasis (TNM) stage and poor tumor differentiation and was an independent predictive marker for poor prognosis in HCC. Overexpression of Cx32 significantly inhibited SN-induced apoptosis by activating the EGFR signaling pathway in vitro and in vivo. Moreover, the expression levels of Cx32 and EGFR were positively correlated in HCC specimens. The CO-IP experiments demonstrated that Cx32 could bind to Src kinase, and the western blotting results revealed that Cx32 increased the levels of EGFR and p-EGFR by upregulating Src expression.

Conclusion

The present study demonstrated that overexpressed and internalized Cx32 was associated with advanced TNM stage and poor tumor differentiation and predicted poor prognosis in HCC. Cx32 facilitated HCC progression by blocking chemotherapy-induced apoptosis in vitro and in vivo via interacting with Src and thus promoting the phosphorylation of EGFR, subsequently activating the EGFR signaling pathway. Cx32 may be a potential biomarker and a new therapeutic target for HCC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Beyer EC, Berthoud VM. Gap junction gene and protein families: Connexins, innexins, and pannexins. Biochim Biophys Acta Biomembr. 2018;1860:5–8.CrossRef Beyer EC, Berthoud VM. Gap junction gene and protein families: Connexins, innexins, and pannexins. Biochim Biophys Acta Biomembr. 2018;1860:5–8.CrossRef
2.
go back to reference Beyer EC, Berthoud VM. Gap junction structure: unraveled, but not fully revealed. F1000Research. 2017;6:568.CrossRef Beyer EC, Berthoud VM. Gap junction structure: unraveled, but not fully revealed. F1000Research. 2017;6:568.CrossRef
3.
go back to reference Aasen T, Mesnil M, Naus CC, Lampe PD, Laird DW. Gap junctions and cancer: communicating for 50 years. Nat Rev Cancer. 2016;16:775–88.CrossRef Aasen T, Mesnil M, Naus CC, Lampe PD, Laird DW. Gap junctions and cancer: communicating for 50 years. Nat Rev Cancer. 2016;16:775–88.CrossRef
4.
go back to reference Mao XY, Li QQ, Gao YF, Zhou HH, Liu ZQ, Jin WL. Gap junction as an intercellular glue: emerging roles in cancer EMT and metastasis. Cancer Lett. 2016;381:133–7.CrossRef Mao XY, Li QQ, Gao YF, Zhou HH, Liu ZQ, Jin WL. Gap junction as an intercellular glue: emerging roles in cancer EMT and metastasis. Cancer Lett. 2016;381:133–7.CrossRef
5.
go back to reference Zhang Y, Tao L, Fan L, Peng Y, Yang K, Zhao Y, et al. Different gap junction-propagated effects on cisplatin transfer result in opposite responses to cisplatin in normal cells versus tumor cells. Sci Rep. 2015;5:12563.CrossRef Zhang Y, Tao L, Fan L, Peng Y, Yang K, Zhao Y, et al. Different gap junction-propagated effects on cisplatin transfer result in opposite responses to cisplatin in normal cells versus tumor cells. Sci Rep. 2015;5:12563.CrossRef
6.
go back to reference Kandouz M, Batist G. Gap junctions and connexins as therapeutic targets in cancer. Expert Opin Ther Targets. 2010;14:681–92.CrossRef Kandouz M, Batist G. Gap junctions and connexins as therapeutic targets in cancer. Expert Opin Ther Targets. 2010;14:681–92.CrossRef
7.
go back to reference Naus CC, Laird DW. Implications and challenges of connexin connections to cancer. Nat Rev Cancer. 2010;10:435–41.CrossRef Naus CC, Laird DW. Implications and challenges of connexin connections to cancer. Nat Rev Cancer. 2010;10:435–41.CrossRef
8.
go back to reference Vinken M, Decrock E, Leybaert L, Bultynck G, Himpens B, Vanhaecke T, et al. Non-channel functions of connexins in cell growth and cell death. Biochim Biophys Acta. 2012;1818:2002–8.CrossRef Vinken M, Decrock E, Leybaert L, Bultynck G, Himpens B, Vanhaecke T, et al. Non-channel functions of connexins in cell growth and cell death. Biochim Biophys Acta. 2012;1818:2002–8.CrossRef
9.
go back to reference Chen SC, Pelletier DB, Ao P, Boynton AL. Connexin43 reverses the phenotype of transformed cells and alters their expression of cyclin/cyclin-dependent kinases. Cell Growth Differ. 1995;6:681–90.PubMed Chen SC, Pelletier DB, Ao P, Boynton AL. Connexin43 reverses the phenotype of transformed cells and alters their expression of cyclin/cyclin-dependent kinases. Cell Growth Differ. 1995;6:681–90.PubMed
10.
go back to reference Kanczuga-Koda L, Sulkowski S, Koda M, Skrzydlewska E, Sulkowska M. Connexin 26 correlates with Bcl-xL and Bax proteins expression in colorectal cancer. World J Gastroenterol. 2005;11:1544–8.CrossRef Kanczuga-Koda L, Sulkowski S, Koda M, Skrzydlewska E, Sulkowska M. Connexin 26 correlates with Bcl-xL and Bax proteins expression in colorectal cancer. World J Gastroenterol. 2005;11:1544–8.CrossRef
11.
go back to reference Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet (London, England). 2012;379:1245–1255. Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet (London, England). 2012;379:1245–1255.
12.
go back to reference Maluccio M, Covey A. Recent progress in understanding, diagnosing, and treating hepatocellular carcinoma. CA Cancer J Clin. 2012;62:394–9.CrossRef Maluccio M, Covey A. Recent progress in understanding, diagnosing, and treating hepatocellular carcinoma. CA Cancer J Clin. 2012;62:394–9.CrossRef
13.
go back to reference Vinken M, De Kock J, Oliveira AG, Menezes GB, Cogliati B, Dagli ML, et al. Modifications in connexin expression in liver development and cancer. Cell Commun Adhes. 2012;19:55–62.CrossRef Vinken M, De Kock J, Oliveira AG, Menezes GB, Cogliati B, Dagli ML, et al. Modifications in connexin expression in liver development and cancer. Cell Commun Adhes. 2012;19:55–62.CrossRef
14.
go back to reference Wu L, Zhou WB, Shen F, Liu W, Wu HW, Zhou SJ, et al. Connexin32mediated antitumor effects of suicide gene therapy against hepatocellular carcinoma: in vitro and in vivo anticancer activity. Mol Med Rep. 2016;13:3213–9.CrossRef Wu L, Zhou WB, Shen F, Liu W, Wu HW, Zhou SJ, et al. Connexin32mediated antitumor effects of suicide gene therapy against hepatocellular carcinoma: in vitro and in vivo anticancer activity. Mol Med Rep. 2016;13:3213–9.CrossRef
15.
go back to reference Dagli ML, Yamasaki H, Krutovskikh V, Omori Y. Delayed liver regeneration and increased susceptibility to chemical hepatocarcinogenesis in transgenic mice expressing a dominant-negative mutant of connexin32 only in the liver. Carcinogenesis. 2004;25:483–92.CrossRef Dagli ML, Yamasaki H, Krutovskikh V, Omori Y. Delayed liver regeneration and increased susceptibility to chemical hepatocarcinogenesis in transgenic mice expressing a dominant-negative mutant of connexin32 only in the liver. Carcinogenesis. 2004;25:483–92.CrossRef
16.
go back to reference Hokaiwado N, Asamoto M, Futakuchi M, Ogawa K, Takahashi S, Shirai T. Both early and late stages of hepatocarcinogenesis are enhanced in Cx32 dominant negative mutant transgenic rats with disrupted gap junctional intercellular communication. J Membr Biol. 2007;218:101–6.CrossRef Hokaiwado N, Asamoto M, Futakuchi M, Ogawa K, Takahashi S, Shirai T. Both early and late stages of hepatocarcinogenesis are enhanced in Cx32 dominant negative mutant transgenic rats with disrupted gap junctional intercellular communication. J Membr Biol. 2007;218:101–6.CrossRef
17.
go back to reference Zhao B, Zhao W, Wang Y, Xu Y, Xu J, Tang K, et al. Connexin32 regulates hepatoma cell metastasis and proliferation via the p53 and Akt pathways. Oncotarget. 2015;6:10116–33.PubMed Zhao B, Zhao W, Wang Y, Xu Y, Xu J, Tang K, et al. Connexin32 regulates hepatoma cell metastasis and proliferation via the p53 and Akt pathways. Oncotarget. 2015;6:10116–33.PubMed
18.
go back to reference Li Q, Omori Y, Nishikawa Y, Yoshioka T, Yamamoto Y, Enomoto K. Cytoplasmic accumulation of connexin32 protein enhances motility and metastatic ability of human hepatoma cells in vitro and in vivo. Int J Cancer. 2007;121:536–46.CrossRef Li Q, Omori Y, Nishikawa Y, Yoshioka T, Yamamoto Y, Enomoto K. Cytoplasmic accumulation of connexin32 protein enhances motility and metastatic ability of human hepatoma cells in vitro and in vivo. Int J Cancer. 2007;121:536–46.CrossRef
19.
go back to reference Kawasaki Y, Omori Y, Li Q, Nishikawa Y, Yoshioka T, Yoshida M, et al. Cytoplasmic accumulation of connexin32 expands cancer stem cell population in human HuH7 hepatoma cells by enhancing its self-renewal. Int J Cancer. 2011;128:51–62.CrossRef Kawasaki Y, Omori Y, Li Q, Nishikawa Y, Yoshioka T, Yoshida M, et al. Cytoplasmic accumulation of connexin32 expands cancer stem cell population in human HuH7 hepatoma cells by enhancing its self-renewal. Int J Cancer. 2011;128:51–62.CrossRef
20.
go back to reference Schwarz M, Wanke I, Wulbrand U, Moennikes O, Buchmann A. Role of connexin32 and beta-catenin in tumor promotion in mouse liver. Toxicol Pathol. 2003;31:99–102.PubMed Schwarz M, Wanke I, Wulbrand U, Moennikes O, Buchmann A. Role of connexin32 and beta-catenin in tumor promotion in mouse liver. Toxicol Pathol. 2003;31:99–102.PubMed
21.
go back to reference Moennikes O, Buchmann A, Romualdi A, Ott T, Werringloer J, Willecke K, et al. Lack of phenobarbital-mediated promotion of hepatocarcinogenesis in connexin32-null mice. Cancer Res. 2000;60:5087–91.PubMed Moennikes O, Buchmann A, Romualdi A, Ott T, Werringloer J, Willecke K, et al. Lack of phenobarbital-mediated promotion of hepatocarcinogenesis in connexin32-null mice. Cancer Res. 2000;60:5087–91.PubMed
22.
go back to reference Zhao Y, Lai Y, Ge H, Guo Y, Feng X, Song J, et al. Non-junctional Cx32 mediates anti-apoptotic and pro-tumor effects via epidermal growth factor receptor in human cervical cancer cells. Cell Death Dis. 2017;8:e2773.CrossRef Zhao Y, Lai Y, Ge H, Guo Y, Feng X, Song J, et al. Non-junctional Cx32 mediates anti-apoptotic and pro-tumor effects via epidermal growth factor receptor in human cervical cancer cells. Cell Death Dis. 2017;8:e2773.CrossRef
23.
go back to reference Lai Y, Fan L, Zhao Y, Ge H, Feng X, Wang Q, et al. Cx32 suppresses extrinsic apoptosis in human cervical cancer cells via the NFkappaB signalling pathway. Int J Oncol. 2017;51:1159–68.CrossRef Lai Y, Fan L, Zhao Y, Ge H, Feng X, Wang Q, et al. Cx32 suppresses extrinsic apoptosis in human cervical cancer cells via the NFkappaB signalling pathway. Int J Oncol. 2017;51:1159–68.CrossRef
24.
go back to reference Tao L, Harris AL. 2-aminoethoxydiphenyl borate directly inhibits channels composed of connexin26 and/or connexin32. Mol Pharmacol. 2007;71:570–9.CrossRef Tao L, Harris AL. 2-aminoethoxydiphenyl borate directly inhibits channels composed of connexin26 and/or connexin32. Mol Pharmacol. 2007;71:570–9.CrossRef
25.
go back to reference Lee SH, Lee WK, Kim N, Kang JH, Kim KH, Kim SG, et al. Renal cell carcinoma is abrogated by p53 stabilization through transglutaminase 2 inhibition, vol. 10; 2018. Lee SH, Lee WK, Kim N, Kang JH, Kim KH, Kim SG, et al. Renal cell carcinoma is abrogated by p53 stabilization through transglutaminase 2 inhibition, vol. 10; 2018.
26.
go back to reference Wang ZS, Wu LQ, Yi X, Geng C, Li YJ, Yao RY. Connexin-43 can delay early recurrence and metastasis in patients with hepatitis B-related hepatocellular carcinoma and low serum alpha-fetoprotein after radical hepatectomy. BMC Cancer. 2013;13:306.CrossRef Wang ZS, Wu LQ, Yi X, Geng C, Li YJ, Yao RY. Connexin-43 can delay early recurrence and metastasis in patients with hepatitis B-related hepatocellular carcinoma and low serum alpha-fetoprotein after radical hepatectomy. BMC Cancer. 2013;13:306.CrossRef
27.
go back to reference Ionta M, Ferreira RA, Pfister SC, Machado-Santelli GM. Exogenous Cx43 expression decrease cell proliferation rate in rat hepatocarcinoma cells independently of functional gap junction. Cancer Cell Int. 2009;9:22.CrossRef Ionta M, Ferreira RA, Pfister SC, Machado-Santelli GM. Exogenous Cx43 expression decrease cell proliferation rate in rat hepatocarcinoma cells independently of functional gap junction. Cancer Cell Int. 2009;9:22.CrossRef
28.
go back to reference Maa MC, Leu TH, McCarley DJ, Schatzman RC, Parsons SJ. Potentiation of epidermal growth factor receptor-mediated oncogenesis by c-Src: implications for the etiology of multiple human cancers. Proc Natl Acad Sci U S A. 1995;92:6981–5.CrossRef Maa MC, Leu TH, McCarley DJ, Schatzman RC, Parsons SJ. Potentiation of epidermal growth factor receptor-mediated oncogenesis by c-Src: implications for the etiology of multiple human cancers. Proc Natl Acad Sci U S A. 1995;92:6981–5.CrossRef
29.
go back to reference Hong X, Wang Q, Yang Y, Zheng S, Tong X, Zhang S, et al. Gap junctions propagate opposite effects in normal and tumor testicular cells in response to cisplatin. Cancer Lett. 2012;317:165–71.CrossRef Hong X, Wang Q, Yang Y, Zheng S, Tong X, Zhang S, et al. Gap junctions propagate opposite effects in normal and tumor testicular cells in response to cisplatin. Cancer Lett. 2012;317:165–71.CrossRef
30.
go back to reference Carette D, Gilleron J, Chevallier D, Segretain D, Pointis G. Connexin a check-point component of cell apoptosis in normal and physiopathological conditions. Biochimie. 2014;101:1–9.CrossRef Carette D, Gilleron J, Chevallier D, Segretain D, Pointis G. Connexin a check-point component of cell apoptosis in normal and physiopathological conditions. Biochimie. 2014;101:1–9.CrossRef
31.
go back to reference Krysko DV, Leybaert L, Vandenabeele P, D'Herde K. Gap junctions and the propagation of cell survival and cell death signals. Apoptosis. 2005;10:459–69.CrossRef Krysko DV, Leybaert L, Vandenabeele P, D'Herde K. Gap junctions and the propagation of cell survival and cell death signals. Apoptosis. 2005;10:459–69.CrossRef
32.
go back to reference Decrock E, Vinken M, De Vuyst E, Krysko DV, D'Herde K, Vanhaecke T, et al. Connexin-related signaling in cell death: to live or let die? Cell Death Differ. 2009;16:524–36.CrossRef Decrock E, Vinken M, De Vuyst E, Krysko DV, D'Herde K, Vanhaecke T, et al. Connexin-related signaling in cell death: to live or let die? Cell Death Differ. 2009;16:524–36.CrossRef
33.
go back to reference Wu W, Fan L, Bao Z, Zhang Y, Peng Y, Shao M, et al. The cytoplasmic translocation of Cx32 mediates cisplatin resistance in ovarian cancer cells. Biochem Biophys Res Commun. 2017;487:292–9.CrossRef Wu W, Fan L, Bao Z, Zhang Y, Peng Y, Shao M, et al. The cytoplasmic translocation of Cx32 mediates cisplatin resistance in ovarian cancer cells. Biochem Biophys Res Commun. 2017;487:292–9.CrossRef
34.
go back to reference Falk MM, Bell CL, Kells Andrews RM, Murray SA. Molecular mechanisms regulating formation, trafficking and processing of annular gap junctions. BMC Cell Biol. 2016;17(Suppl 1):22.CrossRef Falk MM, Bell CL, Kells Andrews RM, Murray SA. Molecular mechanisms regulating formation, trafficking and processing of annular gap junctions. BMC Cell Biol. 2016;17(Suppl 1):22.CrossRef
35.
go back to reference Gilleron J, Carette D, Chevallier D, Segretain D, Pointis G. Molecular connexin partner remodeling orchestrates connexin traffic: from physiology to pathophysiology. Crit Rev Biochem Mol Biol. 2012;47:407–23.CrossRef Gilleron J, Carette D, Chevallier D, Segretain D, Pointis G. Molecular connexin partner remodeling orchestrates connexin traffic: from physiology to pathophysiology. Crit Rev Biochem Mol Biol. 2012;47:407–23.CrossRef
36.
go back to reference Su V, Lau AF. Connexins: mechanisms regulating protein levels and intercellular communication. FEBS Lett. 2014;588:1212–20.CrossRef Su V, Lau AF. Connexins: mechanisms regulating protein levels and intercellular communication. FEBS Lett. 2014;588:1212–20.CrossRef
37.
go back to reference Omori Y, Li Q, Nishikawa Y, Yoshioka T, Yoshida M, Nishimura T, et al. Pathological significance of intracytoplasmic connexin proteins: implication in tumor progression. J Membr Biol. 2007;218:73–7.CrossRef Omori Y, Li Q, Nishikawa Y, Yoshioka T, Yoshida M, Nishimura T, et al. Pathological significance of intracytoplasmic connexin proteins: implication in tumor progression. J Membr Biol. 2007;218:73–7.CrossRef
38.
go back to reference Mennecier G, Derangeon M, Coronas V, Herve JC, Mesnil M. Aberrant expression and localization of connexin43 and connexin30 in a rat glioma cell line. Mol Carcinog. 2008;47:391–401.CrossRef Mennecier G, Derangeon M, Coronas V, Herve JC, Mesnil M. Aberrant expression and localization of connexin43 and connexin30 in a rat glioma cell line. Mol Carcinog. 2008;47:391–401.CrossRef
39.
go back to reference Ezumi K, Yamamoto H, Murata K, Higashiyama M, Damdinsuren B, Nakamura Y, et al. Aberrant expression of connexin 26 is associated with lung metastasis of colorectal cancer. Clinical Cancer Res. 2008;14:677–84.CrossRef Ezumi K, Yamamoto H, Murata K, Higashiyama M, Damdinsuren B, Nakamura Y, et al. Aberrant expression of connexin 26 is associated with lung metastasis of colorectal cancer. Clinical Cancer Res. 2008;14:677–84.CrossRef
40.
go back to reference Komposch K, Sibilia M. EGFR signaling in liver diseases. Int J Mol Sci. 2016;17(1):30. Komposch K, Sibilia M. EGFR signaling in liver diseases. Int J Mol Sci. 2016;17(1):30.
41.
go back to reference Lusk JB, Lam VY, Tolwinski NS. Epidermal growth factor pathway signaling in Drosophila embryogenesis: tools for understanding Cancer. Cancers. 2017;9(2):16. Lusk JB, Lam VY, Tolwinski NS. Epidermal growth factor pathway signaling in Drosophila embryogenesis: tools for understanding Cancer. Cancers. 2017;9(2):16.
42.
go back to reference Tabernero A, Gangoso E, Jaraiz-Rodriguez M, Medina JM. The role of connexin43-Src interaction in astrocytomas: a molecular puzzle. Neuroscience. 2016;323:183–94.CrossRef Tabernero A, Gangoso E, Jaraiz-Rodriguez M, Medina JM. The role of connexin43-Src interaction in astrocytomas: a molecular puzzle. Neuroscience. 2016;323:183–94.CrossRef
43.
go back to reference Duffy HS, Iacobas I, Hotchkiss K, Hirst-Jensen BJ, Bosco A, Dandachi N, et al. The gap junction protein connexin32 interacts with the Src homology 3/hook domain of discs large homolog 1. J Biol Chem. 2007;282:9789–96.CrossRef Duffy HS, Iacobas I, Hotchkiss K, Hirst-Jensen BJ, Bosco A, Dandachi N, et al. The gap junction protein connexin32 interacts with the Src homology 3/hook domain of discs large homolog 1. J Biol Chem. 2007;282:9789–96.CrossRef
44.
go back to reference Roskoski R Jr. Src protein-tyrosine kinase structure, mechanism, and small molecule inhibitors. Pharmacol Res. 2015;94:9–25.CrossRef Roskoski R Jr. Src protein-tyrosine kinase structure, mechanism, and small molecule inhibitors. Pharmacol Res. 2015;94:9–25.CrossRef
45.
go back to reference Chen Z, Oh D, Dubey AK, Yao M, Yang B, Groves JT, et al. EGFR family and Src family kinase interactions: mechanics matters? Curr Opin Cell Biol. 2018;51:97–102.CrossRef Chen Z, Oh D, Dubey AK, Yao M, Yang B, Groves JT, et al. EGFR family and Src family kinase interactions: mechanics matters? Curr Opin Cell Biol. 2018;51:97–102.CrossRef
46.
go back to reference Sato K, Sato A, Aoto M, Fukami Y. C-Src phosphorylates epidermal growth factor receptor on tyrosine 845. Biochem Biophys Res Commun. 1995;215:1078–87.CrossRef Sato K, Sato A, Aoto M, Fukami Y. C-Src phosphorylates epidermal growth factor receptor on tyrosine 845. Biochem Biophys Res Commun. 1995;215:1078–87.CrossRef
47.
go back to reference Stover DR, Becker M, Liebetanz J, Lydon NB. Src phosphorylation of the epidermal growth factor receptor at novel sites mediates receptor interaction with Src and P85 alpha. J Biol Chem. 1995;270:15591–7.CrossRef Stover DR, Becker M, Liebetanz J, Lydon NB. Src phosphorylation of the epidermal growth factor receptor at novel sites mediates receptor interaction with Src and P85 alpha. J Biol Chem. 1995;270:15591–7.CrossRef
48.
go back to reference Biscardi JS, Maa MC, Tice DA, Cox ME, Leu TH, Parsons SJ. C-Src-mediated phosphorylation of the epidermal growth factor receptor on Tyr845 and Tyr1101 is associated with modulation of receptor function. J Biol Chem. 1999;274:8335–43.CrossRef Biscardi JS, Maa MC, Tice DA, Cox ME, Leu TH, Parsons SJ. C-Src-mediated phosphorylation of the epidermal growth factor receptor on Tyr845 and Tyr1101 is associated with modulation of receptor function. J Biol Chem. 1999;274:8335–43.CrossRef
Metadata
Title
Cx32 exerts anti-apoptotic and pro-tumor effects via the epidermal growth factor receptor pathway in hepatocellular carcinoma
Authors
Yuke Xiang
Qin Wang
Yunquan Guo
Hui Ge
Yile Fu
Xiyan Wang
Liang Tao
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2019
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1142-y

Other articles of this Issue 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 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