Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2024

Open Access 01-12-2024 | Research

Tight junction protein cingulin variant is associated with cancer susceptibility by overexpressed IQGAP1 and Rac1-dependent epithelial-mesenchymal transition

Authors: Yi-Ting Huang, Ya-Ting Hsu, Pei-Ying Wu, Yu-Min Yeh, Peng-Chan Lin, Keng-Fu Hsu, Meng-Ru Shen

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2024

Login to get access

Abstract

Background

Cingulin (CGN) is a pivotal cytoskeletal adaptor protein located at tight junctions. This study investigates the link between CGN mutation and increased cancer susceptibility through genetic and mechanistic analyses and proposes a potential targeted therapeutic approach.

Methods

In a high-cancer-density family without known pathogenic variants, we performed tumor-targeted and germline whole-genome sequencing to identify novel cancer-associated variants. Subsequently, these variants were validated in a 222 cancer patient cohort, and CGN c.3560C > T was identified as a potential cancer-risk allele. Both wild-type (WT) (c.3560C > C) and variant (c.3560C > T) were transfected into cancer cell lines and incorporated into orthotopic xenograft mice model for evaluating their effects on cancer progression. Western blot, immunofluorescence analysis, migration and invasion assays, two-dimensional gel electrophoresis with mass spectrometry, immunoprecipitation assays, and siRNA applications were used to explore the biological consequence of CGN c.3560C > T.

Results

In cancer cell lines and orthotopic animal models, CGN c.3560C > T enhanced tumor progression with reduced sensitivity to oxaliplatin compared to the CGN WT. The variant induced downregulation of epithelial marker, upregulation of mesenchymal marker and transcription factor, which converged to initiate epithelial-mesenchymal transition (EMT). Proteomic analysis was conducted to investigate the elements driving EMT in CGN c.3560C > T. This exploration unveiled overexpression of IQGAP1 induced by the variant, contrasting the levels observed in CGN WT. Immunoprecipitation assay confirmed a direct interaction between CGN and IQGAP1. IQGAP1 functions as a regulator of multiple GTPases, particularly the Rho family. This overexpressed IQGAP1 was consistently associated with the activation of Rac1, as evidenced by the analysis of the cancer cell line and clinical sample harboring CGN c.3560C > T. Notably, activated Rac1 was suppressed following the downregulation of IQGAP1 by siRNA. Treatment with NSC23766, a selective inhibitor for Rac1-GEF interaction, resulted in the inactivation of Rac1. This intervention mitigated the EMT program in cancer cells carrying CGN c.3560C > T. Consistently, xenograft tumors with WT CGN showed no sensitivity to NSC23766 treatment, but NSC23766 demonstrated the capacity to attenuate tumor growth harboring c.3560C > T.

Conclusions

CGN c.3560C > T leads to IQGAP1 overexpression, subsequently triggering Rac1-dependent EMT. Targeting activated Rac1 is a strategy to impede the advancement of cancers carrying this specific variant.
Appendix
Available only for authorised users
Literature
1.
go back to reference Futreal PA, Liu Q, Shattuck-Eidens D, Cochran C, Harshman K, Tavtigian S, et al. BRCA1 mutations in primary breast and ovarian carcinomas. Science. 1994;266:120–2.ADSPubMedCrossRef Futreal PA, Liu Q, Shattuck-Eidens D, Cochran C, Harshman K, Tavtigian S, et al. BRCA1 mutations in primary breast and ovarian carcinomas. Science. 1994;266:120–2.ADSPubMedCrossRef
2.
go back to reference Wooster R, Neuhausen SL, Mangion J, Quirk Y, Ford D, Collins N, et al. Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science. 1994;265:2088–90.ADSPubMedCrossRef Wooster R, Neuhausen SL, Mangion J, Quirk Y, Ford D, Collins N, et al. Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science. 1994;265:2088–90.ADSPubMedCrossRef
3.
go back to reference Friedman LS, Ostermeyer EA, Szabo CI, Dowd P, Lynch ED, Rowell SE, et al. Confirmation of BRCA1 by analysis of germline mutations linked to breast and ovarian cancer in ten families. Nat Genet. 1994;8:399–404.PubMedCrossRef Friedman LS, Ostermeyer EA, Szabo CI, Dowd P, Lynch ED, Rowell SE, et al. Confirmation of BRCA1 by analysis of germline mutations linked to breast and ovarian cancer in ten families. Nat Genet. 1994;8:399–404.PubMedCrossRef
4.
go back to reference Vahteristo P, Bartkova J, Eerola H, Syrjakoski K, Ojala S, Kilpivaara O, et al. A CHEK2 genetic variant contributing to a substantial fraction of familial breast cancer. Am J Hum Genet. 2002;71:432–8.PubMedPubMedCentralCrossRef Vahteristo P, Bartkova J, Eerola H, Syrjakoski K, Ojala S, Kilpivaara O, et al. A CHEK2 genetic variant contributing to a substantial fraction of familial breast cancer. Am J Hum Genet. 2002;71:432–8.PubMedPubMedCentralCrossRef
5.
go back to reference Erkko H, Xia B, Nikkila J, Schleutker J, Syrjakoski K, Mannermaa A, et al. A recurrent mutation in PALB2 in Finnish cancer families. Nature. 2007;446:316–9.ADSPubMedCrossRef Erkko H, Xia B, Nikkila J, Schleutker J, Syrjakoski K, Mannermaa A, et al. A recurrent mutation in PALB2 in Finnish cancer families. Nature. 2007;446:316–9.ADSPubMedCrossRef
6.
go back to reference Heikkinen K, Rapakko K, Karppinen SM, Erkko H, Knuutila S, Lundan T, et al. RAD50 and NBS1 are breast cancer susceptibility genes associated with genomic instability. Carcinogenesis. 2006;27:1593–9.PubMedCrossRef Heikkinen K, Rapakko K, Karppinen SM, Erkko H, Knuutila S, Lundan T, et al. RAD50 and NBS1 are breast cancer susceptibility genes associated with genomic instability. Carcinogenesis. 2006;27:1593–9.PubMedCrossRef
7.
go back to reference Kuchenbaecker KB, Hopper JL, Barnes DR, Phillips KA, Mooij TM, Roos-Blom MJ, et al. Risks of breast, ovarian, and contralateral breast Cancer for BRCA1 and BRCA2 mutation carriers. Jama. 2017;317:2402–16.PubMedCrossRef Kuchenbaecker KB, Hopper JL, Barnes DR, Phillips KA, Mooij TM, Roos-Blom MJ, et al. Risks of breast, ovarian, and contralateral breast Cancer for BRCA1 and BRCA2 mutation carriers. Jama. 2017;317:2402–16.PubMedCrossRef
8.
go back to reference Couch FJ, Shimelis H, Hu C, Hart SN, Polley EC, Na J, et al. Associations between Cancer predisposition testing panel genes and breast Cancer. JAMA Onc. 2017;3:1190–6.CrossRef Couch FJ, Shimelis H, Hu C, Hart SN, Polley EC, Na J, et al. Associations between Cancer predisposition testing panel genes and breast Cancer. JAMA Onc. 2017;3:1190–6.CrossRef
9.
go back to reference Balda MS, Gonzalez-Mariscal L, Matter K, Cereijido M, Anderson JM. Assembly of the tight junction: the role of diacylglycerol. J Cell Biol. 1993;123:293–302.PubMedCrossRef Balda MS, Gonzalez-Mariscal L, Matter K, Cereijido M, Anderson JM. Assembly of the tight junction: the role of diacylglycerol. J Cell Biol. 1993;123:293–302.PubMedCrossRef
10.
go back to reference Zihni C, Mills C, Matter K, Balda MS. Tight junctions: from simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol. 2016;17:564–80.PubMedCrossRef Zihni C, Mills C, Matter K, Balda MS. Tight junctions: from simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol. 2016;17:564–80.PubMedCrossRef
11.
go back to reference Citi S, Paschoud S, Pulimeno P, Timolati F, De Robertis F, Jond L, et al. The tight junction protein cingulin regulates gene expression and RhoA signaling. Ann N Y Acad Sci. 2009;1165:88–98.ADSPubMedCrossRef Citi S, Paschoud S, Pulimeno P, Timolati F, De Robertis F, Jond L, et al. The tight junction protein cingulin regulates gene expression and RhoA signaling. Ann N Y Acad Sci. 2009;1165:88–98.ADSPubMedCrossRef
12.
go back to reference Aijaz S, D'Atri F, Citi S, Balda MS, Matter K. Binding of GEF-H1 to the tight junction-associated adaptor cingulin results in inhibition of rho signaling and G1/S phase transition. Dev Cell. 2005;8:777–86.PubMedCrossRef Aijaz S, D'Atri F, Citi S, Balda MS, Matter K. Binding of GEF-H1 to the tight junction-associated adaptor cingulin results in inhibition of rho signaling and G1/S phase transition. Dev Cell. 2005;8:777–86.PubMedCrossRef
13.
go back to reference Moreno-Bueno G, Portillo F, Cano A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene. 2008;27:6958–69.PubMedCrossRef Moreno-Bueno G, Portillo F, Cano A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene. 2008;27:6958–69.PubMedCrossRef
14.
go back to reference Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442–54.PubMedCrossRef Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442–54.PubMedCrossRef
15.
16.
go back to reference Morris HT, Machesky LM. Actin cytoskeletal control during epithelial to mesenchymal transition: focus on the pancreas and intestinal tract. Br J Cancer. 2015;112:613–20.PubMedPubMedCentralCrossRef Morris HT, Machesky LM. Actin cytoskeletal control during epithelial to mesenchymal transition: focus on the pancreas and intestinal tract. Br J Cancer. 2015;112:613–20.PubMedPubMedCentralCrossRef
19.
go back to reference Noritake J, Watanabe T, Sato K, Wang S, Kaibuchi K. IQGAP1: a key regulator of adhesion and migration. J Cell Sci. 2005;118:2085–92.PubMedCrossRef Noritake J, Watanabe T, Sato K, Wang S, Kaibuchi K. IQGAP1: a key regulator of adhesion and migration. J Cell Sci. 2005;118:2085–92.PubMedCrossRef
20.
go back to reference Owen D, Campbell LJ, Littlefield K, Evetts KA, Li Z, Sacks DB, et al. The IQGAP1-Rac1 and IQGAP1-Cdc42 interactions: interfaces differ between the complexes. J Biol Chem. 2008;283:1692–704.PubMedCrossRef Owen D, Campbell LJ, Littlefield K, Evetts KA, Li Z, Sacks DB, et al. The IQGAP1-Rac1 and IQGAP1-Cdc42 interactions: interfaces differ between the complexes. J Biol Chem. 2008;283:1692–704.PubMedCrossRef
24.
go back to reference Gao Y, Dickerson JB, Guo F, Zheng J, Zheng Y. Rational design and characterization of a Rac GTPase-specific small molecule inhibitor. Proc Natl Acad Sci USA. 2004;101:7618–23.ADSPubMedPubMedCentralCrossRef Gao Y, Dickerson JB, Guo F, Zheng J, Zheng Y. Rational design and characterization of a Rac GTPase-specific small molecule inhibitor. Proc Natl Acad Sci USA. 2004;101:7618–23.ADSPubMedPubMedCentralCrossRef
25.
go back to reference Wu X, Tu X, Joeng KS, Hilton MJ, Williams DA, Long F. Rac1 activation controls nuclear localization of beta-catenin during canonical Wnt signaling. Cell. 2008;133:340–53.PubMedPubMedCentralCrossRef Wu X, Tu X, Joeng KS, Hilton MJ, Williams DA, Long F. Rac1 activation controls nuclear localization of beta-catenin during canonical Wnt signaling. Cell. 2008;133:340–53.PubMedPubMedCentralCrossRef
27.
go back to reference Yoon C, Cho SJ, Chang KK, Park DJ, Ryeom SW, Yoon SS. Role of Rac1 pathway in epithelial-to-mesenchymal transition and Cancer stem-like cell phenotypes in gastric adenocarcinoma. MCR. 2017;15:1106–16.PubMedCrossRef Yoon C, Cho SJ, Chang KK, Park DJ, Ryeom SW, Yoon SS. Role of Rac1 pathway in epithelial-to-mesenchymal transition and Cancer stem-like cell phenotypes in gastric adenocarcinoma. MCR. 2017;15:1106–16.PubMedCrossRef
28.
go back to reference Zhou K, Rao J, Zhou ZH, Yao XH, Wu F, Yang J, et al. RAC1-GTP promotes epithelial-mesenchymal transition and invasion of colorectal cancer by activation of STAT3. Lab Invest. 2018;98:989–98.PubMedCrossRef Zhou K, Rao J, Zhou ZH, Yao XH, Wu F, Yang J, et al. RAC1-GTP promotes epithelial-mesenchymal transition and invasion of colorectal cancer by activation of STAT3. Lab Invest. 2018;98:989–98.PubMedCrossRef
29.
go back to reference Rathinam R, Berrier A, Alahari SK. Role of rho GTPases and their regulators in cancer progression. Front Biosci. 2011;16:2561–71.CrossRef Rathinam R, Berrier A, Alahari SK. Role of rho GTPases and their regulators in cancer progression. Front Biosci. 2011;16:2561–71.CrossRef
30.
go back to reference Gómez del Pulgar T, Benitah SA, Valerón PF, Espina C, Lacal JC. Rho GTPase expression in tumourigenesis: evidence for a significant link. BioEssay. 2005;27:602–13.CrossRef Gómez del Pulgar T, Benitah SA, Valerón PF, Espina C, Lacal JC. Rho GTPase expression in tumourigenesis: evidence for a significant link. BioEssay. 2005;27:602–13.CrossRef
31.
go back to reference Lionarons DA, Hancock DC, Rana S, East P, Moore C, Murillo MM, et al. RAC1(P29S) induces a mesenchymal phenotypic switch via serum response factor to promote melanoma development and therapy resistance. Cancer Cell. 2019;36:68–83.e9.PubMedPubMedCentralCrossRef Lionarons DA, Hancock DC, Rana S, East P, Moore C, Murillo MM, et al. RAC1(P29S) induces a mesenchymal phenotypic switch via serum response factor to promote melanoma development and therapy resistance. Cancer Cell. 2019;36:68–83.e9.PubMedPubMedCentralCrossRef
32.
go back to reference Gómez del Pulgar T, Bandrés E, Espina C, Valdés-Mora F, Pérez-Palacios R, García-Amigot F, et al. Differential expression of Rac1 identifies its target genes and its contribution to progression of colorectal cancer. Int J Biochem Cell Biol. 2007;39:2289–302.PubMedCrossRef Gómez del Pulgar T, Bandrés E, Espina C, Valdés-Mora F, Pérez-Palacios R, García-Amigot F, et al. Differential expression of Rac1 identifies its target genes and its contribution to progression of colorectal cancer. Int J Biochem Cell Biol. 2007;39:2289–302.PubMedCrossRef
33.
go back to reference Gastonguay A, Berg T, Hauser AD, Schuld N, Lorimer E, Williams CL. The role of Rac1 in the regulation of NF-κB activity, cell proliferation, and cell migration in non-small cell lung carcinoma. Cancer Biol Ther. 2012;13:647–56.PubMedPubMedCentralCrossRef Gastonguay A, Berg T, Hauser AD, Schuld N, Lorimer E, Williams CL. The role of Rac1 in the regulation of NF-κB activity, cell proliferation, and cell migration in non-small cell lung carcinoma. Cancer Biol Ther. 2012;13:647–56.PubMedPubMedCentralCrossRef
35.
go back to reference Usatyuk PV, Gorshkova IA, He D, Zhao Y, Kalari SK, Garcia JG, et al. Phospholipase D-mediated activation of IQGAP1 through Rac1 regulates hyperoxia-induced p47phox translocation and reactive oxygen species generation in lung endothelial cells. J Biol Chem. 2009;284:15339–52.PubMedPubMedCentralCrossRef Usatyuk PV, Gorshkova IA, He D, Zhao Y, Kalari SK, Garcia JG, et al. Phospholipase D-mediated activation of IQGAP1 through Rac1 regulates hyperoxia-induced p47phox translocation and reactive oxygen species generation in lung endothelial cells. J Biol Chem. 2009;284:15339–52.PubMedPubMedCentralCrossRef
36.
go back to reference Brown MD, Sacks DB. IQGAP1 in cellular signaling: bridging the GAP. Trends Cell Biol. 2006;16:242–9.PubMedCrossRef Brown MD, Sacks DB. IQGAP1 in cellular signaling: bridging the GAP. Trends Cell Biol. 2006;16:242–9.PubMedCrossRef
37.
go back to reference Jacquemet G, Morgan MR, Byron A, Humphries JD, Choi CK, Chen CS, et al. Rac1 is deactivated at integrin activation sites through an IQGAP1-filamin-A-RacGAP1 pathway. J Cell Sci. 2013;126:4121–35.PubMedPubMedCentral Jacquemet G, Morgan MR, Byron A, Humphries JD, Choi CK, Chen CS, et al. Rac1 is deactivated at integrin activation sites through an IQGAP1-filamin-A-RacGAP1 pathway. J Cell Sci. 2013;126:4121–35.PubMedPubMedCentral
38.
go back to reference Tanos BE, Perez Bay AE, Salvarezza S, Vivanco I, Mellinghoff I, Osman M, et al. IQGAP1 controls tight junction formation through differential regulation of claudin recruitment. J Cell Sci. 2015;128:853–62.PubMedPubMedCentral Tanos BE, Perez Bay AE, Salvarezza S, Vivanco I, Mellinghoff I, Osman M, et al. IQGAP1 controls tight junction formation through differential regulation of claudin recruitment. J Cell Sci. 2015;128:853–62.PubMedPubMedCentral
39.
go back to reference Fukata M, Nakagawa M, Itoh N, Kawajiri A, Yamaga M, Kuroda S, et al. Involvement of IQGAP1, an effector of Rac1 and Cdc42 GTPases, in cell-cell dissociation during cell scattering. Mol Cell Biol. 2001;21:2165–83.PubMedPubMedCentralCrossRef Fukata M, Nakagawa M, Itoh N, Kawajiri A, Yamaga M, Kuroda S, et al. Involvement of IQGAP1, an effector of Rac1 and Cdc42 GTPases, in cell-cell dissociation during cell scattering. Mol Cell Biol. 2001;21:2165–83.PubMedPubMedCentralCrossRef
40.
go back to reference Hage B, Meinel K, Baum I, Giehl K, Menke A. Rac1 activation inhibits E-cadherin-mediated adherens junctions via binding to IQGAP1 in pancreatic carcinoma cells. Cell Signal. 2009;7(1):1–13. Hage B, Meinel K, Baum I, Giehl K, Menke A. Rac1 activation inhibits E-cadherin-mediated adherens junctions via binding to IQGAP1 in pancreatic carcinoma cells. Cell Signal. 2009;7(1):1–13.
41.
go back to reference Dokmanovic M, Hirsch DS, Shen Y, Wu WJ. Rac1 contributes to trastuzumab resistance of breast cancer cells: Rac1 as a potential therapeutic target for the treatment of trastuzumab-resistant breast cancer. Mol Cancer Ther. 2009;8:1557–69.PubMedCrossRef Dokmanovic M, Hirsch DS, Shen Y, Wu WJ. Rac1 contributes to trastuzumab resistance of breast cancer cells: Rac1 as a potential therapeutic target for the treatment of trastuzumab-resistant breast cancer. Mol Cancer Ther. 2009;8:1557–69.PubMedCrossRef
42.
go back to reference Xie JW, Haslam SZ. Extracellular matrix, Rac1 signaling, and estrogen-induced proliferation in MCF-7 breast cancer cells. Breast Cancer Res Treat. 2008;110:257–68.PubMedCrossRef Xie JW, Haslam SZ. Extracellular matrix, Rac1 signaling, and estrogen-induced proliferation in MCF-7 breast cancer cells. Breast Cancer Res Treat. 2008;110:257–68.PubMedCrossRef
43.
44.
go back to reference Nagy R, Sweet K, Eng C. Highly penetrant hereditary cancer syndromes. Oncon. 2004;23:6445–70. Nagy R, Sweet K, Eng C. Highly penetrant hereditary cancer syndromes. Oncon. 2004;23:6445–70.
45.
go back to reference Kurian AW, Hare EE, Mills MA, Kingham KE, McPherson L, Whittemore AS, et al. Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. J Clin Oncol. 2014;32:2001–9.PubMedPubMedCentralCrossRef Kurian AW, Hare EE, Mills MA, Kingham KE, McPherson L, Whittemore AS, et al. Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. J Clin Oncol. 2014;32:2001–9.PubMedPubMedCentralCrossRef
46.
go back to reference Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ, Weissman SM, Yackowski L, et al. Pathogenic and likely pathogenic variant prevalence among the first 10,000 patients referred for next-generation cancer panel testing. Gen Med. 2016;18:823–32. Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ, Weissman SM, Yackowski L, et al. Pathogenic and likely pathogenic variant prevalence among the first 10,000 patients referred for next-generation cancer panel testing. Gen Med. 2016;18:823–32.
47.
go back to reference LaDuca H, Polley EC, Yussuf A, Hoang L, Gutierrez S, Hart SN, et al. A clinical guide to hereditary cancer panel testing: evaluation of gene-specific cancer associations and sensitivity of genetic testing criteria in a cohort of 165,000 high-risk patients. Gen Med. 2020;22:407–15. LaDuca H, Polley EC, Yussuf A, Hoang L, Gutierrez S, Hart SN, et al. A clinical guide to hereditary cancer panel testing: evaluation of gene-specific cancer associations and sensitivity of genetic testing criteria in a cohort of 165,000 high-risk patients. Gen Med. 2020;22:407–15.
Metadata
Title
Tight junction protein cingulin variant is associated with cancer susceptibility by overexpressed IQGAP1 and Rac1-dependent epithelial-mesenchymal transition
Authors
Yi-Ting Huang
Ya-Ting Hsu
Pei-Ying Wu
Yu-Min Yeh
Peng-Chan Lin
Keng-Fu Hsu
Meng-Ru Shen
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2024
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-024-02987-z

Other articles of this Issue 1/2024

Journal of Experimental & Clinical Cancer Research 1/2024 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