1
|
Zhang D, Li L, Ma F. Integrative analyses identified gap junction beta-2 as a prognostic biomarker and therapeutic target for breast cancer. CANCER INNOVATION 2024; 3:e128. [PMID: 38948248 PMCID: PMC11212300 DOI: 10.1002/cai2.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/17/2023] [Accepted: 02/01/2024] [Indexed: 07/02/2024]
Abstract
Background Increasing evidence has shown that connexins are involved in the regulation of tumor development, immune escape, and drug resistance. This study investigated the gene expression patterns, prognostic values, and potential mechanisms of connexins in breast cancer. Methods We conducted a comprehensive analysis of connexins using public gene and protein expression databases and clinical samples from our institution. Connexin mRNA expressions in breast cancer and matched normal tissues were compared, and multiomics studies were performed. Results Gap junction beta-2 mRNA was overexpressed in breast cancers of different pathological types and molecular subtypes, and its high expression was associated with poor prognosis. The tumor membrane of the gap junction beta-2 mutated group was positive, and the corresponding protein was expressed. Somatic mutation and copy number variation of gap junction beta-2 are rare in breast cancer. The gap junction beta-2 transcription level in the p110α subunit of the phosphoinositide 3-kinase mutant subgroup was higher than that in the wild-type subgroup. Gap junction beta-2 was associated with the phosphoinositide 3-kinase-Akt signaling pathway, extracellular matrix-receptor interaction, focal adhesion, and proteoglycans in cancer. Furthermore, gap junction beta-2 overexpression may be associated with phosphoinositide 3-kinase and histone deacetylase inhibitor resistance, and its expression level correlated with infiltrating CD8+ T cells, macrophages, neutrophils, and dendritic cells. Conclusions Gap junction beta-2 may be a promising therapeutic target for targeted therapy and immunotherapy and may be used to predict breast cancer prognosis.
Collapse
Affiliation(s)
- Di Zhang
- Department of Medical OncologyQilu Hospital of Shandong UniversityJinanChina
- Department of Medical OncologyQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lixi Li
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fei Ma
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Xu H, Wang X, Zhu F, Guo S, Chao Z, Cao C, Lu Z, Zhu H, Wang M, Zhu F, Yang J, Zeng R, Yao Y. Comprehensive Pan-Cancer Analysis of Connexin 43 as a Potential Biomarker and Therapeutic Target in Human Kidney Renal Clear Cell Carcinoma (KIRC). MEDICINA (KAUNAS, LITHUANIA) 2024; 60:780. [PMID: 38792963 PMCID: PMC11123162 DOI: 10.3390/medicina60050780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: Connexin 43 (Cx43) is involved in the transfer of small signaling molecules between neighboring cells, thereby exerting a major influence on the initiation and progression of tumorigenesis. However, there is a lack of systematic research on Cx43 expression and its predictive role in clinical diagnosis and prognosis in pan-cancer. Materials and Methods: Several biological databases were used to evaluate the expression levels of GJA1 (encoding Cx43) and its diagnostic and prognostic significance in pan-cancer. We targeted kidney renal clear cell carcinoma (KIRC) and investigated the relationship between GJA1 expression and different clinical features of KIRC patients. Then, we performed cell-based experiments to partially confirm our results and predicted several proteins that were functionally related to Cx43. Results: The expression of GJA1 has a high level of accuracy in predicting KIRC. High GJA1 expression was remarkably correlated with a favorable prognosis, and this expression was reduced in groups with poor clinical features in KIRC. Cell experiments confirmed the inhibitory effects of increased GJA1 expression on the migratory capacity of human renal cancer (RCC) cell lines, and protein-protein interaction (PPI) analysis predicted that CDH1 and CTNNB1 were closely related to Cx43. Conclusions: GJA1 could be a promising independent favorable prognostic factor for KIRC, and upregulation of GJA1 expression could inhibit the migratory capacity of renal cancer cells.
Collapse
Affiliation(s)
- Huzi Xu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Xiuru Wang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Fan Zhu
- Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Shuiming Guo
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Zheng Chao
- Division of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China;
| | - Chujin Cao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Zhihui Lu
- Division of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China;
| | - Han Zhu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Meng Wang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Fengming Zhu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Juan Yang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Rui Zeng
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
| | - Ying Yao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; (H.X.); (X.W.); (S.G.); (C.C.); (H.Z.); (M.W.); (F.Z.); (J.Y.)
- Division of Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| |
Collapse
|
3
|
Provera MD, Straign DM, Karimpour P, Ihle CL, Owens P. Bone morphogenetic protein pathway responses and alterations of osteogenesis in metastatic prostate cancers. Cancer Rep (Hoboken) 2023; 6:e1707. [PMID: 36054271 PMCID: PMC9940003 DOI: 10.1002/cnr2.1707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/07/2022] [Accepted: 07/27/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Prostate cancer is a common cancer in men that annually results in more than 33 000 US deaths. Mortality from prostate cancer is largely from metastatic disease, reflecting on the great strides in the last century of treatments in care for the localized disease. Metastatic castrate resistant prostate cancer (mCRPC) will commonly travel to the bone, creating unique bone pathology that requires nuanced treatments in those sites with surgical, radio and chemotherapeutic interventions. The bone morphogenetic protein (BMP) pathway has been historically studied in the capacity to regulate the osteogenic nature of new bone. New mineralized bone generation is a frequent and common observation in mCRPC and referred to as blastic bone lesions. Less common are bone destructive lesions that are termed lytic. METHODS We queried the cancer genome atlas (TCGA) prostate cancer databases for the expression of the BMP pathway and found that distinct gene expression of the ligands, soluble antagonists, receptors, and intracellular mediators were altered in localized versus metastatic disease. Human prostate cancer cell lines have an innate ability to promote blastic- or lytic-like bone lesions and we hypothesized that inhibiting BMP signaling in these cell lines would result in a distinct change in osteogenesis gene expression with BMP inhibition. RESULTS We found unique and common changes by comparing these cell lines response and unique BMP pathway alterations. We treated human PCa cell lines with distinct bone pathologic phenotypes with the BMP inhibitor DMH1 and found distinct osteogenesis responses. We analyzed distinct sites of metastatic PCa in the TCGA and found that BMP signaling was selectively altered in commons sites such as lymph node, bone and liver compared to primary tumors. CONCLUSIONS Overall we conclude that BMPs in metastatic prostate cancer are important signals and functional mediators of diverse processes that have potential for individualized precision oncology in mCRPC.
Collapse
Affiliation(s)
- Meredith D. Provera
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | - Desiree M. Straign
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | | | - Claire L. Ihle
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | - Philip Owens
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
- Department of Veterans Affairs, Research Service, Eastern Colorado Health Care SystemAuroraColoradoUSA
| |
Collapse
|
4
|
Qi J, Yin J, Ding G. A Connexin-Based Biomarker Model Applicable for Prognosis and Immune Landscape Assessment in Lung Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:9261339. [PMID: 36276289 PMCID: PMC9581606 DOI: 10.1155/2022/9261339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 12/03/2022]
Abstract
Purpose Gap junction protein (Connexin) family is the basic unit of cellular connection, whose multiple members were recently demonstrated to be associated with tumor progression. However, the expression pattern and prognostic value of connexin in lung adenocarcinoma (LUAD) have not yet been elucidated. Methods Consensus cluster algorithm was first applied to determine a novel molecular subtype in LUAD based on connexin genes. The differentially expressed genes (DEGs) between two clusters were obtained to include in Cox regression analyses for the model construction. To examine the predictive capacity of the signature, survival curves and ROC plots were conducted. We implemented GSEA method to uncover the function effects enriched in the risk model. Moreover, the tumor immune microenvironment in LUAD was depicted by CIBERSORT and ssGSEA methods. Results The integrated LUAD cohort (TCGA-LUAD and GSE68465) were clustered into two subtypes (C1 = 217 and C2 = 296) based on 21 connexins and the clinical outcomes of LUAD cases in the two clusters showed remarkable discrepancy. Next, we collected 222 DEGs among two subclusters to build a prognostic model using stepwise Cox analyses. Our proposed model consisted of six genes that accurately forecast patient outcomes and differentiate patient risk. GSEA indicated that high-risk group was involved in tumor relevant pathways were activated in high-risk group, such as PI3K/AKT signaling, TGF-β pathway, and p53 pathway. Furthermore, LUAD cases with high-risk presented higher infiltration level of M2 macrophage and neutrophil, suggesting high-risk group were more likely to generate an immunosuppressive status. Conclusion Our data identified a novel connexin-based subcluster in LUAD and further created a risk signature which plays a central part in prognosis assessment and clinical potency.
Collapse
Affiliation(s)
- Junqing Qi
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jun Yin
- Department of Cardiothoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Guowen Ding
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
5
|
Jones JC, Bodenstine TM. Connexins and Glucose Metabolism in Cancer. Int J Mol Sci 2022; 23:ijms231710172. [PMID: 36077565 PMCID: PMC9455984 DOI: 10.3390/ijms231710172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Connexins are a family of transmembrane proteins that regulate diverse cellular functions. Originally characterized for their ability to mediate direct intercellular communication through the formation of highly regulated membrane channels, their functions have been extended to the exchange of molecules with the extracellular environment, and the ability to modulate numerous channel-independent effects on processes such as motility and survival. Notably, connexins have been implicated in cancer biology for their context-dependent roles that can both promote or suppress cancer cell function. Moreover, connexins are able to mediate many aspects of cellular metabolism including the intercellular coupling of nutrients and signaling molecules. During cancer progression, changes to substrate utilization occur to support energy production and biomass accumulation. This results in metabolic plasticity that promotes cell survival and proliferation, and can impact therapeutic resistance. Significant progress has been made in our understanding of connexin and cancer biology, however, delineating the roles these multi-faceted proteins play in metabolic adaptation of cancer cells is just beginning. Glucose represents a major carbon substrate for energy production, nucleotide synthesis, carbohydrate modifications and generation of biosynthetic intermediates. While cancer cells often exhibit a dependence on glycolytic metabolism for survival, cellular reprogramming of metabolic pathways is common when blood perfusion is limited in growing tumors. These metabolic changes drive aggressive phenotypes through the acquisition of functional traits. Connections between glucose metabolism and connexin function in cancer cells and the surrounding stroma are now apparent, however much remains to be discovered regarding these relationships. This review discusses the existing evidence in this area and highlights directions for continued investigation.
Collapse
|
6
|
Lv Z, Wang X, Zhu C, Wang K. The global status of research in prostate cancer bone metastasis: A bibliometric and visualized analysis. Front Med (Lausanne) 2022; 9:931422. [PMID: 35991630 PMCID: PMC9381755 DOI: 10.3389/fmed.2022.931422] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
Background Prostate cancer (PCa) is a serious threat to the health of elderly aged groups. It is very important to understand the occurrence and development of PCa for early diagnosis, treatment and metastasis control. This study aims to elucidate the international frontier research direction and literature distribution through bibliometric and visual analyses of PCa bone metastasis. Methods Data were obtained from the Web of Science core collection database, which collected 2,246 papers related to PCa bone metastasis from 1 January 2012 to 31 December 2021. The collected data were analyzed using the VOSviewer software for citation, co-authorship, co-citation, bibliometric coupling, and co-occurrence. Results Over the past decade, published papers have increased annually. The United States of America has published 890 papers with 29,161 citations, far more than any other country, and it has the most extensive collaboration with other countries. For example, 33 articles by Saad Fred were cited 2,721 times, and 91 articles from the University of Texas MD Anderson CANC CTR were cited 3,037 times, the most cited author and organization. Peng Xinsheng and Duke UNIV comprise the most active collaborative author and organization, respectively. The most co-cited journal was CANCER RES, with 3,195 citations. Studies of PCa bone metastasis can be divided into four categories: “basic research,” “auxiliary diagnosis and treatment,” “clinical trial,” and “prognosis.” Conclusion Our results provide a comprehensive overview of the research priorities and future directions of PCa bone metastasis, which can further accurately guide researchers in diagnosis, treatment, and personalized prevention.
Collapse
Affiliation(s)
- Zongwei Lv
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chunming Zhu
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Chunming Zhu,
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
- Kefeng Wang,
| |
Collapse
|
7
|
Valiunas V, Gordon C, Valiuniene L, Devine D, Lin RZ, Cohen IS, Brink PR. Intercellular delivery of therapeutic oligonucleotides. J Drug Deliv Sci Technol 2022; 72:103404. [PMID: 36721641 PMCID: PMC9886232 DOI: 10.1016/j.jddst.2022.103404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
One promising approach to cancer therapeutics is to induce changes in gene expression that either reduce cancer cell proliferation or induce cancer cell death. Therefore, delivering oligonucleotides (siRNA/miRNA) that target specific genes or gene programs might have a potential therapeutic benefit. The aim of this study was to examine the potential of cell-based delivery of oligonucleotides to cancer cells via two naturally occurring intercellular pathways: gap junctions and vesicular/exosomal traffic. We utilized human mesenchymal stem cells (hMSCs) as delivery cells and chose to deliver in vitro two synthetic oligonucleotides, AllStars HS Cell Death siRNA and miR-16 mimic, as toxic (therapeutic) oligonucleotides targeting three cancer cell lines: prostate (PC3), pancreatic (PANC1) and cervical (HeLa). Both oligonucleotides dramatically reduced cell proliferation and/or induced cell death when transfected directly into target cells and delivery hMSCs. The delivery and target cells we chose express gap junction connexin 43 (Cx43) endogenously (PC3, PANC1, hMSC) or via stable transfection (HeLaCx43). Co-culture of hMSCs (transfected with either toxic oligonucleotide) with any of Cx43 expressing cancer cells induced target cell death (~20% surviving) or senescence (~85% proliferation reduction) over 96 hours. We eliminated gap junction-mediated delivery by using connexin deficient HeLaWT cells or knocking out endogenous Cx43 in PANC1 and PC3 cells via CRISPR/Cas9. Subsequently, all Cx43 deficient target cells co-cultured with the same toxic oligonucleotide loaded hMSCs proliferated, albeit at significantly slower rates, with cell number increasing on average ~2.2-fold (30% of control cells) over 96 hours. Our results show that both gap junction and vesicular/exosomal intercellular delivery pathways from hMSCs to target cancer cells deliver oligonucleotides and function to either induce cell death or significantly reduce their proliferation. Thus, hMSC-based cellular delivery is an effective method of delivering synthetic oligonucleotides that can significantly reduce tumor cell growth and should be further investigated as a possible approach to cancer therapy.
Collapse
Affiliation(s)
- Virginijus Valiunas
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Chris Gordon
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Laima Valiuniene
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Daniel Devine
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Richard Z Lin
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Ira S Cohen
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Peter R Brink
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology Stony Brook University, Stony Brook, NY 11794-8661, USA
| |
Collapse
|
8
|
Maliekal TT, Dharmapal D, Sengupta S. Tubulin Isotypes: Emerging Roles in Defining Cancer Stem Cell Niche. Front Immunol 2022; 13:876278. [PMID: 35693789 PMCID: PMC9179084 DOI: 10.3389/fimmu.2022.876278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Although the role of microtubule dynamics in cancer progression is well-established, the roles of tubulin isotypes, their cargos and their specific function in the induction and sustenance of cancer stem cells (CSCs) were poorly explored. But emerging reports urge to focus on the transport function of tubulin isotypes in defining orchestrated expression of functionally critical molecules in establishing a stem cell niche, which is the key for CSC regulation. In this review, we summarize the role of specific tubulin isotypes in the transport of functional molecules that regulate metabolic reprogramming, which leads to the induction of CSCs and immune evasion. Recently, the surface expression of GLUT1 and GRP78 as well as voltage-dependent anion channel (VDAC) permeability, regulated by specific isotypes of β-tubulins have been shown to impart CSC properties to cancer cells, by implementing a metabolic reprogramming. Moreover, βIVb tubulin is shown to be critical in modulating EphrinB1signaling to sustain CSCs in oral carcinoma. These tubulin-interacting molecules, Ephrins, GLUT1 and GRP78, are also important regulators of immune evasion, by evoking PD-L1 mediated T-cell suppression. Thus, the recent advances in the field implicate that tubulins play a role in the controlled transport of molecules involved in CSC niche. The indication of tubulin isotypes in the regulation of CSCs offers a strategy to specifically target those tubulin isotypes to eliminate CSCs, rather than the general inhibition of microtubules, which usually leads to therapy resistance.
Collapse
Affiliation(s)
- Tessy Thomas Maliekal
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Regional Centre for Biotechnology, Faridabad, India
- *Correspondence: Tessy Thomas Maliekal, ; Suparna Sengupta,
| | - Dhrishya Dharmapal
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- University of Kerala, Department of Biotechnology, Thiruvananthapuram, India
| | - Suparna Sengupta
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Regional Centre for Biotechnology, Faridabad, India
- University of Kerala, Department of Biotechnology, Thiruvananthapuram, India
- *Correspondence: Tessy Thomas Maliekal, ; Suparna Sengupta,
| |
Collapse
|
9
|
Niu G, Zhang X, Hong R, Yang X, Gu J, Song T, Hu Z, Chen L, Wang X, Xia J, Ke Z, Ren J, Hong L. GJA1 promotes hepatocellular carcinoma progression by mediating TGF-β-induced activation and the epithelial-mesenchymal transition of hepatic stellate cells. Open Med (Wars) 2021; 16:1459-1471. [PMID: 34693020 PMCID: PMC8486017 DOI: 10.1515/med-2021-0344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 08/12/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
Introduction Gap junction protein, alpha 1 (GJA1), which is correlated with recurrences and unfavorable prognoses in hepatocellular carcinomas (HCCs), is one of the specific proteins expressed by activated hepatic stellate cells (HSCs). Methods Expression of GJA1 was compared between HCCs and nontumor tissues (NTs), between hepatic cirrhosis and NTs, and between primary and metastatic HCCs using transcriptomic datasets from the Gene Expression Omnibus and the Integrative Molecular Database of Hepatocellular Carcinoma. The in vitro activities of GJA1 were investigated in cultured HSCs and HCC cells. The underlying mechanism was characterized using Gene Set Enrichment Analysis and validated by western blotting. Results The expression of GJA1 was significantly increased in HCCs and hepatic cirrhosis compared to that in NTs. GJA1 was also overexpressed in pulmonary metastases from HCCs when compared with HCCs without metastasis. Overexpression of GJA1 promoted while knockdown of GJA1 inhibited proliferation and transforming growth factor (TGF)-β-mediated activation and migration of cultured HSCs. Overexpression of GJA1 by lentivirus infection promoted proliferation and migration, while conditioned medium from HSCs overexpressing GJA1 promoted migration but inhibited proliferation of Hep3B and PLC-PRF-5 cells. Lentivirus infection with shGJA1 or conditioned medium from shGJA1-infected HSCs inhibited the proliferation and migration of HCCLM3 cells that had a high propensity toward lung metastasis. Mechanistically, GJA1 induced the epithelial–mesenchymal transition (EMT) in HSCs and HCCLM3 cells. Conclusion GJA1 promoted HCC progression by inducing HSC activation and the EMT in HSCs. GJA1 is potentially regulated by TGF-β and thus may be a therapeutic target to inhibit HCC progression.
Collapse
Affiliation(s)
- Gengming Niu
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Xiaotian Zhang
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Runqi Hong
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Ximin Yang
- Department of Radiology, Dongying New District Hospital, Dongying, Shandong Province, 257000, People's Republic of China
| | - Jiawei Gu
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Tao Song
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Zhiqing Hu
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Liang Chen
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Xin Wang
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Jie Xia
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, Minhang District, Shanghai, 200240, People's Republic of China
| | - Zhongwei Ke
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Jun Ren
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Liang Hong
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| |
Collapse
|
10
|
Glucose-limiting conditions induce an invasive population of MDA-MB-231 breast cancer cells with increased connexin 43 expression and membrane localization. J Cell Commun Signal 2021; 15:223-236. [PMID: 33591483 PMCID: PMC7991056 DOI: 10.1007/s12079-020-00601-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 12/09/2020] [Indexed: 01/10/2023] Open
Abstract
Gap junctional intercellular communication (GJIC) is a homeostatic process mediated by membrane channels composed of a protein family known as connexins. Alterations to channel activity can modulate suppression or facilitation of cancer progression. These varying roles are influenced by the cancer cell genetic profile and the context-dependent mechanisms of a dynamic extracellular environment that encompasses fluctuations to nutrient availability. To better explore the effects of altered cellular metabolism on GJIC in breast cancer, we generated a derivative of the triple-negative breast cancer cell line MDA-MB-231 optimized for growth in low-glucose. Reduced availability of glucose is commonly encountered during tumor development and leads to metabolic reprogramming in cancer cells. MDA-MB-231 low-glucose adapted cells exhibited a larger size with improved cell–cell contact and upregulation of cadherin-11. Additionally, increased protein levels of connexin 43 and greater plasma membrane localization were observed with a corresponding improvement in GJIC activity compared to the parental cell line. Since GJIC has been shown to affect cellular invasion in multiple cancer cell types, we evaluated the invasive qualities of these cells using multiple three-dimensional Matrigel growth models. Results of these experiments demonstrated a significantly more invasive phenotype. Moreover, a decrease in invasion was noted when GJIC was inhibited. Our results indicate a potential response of triple-negative breast cancer cells to reduced glucose availability that results in changes to GJIC and invasiveness. Delineation of this relationship may help elucidate mechanisms by which altered cancer cell metabolism affects GJIC and how cancer cells respond to nutrient availability in this regard.
Collapse
|
11
|
Connexins and cAMP Cross-Talk in Cancer Progression and Metastasis. Cancers (Basel) 2020; 13:cancers13010058. [PMID: 33379194 PMCID: PMC7795795 DOI: 10.3390/cancers13010058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Different connexins play diverse roles in cancers, either tumor-suppressing or tumor-promoting. In lung cancer, Cx43 serves as a tumor suppressor at the early stage, but it can also be a tumor-promotor at an advanced stage and during metastasis. Moreover, other connexins, including Cx26, Cx31.1, and Cx32, can be tumor suppressors. In contrast, Cx30.3 can be a tumor-promotor. The roles of different connexins in different cancers have also been established. Cx43 acts as a tumor suppressor in colorectal cancer, breast cancer, and glioma, whereas Cx32 can be a suppressor in liver tumors and hepatocarcinogenesis. Cx26 can be a tumor suppressor in mammary tumors; in contrast, it can be a tumor-promotor in melanoma. Existing drugs/molecules targeting the cAMP/PKA/connexin axis act to regulate channel opening/closing. Mimic peptides, such as Gap19, Gap26, and Gap 27 block hemichannels, mimetic peptides, and CT9/CT10 and promote hemichannel opening and also hemichannel closing. Abstract Connexin-containing gap junctions mediate the direct exchange of small molecules between cells, thus promoting cell–cell communication. Connexins (Cxs) have been widely studied as key tumor-suppressors. However, certain Cx subtypes, such as Cx43 and Cx26, are overexpressed in metastatic tumor lesions. Cyclic adenosine monophosphate (cAMP) signaling regulates Cx expression and function via transcriptional control and phosphorylation. cAMP also passes through gap junction channels between adjacent cells, regulating cell cycle progression, particularly in cancer cell populations. Low levels of cAMP are sufficient to activate key effectors. The present review evaluates the mechanisms underlying Cx regulation by cAMP signaling and the role of gap junctions in cancer progression and metastasis. A deeper understanding of these processes might facilitate the development of novel anticancer drugs.
Collapse
|
12
|
Clézardin P, Coleman R, Puppo M, Ottewell P, Bonnelye E, Paycha F, Confavreux CB, Holen I. Bone metastasis: mechanisms, therapies, and biomarkers. Physiol Rev 2020; 101:797-855. [PMID: 33356915 DOI: 10.1152/physrev.00012.2019] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Skeletal metastases are frequent complications of many cancers, causing bone complications (fractures, bone pain, disability) that negatively affect the patient's quality of life. Here, we first discuss the burden of skeletal complications in cancer bone metastasis. We then describe the pathophysiology of bone metastasis. Bone metastasis is a multistage process: long before the development of clinically detectable metastases, circulating tumor cells settle and enter a dormant state in normal vascular and endosteal niches present in the bone marrow, which provide immediate attachment and shelter, and only become active years later as they proliferate and alter the functions of bone-resorbing (osteoclasts) and bone-forming (osteoblasts) cells, promoting skeletal destruction. The molecular mechanisms involved in mediating each of these steps are described, and we also explain how tumor cells interact with a myriad of interconnected cell populations in the bone marrow, including a rich vascular network, immune cells, adipocytes, and nerves. We discuss metabolic programs that tumor cells could engage with to specifically grow in bone. We also describe the progress and future directions of existing bone-targeted agents and report emerging therapies that have arisen from recent advances in our understanding of the pathophysiology of bone metastases. Finally, we discuss the value of bone turnover biomarkers in detection and monitoring of progression and therapeutic effects in patients with bone metastasis.
Collapse
Affiliation(s)
- Philippe Clézardin
- INSERM, Research Unit UMR_S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, Lyon, France.,Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Rob Coleman
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Margherita Puppo
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Penelope Ottewell
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Edith Bonnelye
- INSERM, Research Unit UMR_S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, Lyon, France
| | - Frédéric Paycha
- Service de Médecine Nucléaire, Hôpital Lariboisière, Paris, France
| | - Cyrille B Confavreux
- INSERM, Research Unit UMR_S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, Lyon, France.,Service de Rhumatologie Sud, CEMOS-Centre Expert des Métastases Osseuses, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Ingunn Holen
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
13
|
Mulkearns-Hubert EE, Reizes O, Lathia JD. Connexins in Cancer: Jekyll or Hyde? Biomolecules 2020; 10:E1654. [PMID: 33321749 PMCID: PMC7764653 DOI: 10.3390/biom10121654] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022] Open
Abstract
The expression, localization, and function of connexins, the protein subunits that comprise gap junctions, are often altered in cancer. In addition to cell-cell coupling through gap junction channels, connexins also form hemichannels that allow communication between the cell and the extracellular space and perform non-junctional intracellular activities. Historically, connexins have been considered tumor suppressors; however, they can also serve tumor-promoting functions in some contexts. Here, we review the literature surrounding connexins in cancer cells in terms of specific connexin functions and propose that connexins function upstream of most, if not all, of the hallmarks of cancer. The development of advanced connexin targeting approaches remains an opportunity for the field to further interrogate the role of connexins in cancer phenotypes, particularly through the use of in vivo models. More specific modulators of connexin function will both help elucidate the functions of connexins in cancer and advance connexin-specific therapies in the clinic.
Collapse
Affiliation(s)
- Erin E. Mulkearns-Hubert
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (O.R.); (J.D.L.)
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (O.R.); (J.D.L.)
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College, Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Justin D. Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (O.R.); (J.D.L.)
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College, Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, 44195, USA
| |
Collapse
|
14
|
Curtis KJ, Schiavi J, Mc Garrigle MJ, Kumar V, McNamara LM, Niebur GL. Mechanical stimuli and matrix properties modulate cancer spheroid growth in three-dimensional gelatin culture. J R Soc Interface 2020; 17:20200568. [PMID: 33323051 PMCID: PMC7811591 DOI: 10.1098/rsif.2020.0568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Most patients who succumb to cancer have metastases to bone that contribute to their death. Cancer cells that metastasize to bone are regularly subjected to mechanical stimuli that may affect their proliferation, growth and protein expression. Understanding why some cancer cells thrive in this environment could provide insight into new approaches to prevent or treat metastasis to bone. We used 4T1 cells as a model of breast cancer cells, and implanted them in gelatin hydrogels with moduli of 1 or 2.7 kPa to mimic the properties of bone marrow. The constructs were subjected to either perfusion of media through the hydrogel or combined perfusion and cyclic mechanical compression for 1 h d-1 for 4 d. Controls were cultured in free-swelling conditions. The cells formed spheroids during the 4 d of culture, with larger spheroids in the statically cultured constructs than in perfusion or compressed constructs. In stiffer gelatin, smaller spheroids formed in compressed constructs than perfusion alone, while compression had no effect compared to perfusion in the softer gelatin. Immunostaining indicated that the spheroids expressed osteopontin, parathyroid hormone-related protein and fibronectin, which are all hallmarks of bone metastasis. The proliferative marker Ki67 was present in all spheroids on day 4. In the 1 kPa gelatin, Ki67 staining intensity was greater in the statically cultured, free-swelling constructs than in bioreactor culture, regardless of dynamic compression. By contrast, proliferation was higher in the compressed gelatins compared to perfusion alone in the 2.7 kPa constructs, although the spheroids were smaller, on average. This suggests the stiffer gelatin may restrict spheroid growth at the same time that it enhances mechanobiological signalling during compression. Taken together, 4T1 breast cancer cells are mechanically sensitive, and mechanical stimuli can alter their proliferation and protein expression within soft materials with mechanical properties similar to bone marrow. As such, both in vivo and in vitro models of cancer metastasis should consider the role of the mechanical environment in the bone.
Collapse
Affiliation(s)
- Kimberly J. Curtis
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, Harper Cancer Research Institute, University of Notre Dame, IN 46556, USA
| | - Jessica Schiavi
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Myles J. Mc Garrigle
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Vatsal Kumar
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Laoise M. McNamara
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Glen L. Niebur
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, Harper Cancer Research Institute, University of Notre Dame, IN 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, IN 46556, USA
| |
Collapse
|
15
|
Connexins-Therapeutic Targets in Cancers. Int J Mol Sci 2020; 21:ijms21239119. [PMID: 33266154 PMCID: PMC7730856 DOI: 10.3390/ijms21239119] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Connexins (Cx) are members of a protein family that forms intercellular channels localised in gap junction (GJ) plaques and single transmembrane channels called hemichannels. They participate in intercellular communication or communication between the intracellular and extracellular environments. Connexins affect cell homeostasis, growth and differentiation by enabling the exchange of metabolites or by interfering with various signalling pathways. Alterations in the functionality and the expression of connexins have been linked to the occurrence of many diseases. Connexins have been already linked to cancers, cardiac and brain disorders, chronic lung and kidney conditions and wound healing processes. Connexins have been shown either to suppress cancer tumour growth or to increase tumorigenicity by promoting cancer cell growth, migration and invasiveness. A better understanding of the complexity of cancer biology related to connexins and intercellular communication could result in the design of novel therapeutic strategies. The modulation of connexin expression may be an effective therapeutic approach in some types of cancers. Therefore, one important challenge is the search for mechanisms and new drugs, selectively modulating the expression of various connexin isoforms. We performed a systematic literature search up to February 2020 in the electronic databases PubMed and EMBASE. Our search terms were as follows: connexins, hemichannels, cancer and cancer treatment. This review aims to provide information about the role of connexins and gap junctions in cancer, as well as to discuss possible therapeutic options that are currently being studied.
Collapse
|
16
|
MYC as a Multifaceted Regulator of Tumor Microenvironment Leading to Metastasis. Int J Mol Sci 2020; 21:ijms21207710. [PMID: 33081056 PMCID: PMC7589112 DOI: 10.3390/ijms21207710] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
The Myc family of oncogenes is deregulated in many types of cancer, and their over-expression is often correlated with poor prognosis. The Myc family members are transcription factors that can coordinate the expression of thousands of genes. Among them, c-Myc (MYC) is the gene most strongly associated with cancer, and it is the focus of this review. It regulates the expression of genes involved in cell proliferation, growth, differentiation, self-renewal, survival, metabolism, protein synthesis, and apoptosis. More recently, novel studies have shown that MYC plays a role not only in tumor initiation and growth but also has a broader spectrum of functions in tumor progression. MYC contributes to angiogenesis, immune evasion, invasion, and migration, which all lead to distant metastasis. Moreover, MYC is able to promote tumor growth and aggressiveness by recruiting stromal and tumor-infiltrating cells. In this review, we will dissect all of these novel functions and their involvement in the crosstalk between tumor and host, which have demonstrated that MYC is undoubtedly the master regulator of the tumor microenvironment. In sum, a better understanding of MYC’s role in the tumor microenvironment and metastasis development is crucial in proposing novel and effective cancer treatment strategies.
Collapse
|
17
|
Boucher J, Balandre AC, Debant M, Vix J, Harnois T, Bourmeyster N, Péraudeau E, Chépied A, Clarhaut J, Debiais F, Monvoisin A, Cronier L. Cx43 Present at the Leading Edge Membrane Governs Promigratory Effects of Osteoblast-Conditioned Medium on Human Prostate Cancer Cells in the Context of Bone Metastasis. Cancers (Basel) 2020; 12:cancers12103013. [PMID: 33081404 PMCID: PMC7602984 DOI: 10.3390/cancers12103013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In its late stages, prostate cancer (PCa) is characterized by a high propensity to form osteoblastic bone metastases, mainly treated by palliative approaches. In a previous work, we demonstrated that a gap junctional protein, connexin43 (Cx43) is implicated both in the increase of aggressiveness of PCa cells and in their impact on bone. To analyze the reciprocal part of the dialogue, the current study addresses the role of Cx43 in the impact of bone microenvironment on PCa cells abilities. Using Cx43-overexpressing PCa cell lines, we determined that Cx43 is necessary for promigratory effect induced by osteoblastic conditioned media (ObCM) on individual cells. Next, we demonstrated the requirement of Cx43 membrane localization at the leading edge and the involvement of the cytoplasmic part in this ObCM-induced migration. Overall, our findings precise the role of Cx43 during PCa progression and its putative use as aggressiveness marker and as potential therapeutic targets. Abstract Among the different interacting molecules implicated in bone metastases, connexin43 (Cx43) may increase sensitivity of prostate cancer (PCa) cells to bone microenvironment, as suggested by our in silico and human tissue samples analyses that revealed increased level of Cx43 expression with PCa progression and a Cx43 specific expression in bone secondary sites. The goal of the present study was to understand how Cx43 influences PCa cells sensitivity and aggressiveness to bone microenvironment. By means of Cx43-overexpressing PCa cell lines, we revealed a Cx43-dependent promigratory effect of osteoblastic conditioned media (ObCM). This effect on directional migration relied on the presence of Cx43 at the plasma membrane and not on gap junctional intercellular communication and hemichannel functions. ObCM stimulation induced Rac1 activation and Cx43 interaction with cortactin in protrusions of migrating PCa cells. Finally, by transfecting two different truncated forms of Cx43 in LNCaP cells, we determined that the carboxy terminal (CT) part of Cx43 is crucial for the responsiveness of PCa cells to ObCM. Our study demonstrates that Cx43 level and its membrane localization modulate the phenotypic response of PCa cells to osteoblastic microenvironment and that its CT domain plays a pivotal role.
Collapse
Affiliation(s)
- Jonathan Boucher
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Annie-Claire Balandre
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Marjolaine Debant
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Justine Vix
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
- Department of Rheumatology, University Hospital Center of Poitiers, 2 Rue de la Milétrie, 86021 Poitiers, France
| | - Thomas Harnois
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Nicolas Bourmeyster
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Elodie Péraudeau
- University Hospital Center of Poitiers, 2 rue de la Milétrie, 86021 Poitiers, France; (E.P.); (J.C.)
- CNRS UMR 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), University of Poitiers, 4 Rue Michel Brunet, TSA 51106, CEDEX 09, 86073 Poitiers, France
| | - Amandine Chépied
- Laboratory of Experimental and Clinical Neurosciences, LNEC-INSERM U1084, UBM-Laboratoire de Cancérologie Biologique, CHU de Poitiers, 2 Rue de la Milétrie, 86000 Poitiers, France;
| | - Jonathan Clarhaut
- University Hospital Center of Poitiers, 2 rue de la Milétrie, 86021 Poitiers, France; (E.P.); (J.C.)
- CNRS UMR 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), University of Poitiers, 4 Rue Michel Brunet, TSA 51106, CEDEX 09, 86073 Poitiers, France
| | - Françoise Debiais
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
- Department of Rheumatology, University Hospital Center of Poitiers, 2 Rue de la Milétrie, 86021 Poitiers, France
| | - Arnaud Monvoisin
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
| | - Laurent Cronier
- CNRS ERL7003, Laboratory Signalisation et Transports Ioniques Membranaires (STIM), University of Poitiers, 1 rue Georges Bonnet, TSA 51106, CEDEX 09, 86073 Poitiers, France; (J.B.); (A.-C.B.); (M.D.); (J.V.); (T.H.); (N.B.); (F.D.); (A.M.)
- Correspondence: ; Tel.: +33-5-49-45-37-52
| |
Collapse
|
18
|
Antagonistic Functions of Connexin 43 during the Development of Primary or Secondary Bone Tumors. Biomolecules 2020; 10:biom10091240. [PMID: 32859065 PMCID: PMC7565206 DOI: 10.3390/biom10091240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
Despite research and clinical advances during recent decades, bone cancers remain a leading cause of death worldwide. There is a low survival rate for patients with primary bone tumors such as osteosarcoma and Ewing’s sarcoma or secondary bone tumors such as bone metastases from prostate carcinoma. Gap junctions are specialized plasma membrane structures consisting of transmembrane channels that directly link the cytoplasm of adjacent cells, thereby enabling the direct exchange of small signaling molecules between cells. Discoveries of human genetic disorders due to genetic mutations in gap junction proteins (connexins) and experimental data using connexin knockout mice have provided significant evidence that gap-junctional intercellular communication (Gj) is crucial for tissue function. Thus, the dysfunction of Gj may be responsible for the development of some diseases. Gj is thus a main mechanism for tumor cells to communicate with other tumor cells and their surrounding microenvironment to survive and proliferate. If it is well accepted that a low level of connexin expression favors cancer cell proliferation and therefore primary tumor development, more evidence is suggesting that a high level of connexin expression stimulates various cellular process such as intravasation, extravasation, or migration of metastatic cells. If so, connexin expression would facilitate secondary tumor dissemination. This paper discusses evidence that suggests that connexin 43 plays an antagonistic role in the development of primary bone tumors as a tumor suppressor and secondary bone tumors as a tumor promoter.
Collapse
|
19
|
Direct Intercellular Communications and Cancer: A Snapshot of the Biological Roles of Connexins in Prostate Cancer. Cancers (Basel) 2019; 11:cancers11091370. [PMID: 31540089 PMCID: PMC6770088 DOI: 10.3390/cancers11091370] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/04/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Tissue homeostasis is the result of a complex intercellular network controlling the behavior of every cell for the survival of the whole organism. In mammalian tissues, cells do communicate via diverse long- and short-range communication mechanisms. While long-range communication involves hormones through blood circulation and neural transmission, short-range communication mechanisms include either paracrine diffusible factors or direct interactions (e.g., gap junctions, intercellular bridges and tunneling nanotubes) or a mixture of both (e.g., exosomes). Tumor growth represents an alteration of tissue homeostasis and could be the consequence of intercellular network disruption. In this network, direct short-range intercellular communication seems to be particularly involved. The first type of these intercellular communications thought to be involved in cancer progression were gap junctions and their protein subunits, the connexins. From these studies came the general assumption that global decreased connexin expression is correlated to tumor progression and increased cell proliferation. However, this assumption appeared more complicated by the fact that connexins may act also as pro-tumorigenic. Then, the concept that direct intercellular communication could be involved in cancer has been expanded to include new forms of intercellular communication such as tunneling nanotubes (TNTs) and exosomes. TNTs are intercellular bridges that allow free exchange of small molecules or even mitochondria depending on the presence of gap junctions. The majority of current research shows that such exchanges promote cancer progression by increasing resistance to hypoxia and chemotherapy. If exosomes are also involved in these mechanisms, more studies are needed to understand their precise role. Prostate cancer (PCa) represents a type of malignancy with one of the highest incidence rates worldwide. The precise role of these types of direct short-range intercellular communication has been considered in the progression of PCa. However, even though data are in favor of connexins playing a key role in PCa progression, a clear understanding of the role of TNTs and exosomes is needed to define their precise role in this malignancy. This review article summarizes the current view of the main mechanisms involved in short-range intercellular communication and their implications in cancer and delves into the biological, predictive and therapeutic role of connexins in PCa.
Collapse
|
20
|
Wu JI, Wang LH. Emerging roles of gap junction proteins connexins in cancer metastasis, chemoresistance and clinical application. J Biomed Sci 2019; 26:8. [PMID: 30642339 PMCID: PMC6332853 DOI: 10.1186/s12929-019-0497-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Connexin, a four-pass transmembrane protein, contributes to assembly of gap junctions among neighboring cells and thus facilitates gap junctional intercellular communication (GJIC). Traditionally, the roles of connexins were thought to mediate formation of hemichannels and GJIC assembly for transportation of ions and small molecules. Many studies have observed loss of GJIC, due to reduced expression or altered cytoplasmic localization of connexins, in primary tumor cells. Connexins are generally considered tumor-suppressive. However, recent studies of clinical samples suggested a different role of connexins in that expression levels and membrane localization of connexins, including Connexin 43 (Cx43, GJA1) and Connexin 26 (Cx26, GJB2), were found to be enhanced in metastatic lesions of cancer patients. Cx43- and Cx26-mediated GJIC was found to promote cancer cell migration and adhesion to the pulmonary endothelium. Regulatory circuits involved in the induction of connexins and their functional effects have also been reported in various types of cancer. Connexins expressed in stromal cells were correlated with metastasis and were implicated in regulating metastatic behaviors of cancer cells. Recent studies have revealed that connexins can contribute to cellular phenotypes via multiple ways, namely 1) GJIC, 2) C-terminal tail-mediated signaling, and 3) cell-cell adhesion during gap junction formation. Both expression levels and the subcellular localization could participate determining the functional roles of connexins in cancer. Compounds targeting connexins were thus tested as potential therapeutics intervening metastasis or chemoresistance. This review focuses on the recent findings in the correlation between the expression of connexins and patients’ prognosis, their roles in metastasis and chemoresistance, as well as the implications and concerns of using connexin-targeting drugs as anti-metastatic therapeutics. Overall, connexins may serve as biomarkers for cancer prognosis and as therapeutic targets for intervening metastasis and chemoresistance.
Collapse
Affiliation(s)
- Jun-I Wu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Lu-Hai Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan. .,Department of Life Sciences, National Central University, Taoyuan, Taiwan. .,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan. .,Chinese Medical Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
21
|
Wang Q, Zhou C, Li X, Cai L, Zou J, Zhang D, Xie J, Lai W. TGF-β1 promotes gap junctions formation in chondrocytes via Smad3/Smad4 signalling. Cell Prolif 2018; 52:e12544. [PMID: 30444057 PMCID: PMC6495951 DOI: 10.1111/cpr.12544] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/04/2018] [Accepted: 09/21/2018] [Indexed: 02/05/2023] Open
Abstract
Objectives Connexin‐mediated functional gap junction intercellular communication (GJIC) has a vital role in development, homeostasis and pathology. Transforming growth factor‐β1 (TGF‐β1), as one of the most vital factors in chondrocytes, promotes cartilage precursor cell differentiation and chondrocyte proliferation, migration and metabolism. However, how TGF‐β1 mediates GJIC in chondrocytes remains unclear. This study aims to determine the influence of TGF‐β1 on GJIC in mouse chondrocytes and its underlying mechanism. Methods qPCR and mRNA microarray were used to verify the expression of genes in the TGF‐β and connexin families in cartilage and chondrocytes. A scrape loading/dye transfer assay was performed to explore GJIC. Western blot analysis was used to detect connexin43 (Cx43) and Smad signalling components. Immunofluorescence staining was performed to characterize protein distribution. Results The TGF‐β1 mRNA was the highest expressed member of the TGFβ super family in cartilage. TGF‐β1 promoted functional GJIC through increased expression of Cx43. TGF‐β1‐mediated GJIC required the participation of TGF‐β type I receptor. TGF‐β1 activated Smad3 and Smad4 signalling to facilitate their nuclear translocation. The Smad3 and Smad4 signalling proteins bound to the promoter of Gja1 and thus initiated Cx43 gene expression. Conclusions For the first time, these results revealed a vital role of TGF‐β1 in cell‐cell communication in chondrocytes via gap junction formation. We describe the regulatory mechanism, the involvement of TGF‐β type I receptor and the nuclear translocation of Smad3/4.
Collapse
Affiliation(s)
- Qingxuan Wang
- State Key Laboratory of Oral Diseases, Orthodontics Department, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, Orthodontics Department, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaobing Li
- State Key Laboratory of Oral Diseases, Orthodontics Department, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, Orthodontics Department, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Zou
- State Key Laboratory of Oral Diseases, Orthodontics Department, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, Orthodontics Department, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, Orthodontics Department, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenli Lai
- State Key Laboratory of Oral Diseases, Orthodontics Department, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Connexins and Pannexins: Important Players in Tumorigenesis, Metastasis and Potential Therapeutics. Int J Mol Sci 2018; 19:ijms19061645. [PMID: 29865195 PMCID: PMC6032133 DOI: 10.3390/ijms19061645] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
Since their characterization more than five decades ago, gap junctions and their structural proteins-the connexins-have been associated with cancer cell growth. During that period, the accumulation of data and molecular knowledge about this association revealed an apparent contradictory relationship between them and cancer. It appeared that if gap junctions or connexins can down regulate cancer cell growth they can be also implied in the migration, invasion and metastatic dissemination of cancer cells. Interestingly, in all these situations, connexins seem to be involved through various mechanisms in which they can act either as gap-junctional intercellular communication mediators, modulators of signalling pathways through their interactome, or as hemichannels, which mediate autocrine/paracrine communication. This complex involvement of connexins in cancer progression is even more complicated by the fact that their hemichannel function may overlap with other gap junction-related proteins, the pannexins. Despite this complexity, the possible involvements of connexins and pannexins in cancer progression and the elucidation of the mechanisms they control may lead to use them as new targets to control cancer progression. In this review, the involvements of connexins and pannexins in these different topics (cancer cell growth, invasion/metastasis process, possible cancer therapeutic targets) are discussed.
Collapse
|
23
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
24
|
Lai Y, Fan L, Zhao Y, Ge H, Feng X, Wang Q, Zhang X, Peng Y, Wang X, Tao L. Cx32 suppresses extrinsic apoptosis in human cervical cancer cells via the NF‑κB signalling pathway. Int J Oncol 2017; 51:1159-1168. [PMID: 28902345 DOI: 10.3892/ijo.2017.4106] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/28/2017] [Indexed: 01/22/2023] Open
Abstract
Tumour necrosis factor α (TNFα) and TNF‑related apoptosis inducing ligand (TRAIL) usually trigger either survival or apoptosis signals in various cell types, and nuclear factor κB (NF‑κB) is a key factor that regulates their biological effects. Connexin 32 (Cx32) is a gap junction (GJ) protein that plays vital roles in tumourigenesis and tumour progression. Our previous study explored abnormal Cx32 expression in para‑nuclear areas, exacerbated prognostic parameters and suppressed streptonigrin/cisplatin-induced apoptosis in human cervical cancer (CaCx) cells. In this study, we investigated the role of Cx32 in the extrinsic apoptosis pathway of CaCx cells. In transgenic HeLa cells and C-33A cells, Cx32 expression was manipulated using doxycycline or Cx32 siRNA. GJ inhibitors or low density culturing was used to change the status of gap junction intracellular communication (GJIC). We found that apoptosis induced by TNFα and TRAIL was suppressed by Cx32 expression despite the presence or absense of GJIC. We also found that Cx32 upregulated the expression of nuclear NF‑κB and its downstream targets c-IAP1, MMP‑2, and MMP‑9 in HeLa‑Cx32 and C-33A cells. Following our previous study design, our clinical data showed that NF‑κB and MMP‑2 levels increased in human CaCx specimens with high Cx32 expression compared to levels in para‑carcinoma of cervical specimens. SC75741 and JSH-23, NF‑кB signalling pathway inhibitors, inhibited the anti-apoptotic effects of Cx32. In conclusion, Cx32 suppressed TNFα /TRAIL-induced extrinsic apoptosis by upregulating the NF‑κB signalling pathway. This study demonstrates a novel mechanism for Cx32's anti-apoptotic effect and provides a reasonable explanation for the pro-tumour effect of Cx32 in human CaCx cells.
Collapse
Affiliation(s)
- Yongchang Lai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Lixia Fan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yifan Zhao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Hui Ge
- Tumor Research Institute, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Xue Feng
- Tumor Research Institute, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaomin Zhang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yuexia Peng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiyan Wang
- Tumor Research Institute, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Liang Tao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
25
|
Boucher J, Monvoisin A, Vix J, Mesnil M, Thuringer D, Debiais F, Cronier L. Connexins, important players in the dissemination of prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:202-215. [PMID: 28693897 DOI: 10.1016/j.bbamem.2017.06.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/22/2017] [Accepted: 06/29/2017] [Indexed: 12/25/2022]
Abstract
Over the past 50years, increasing experimental evidences have established that connexins (Cxs) and gap junctional intercellular communication (GJIC) ensure an important role in both the onset and development of cancerous processes. In the present review, we focus on the impact of Cxs and GJIC during the development of prostate cancer (PCa), from the primary growth mainly localized in acinar glands and ducts to the distant metastasis mainly concentrated in bone. As observed in several other types of solid tumours, Cxs and especially Cx43 exhibit an ambivalent role with a tumour suppressor effect in the early stages and, conversely, a rather pro-tumoural profile for most of invasion and dissemination steps to secondary sites. We report here the current knowledge on the function of Cxs during PCa cells migration, cytoskeletal dynamics, proteinases activities and the cross talk with the surrounding stromal cells in the microenvironment of the tumour and the bones. In addition, we discuss the role of Cxs in the bone tropism even if the prostate model is rarely used to study the complete sequence of cancer dissemination compared to breast cancer or melanoma. Even if not yet fully understood, these recent findings on Cxs provide new insights into their molecular mechanisms associated with progression and bone targeted behaviour of PCa. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Jonathan Boucher
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | - Arnaud Monvoisin
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | - Justine Vix
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France; Department of Rheumatology, C.H.U. la Milétrie, Poitiers, France
| | - Marc Mesnil
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | | | - Françoise Debiais
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France; Department of Rheumatology, C.H.U. la Milétrie, Poitiers, France
| | - Laurent Cronier
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France.
| |
Collapse
|
26
|
Mesnil M, Aasen T, Boucher J, Chépied A, Cronier L, Defamie N, Kameritsch P, Laird DW, Lampe PD, Lathia JD, Leithe E, Mehta PP, Monvoisin A, Pogoda K, Sin WC, Tabernero A, Yamasaki H, Yeh ES, Dagli MLZ, Naus CC. An update on minding the gap in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:237-243. [PMID: 28655619 DOI: 10.1016/j.bbamem.2017.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 01/08/2023]
Abstract
This article is a report of the "International Colloquium on Gap junctions: 50Years of Impact on Cancer" that was held 8-9 September 2016, at the Amphitheater "Pôle Biologie Santé" of the University of Poitiers (Poitiers, France). The colloquium was organized by M Mesnil (Université de Poitiers, Poitiers, France) and C Naus (University of British Columbia, Vancouver, Canada) to celebrate the 50th anniversary of the seminal work published in 1966 by Loewenstein and Kanno [Intercellular communication and the control of tissue growth: lack of communication between cancer cells, Nature, 116 (1966) 1248-1249] which initiated studies on the involvement of gap junctions in carcinogenesis. During the colloquium, 15 participants presented reviews or research updates in the field which are summarized below.
Collapse
Affiliation(s)
- Marc Mesnil
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France.
| | - Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, 08035 Barcelona, Spain
| | - Jonathan Boucher
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Amandine Chépied
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Laurent Cronier
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Norah Defamie
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Petra Kameritsch
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario N6A 5C1, Canada
| | - Paul D Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Justin D Lathia
- Cleveland Clinic, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, and Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, NO-0424 Oslo, Norway
| | - Parmender P Mehta
- Department of Biochemistry and Molecular Biology, University of Nebraska, Medical Center, Omaha, NE 68198, USA
| | - Arnaud Monvoisin
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Kristin Pogoda
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany
| | - Wun-Chey Sin
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Arantxa Tabernero
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca 37007, Spain
| | | | - Elizabeth S Yeh
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29412, USA
| | - Maria Lucia Zaidan Dagli
- Laboratory of Experimental and Comparative Oncology, School of Veterinary Medicine and Animal Science of the University of São Paulo, São Paulo, SP CEP 05508-900, Brazil
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
27
|
Zhao Y, Lai Y, Ge H, Guo Y, Feng X, Song J, Wang Q, Fan L, Peng Y, Cao M, Harris AL, Wang X, Tao L. 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. [PMID: 28492539 PMCID: PMC5520707 DOI: 10.1038/cddis.2017.183] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022]
Abstract
The role of connexin proteins (Cx), which form gap junctions (GJ), in progression and chemotherapeutic sensitivity of cervical cancer (CaCx), is unclear. Using cervix specimens (313 CaCx, 78 controls) and CaCx cell lines, we explored relationships among Cx expression, prognostic variables and mechanisms that may link them. In CaCx specimens, Cx32 was upregulated and cytoplasmically localized, and three other Cx downregulated, relative to controls. Cx32 expression correlated with advanced FIGO staging, differentiation and increased tumor size. In CaCx cell lines, Cx32 expression suppressed streptonigrin/cisplatin-induced apoptosis in the absence of functional GJ. In CaCx specimens and cell lines, expression of Cx32 upregulated epidermal growth factor receptor (EGFR) expression. Inhibition of EGFR signaling abrogated the anti-apoptotic effect of Cx32 expression. In conclusion, upregulated Cx32 in CaCx cells produces anti-apoptotic, pro-tumorigenic effects in vivo and vitro. Abnormal Cx32 expression/localization in CaCx appears to be both a mechanism and biomarker of chemotherapeutic resistance.
Collapse
Affiliation(s)
- Yifan Zhao
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
- Department of Anesthesiology, Sun Yat-Sen
Memorial Hospital, Sun Yat-Sen University, Guangzhou
510120, China
| | - Yongchang Lai
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| | - Hui Ge
- Tumor Research Institute, Xinjiang
Medical University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Yunquan Guo
- Department of Pathology, Xinjiang Medical
University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Xue Feng
- Tumor Research Institute, Xinjiang
Medical University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Jia Song
- Tumor Research Institute, Xinjiang
Medical University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Qin Wang
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| | - Lixia Fan
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| | - Yuexia Peng
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| | - Minghui Cao
- Department of Anesthesiology, Sun Yat-Sen
Memorial Hospital, Sun Yat-Sen University, Guangzhou
510120, China
| | - Andrew L Harris
- Department of Pharmacology, Physiology
and Neuroscience, New Jersey Medical School - Rutgers University,
Newark, NJ
07103, USA
| | - Xiyan Wang
- Tumor Research Institute, Xinjiang
Medical University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Liang Tao
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| |
Collapse
|
28
|
Piwowarczyk K, Paw M, Ryszawy D, Rutkowska-Zapała M, Madeja Z, Siedlar M, Czyż J. Connexin43 high prostate cancer cells induce endothelial connexin43 up-regulation through the activation of intercellular ERK1/2-dependent signaling axis. Eur J Cell Biol 2017; 96:337-346. [PMID: 28396058 DOI: 10.1016/j.ejcb.2017.03.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/24/2017] [Accepted: 03/31/2017] [Indexed: 01/04/2023] Open
Abstract
Connexin(Cx)43 regulates the invasive potential of prostate cancer cells and participates in their extravasation. To address the role of endothelial Cx43 in this process, we analyzed Cx43 regulation in human umbilical vein endothelial cells in the proximity of Cx43high (DU-145 and MAT-LyLu) and Cx43low prostate cancer cells (PC-3 and AT-2). Endothelial Cx43 up-regulation was observed during the diapedesis of DU-145 and MAT-LyLu cells. This process was attenuated by transient Cx43 silencing in cancer cells and by chemical inhibition of ERK1/2-dependent signaling in endothelial cells. Cx43 expression in endothelial cells was insensitive to the inhibition of gap junctional intercellular coupling between Cx43high prostate cancer and endothelial cells by 18α-glycyrrhetinic acid. Instead, endothelial Cx43 up-regulation was correlated with the local contraction of endothelial cells and with their activation in the proximity of Cx43high DU-145 and MAT-LyLu cells. It was also sensitive to pro-inflammatory factors secreted by peripheral blood monocytes, such as TNFα. In contrast to Cx43low AT-2 cells, Cx43low PC-3 cells produced angioactive factors that locally activated the endothelial cells in the absence of endothelial Cx43 up-regulation. Collectively, these data show that Cx43low and Cx43high prostate cancer cells can adapt discrete, Cx43-independent and Cx43-dependent strategies of diapedesis. Our observations identify a novel strategy of prostate cancer cell diapedesis, which depends on the activation of intercellular Cx43/ERK1/2/Cx43 signaling axis at the interfaces between Cx43high prostate cancer and endothelial cells.
Collapse
Affiliation(s)
- Katarzyna Piwowarczyk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Milena Paw
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Damian Ryszawy
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Magdalena Rutkowska-Zapała
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Jarosław Czyż
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
29
|
Alteration of osteoblast arrangement via direct attack by cancer cells: New insights into bone metastasis. Sci Rep 2017; 7:44824. [PMID: 28303941 PMCID: PMC5356003 DOI: 10.1038/srep44824] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/15/2017] [Indexed: 11/09/2022] Open
Abstract
Intact bone tissue exhibits a characteristic anisotropic microstructure derived from collagen fiber alignment and the related c-axis orientation of apatite crystals, which govern the mechanical properties of bone tissue. In contrast, tumor-invaded bone exhibits a disorganized, less-aligned microstructure that results in severely disrupted mechanical function. Despite its importance both in basic principle and in therapeutic applications, the classical understanding of bone metastasis is limited to alterations in bone mass regulated by metastatic cancer cells. In this study, we demonstrate a novel mechanism underlying the disruption of bone tissue anisotropy in metastasized bone. We observed that direct attack by cancer cells on osteoblasts induces the less-organized osteoblast arrangement. Importantly, the crystallographic anisotropy of bone tissue is quantitatively determined by the level of osteoblast arrangement. Osteoblast arrangement was significantly disrupted by physical contact with cancer cells such as osteolytic melanoma B16F10, breast cancer MDA-MB-231, and osteoblastic prostate cancer MDA-PCa-2b cells. The present findings demonstrate that the abnormal arrangement of osteoblasts induced by physical contact with cancer cells facilitates the disorganized microstructure of metastasized bone.
Collapse
|
30
|
Abstract
Fifty years ago, tumour cells were found to lack electrical coupling, leading to the hypothesis that loss of direct intercellular communication is commonly associated with cancer onset and progression. Subsequent studies linked this phenomenon to gap junctions composed of connexin proteins. Although many studies support the notion that connexins are tumour suppressors, recent evidence suggests that, in some tumour types, they may facilitate specific stages of tumour progression through both junctional and non-junctional signalling pathways. This Timeline article highlights the milestones connecting gap junctions to cancer, and underscores important unanswered questions, controversies and therapeutic opportunities in the field.
Collapse
Affiliation(s)
- Trond Aasen
- (Co-corresponding authors) Correspondence to
T.A. () and D.W.L.
()
| | - Marc Mesnil
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences
Fondamentales et Appliquées, Université de Poitiers, Poitiers,
France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, The Life
Sciences Institute, University of British Columbia, Vancouver, British
Columbia, Canada
| | - Paul D. Lampe
- Translational Research Program, Fred Hutchinson Cancer Research
Center, Seattle, United States
| | - Dale W. Laird
- (Co-corresponding authors) Correspondence to
T.A. () and D.W.L.
()
| |
Collapse
|
31
|
Hamon L, Savarin P, Pastré D. Polyamine signal through gap junctions: A key regulator of proliferation and gap-junction organization in mammalian tissues? Bioessays 2016; 38:498-507. [DOI: 10.1002/bies.201500195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Loic Hamon
- Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques; INSERM U1204 and Université Evry-Val d'Essonne; Evry France
| | - Philippe Savarin
- Centre National de Recherche Scientifique (CNRS), Equipe Spectroscopie des Biomolécules et des Milieux Biologiques (SBMB); Université Paris 13, Sorbonne Paris Cité, Laboratoire Chimie, Structures, Propriétés de Biomatériaux et d'Agents Thérapeutiques (CSPBAT), Unité Mixte de Recherche (UMR) 7244; Bobigny France
| | - David Pastré
- Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques; INSERM U1204 and Université Evry-Val d'Essonne; Evry France
| |
Collapse
|
32
|
Oncogenic extracellular HSP70 disrupts the gap-junctional coupling between capillary cells. Oncotarget 2016; 6:10267-83. [PMID: 25868858 PMCID: PMC4496354 DOI: 10.18632/oncotarget.3522] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/17/2015] [Indexed: 12/31/2022] Open
Abstract
High levels of circulating heat shock protein 70 (HSP70) are detected in many cancers. In order to explore the effects of extracellular HSP70 on human microvascular endothelial cells (HMEC), we initially used gap-FRAP technique. Extracellular human HSP70 (rhHSP70), but not rhHSP27, blocks the gap-junction intercellular communication (GJIC) between HMEC, disrupts the structural integrity of HMEC junction plaques, and decreases connexin43 (Cx43) expression, which correlates with the phosphorylation of Cx43 serine residues. Further exploration of these effects identified a rapid transactivation of the Epidermal Growth Factor Receptor in a Toll-Like Receptor 4-dependent manner, preceding its internalization. In turn, cytosolic Ca2+ oscillations are generated. Both GJIC blockade and Ca2+ mobilization partially depend on ATP release through Cx43 and pannexin (Panx-1) channels, as demonstrated by blocking activity or expression of channels, and inactivating extracellular ATP. By monitoring dye-spreading into adjacent cells, we show that HSP70 released from human monocytes in response to macrophage colony-stimulating factor, prevents the formation of GJIC between monocytes and HMEC. Therapeutic manipulation of this pathway could be of interest in inflammatory and tumor growth.
Collapse
|
33
|
Connexin 43 expression is associated with increased malignancy in prostate cancer cell lines and functions to promote migration. Oncotarget 2016; 6:11640-51. [PMID: 25960544 PMCID: PMC4484482 DOI: 10.18632/oncotarget.3449] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/24/2015] [Indexed: 12/13/2022] Open
Abstract
Impaired expression of connexins, the gap junction subunits that facilitate direct cell-cell communication, have been implicated in prostate cancer growth. To elucidate the crucial role of connexins in prostate cancer progression, we performed a systematic quantitative RT-PCR screening of connexin expression in four representative prostate cancer cell lines across the spectrum of malignancy. Transcripts of several connexin subunits were detected in all four cell lines, and connexin 43 (Cx43) showed marked elevation at both RNA and protein levels in cells with increased metastatic potential. Analysis of gap-junction-mediated intercellular communication revealed homocellular coupling in PC-3 cells, which had the highest Cx43 expression, with minimal coupling in LNCaP cells where Cx43 expression was very low. Treatment with the gap junction inhibitor carbenoxolone or connexin mimetic peptide ACT-1 did not impair cell growth, suggesting that growth is independent of functional gap junctions. PC-3 cells with Cx43 expression reduced by shRNA showed decreased migration in monolayer wound healing assay, as well as decreased transwell invasion capacities when compared to control cells expressing non-targeting shRNA. These results, together with the correlation between Cx43 expression levels and the metastatic capacity of the cell lines, suggest a role of Cx43 in prostate cancer invasion and metastasis.
Collapse
|
34
|
Puzzo L, Caltabiano R, Parenti R, Trapasso S, Allegra E. Connexin 43 (Cx43) Expression in Laryngeal Squamous Cell Carcinomas: Preliminary Data on Its Possible Prognostic Role. Head Neck Pathol 2016; 10:292-7. [PMID: 26748803 PMCID: PMC4972757 DOI: 10.1007/s12105-016-0685-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 01/03/2016] [Indexed: 12/31/2022]
Abstract
The aim of the report is to evaluate the prognostic and predictive role of Connexin 43 (Cx43) expression in laryngeal squamous cell carcinomas. Eighty-seven previously untreated patients submitted to laryngectomy ± neck dissection ± radiotherapy were enrolled in this retrospective study. The original primary tumor slides were reassessed, tumor grade and stage reviewed, and Cx43 immunohistochemical analysis performed: only cytoplasmic membranous staining of Cx43 has been shown. Neither significant correlation has been showed for clinical T (p = 0.75) and N (p = 0.81), while significant correlation has been found with grading (p < 0.0001) and pathological N (p < 0.0001). Five year overall survival (OS) of the 87 patients was 54 %; 5 year OS was 59.6 % in Cx43 positive patients and 37.1 % in Cx43 negative patients, but also this difference did not reach statistical significance (p = 0.058). Our best findings were: poorly differentiated carcinomas had low or negative Cx43 expression; moderately differentiated tumors without node metastasis and no radiotherapy but with Cx43 expression had a better outcome; moderately differentiated tumors without node metastasis and no radiotherapy but without Cx43 expression had a worse outcome; moderately differentiated tumors with node metastasis and radiotherapy but without Cx43 expression had a better outcome. Interestingly, in G2 head and neck squamous cell carcinomas with lymph node metastasis at the time of diagnosis, Cx43 aberrant overexpression could identify a subset of patients with poor prognosis, far less responsive to radio/chemotherapy.
Collapse
Affiliation(s)
- Lidia Puzzo
- Department “G.F.Ingrassia”, Section of Anatomic Pathology, University of Catania, Via S. Sofia, 87, 95125 Catania, Italy
| | - Rosario Caltabiano
- Department “G.F.Ingrassia”, Section of Anatomic Pathology, University of Catania, Via S. Sofia, 87, 95125 Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Via S. Sofia, 64, 95125 Catania, Italy
| | - Serena Trapasso
- Department of Medical and Surgical Sciences – Section of Otolaryngology, Magna Graecia University of Catanzaro, Viale Europa, Località Germaneto, 88100 Catanzaro, Italy
| | - Eugenia Allegra
- Department of Medical and Surgical Sciences – Section of Otolaryngology, Magna Graecia University of Catanzaro, Viale Europa, Località Germaneto, 88100 Catanzaro, Italy
| |
Collapse
|
35
|
Upregulation of connexin43 contributes to PX-12-induced oxidative cell death. Tumour Biol 2015; 37:7535-46. [PMID: 26684802 DOI: 10.1007/s13277-015-4620-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/10/2015] [Indexed: 01/24/2023] Open
Abstract
Thioredoxin (Trx) is a small redox protein that underlies aggressive tumor growth and resistance to chemotherapy. Inhibition of Trx with the chemical inhibitor PX-12 suppresses tumor growth and induces cell apoptosis. Currently, the mechanism underlying the therapeutic actions of PX-12 and the molecules influencing cell susceptibility to PX-12 are incompletely understood. Given that connexin43 (Cx43), a tumor suppressor, regulates tumor cell susceptibility to chemotherapy, we examined the possible involvement of Cx43 in PX-12-induced cell death. Exposure of cells to PX-12 led to a loss of cell viability, which was associated with the activation of oxidative sensitive c-Jun N-terminal kinase (JNK). Inhibition of JNK or supplement of cells with anti-oxidants prevented the cell-killing action of PX-12. The forced expression of Cx43 in normal and tumor cells increased cell sensitivity to PX-12-induced JNK activation and cell death. In contrast, the downregulation of Cx43 with siRNA or the suppression of gap junctions with chemical inhibitors attenuated JNK activation and enhanced cell resistance to PX-12. Further analysis revealed that PX-12 at low concentrations induced a JNK-dependent elevation in the Cx43 protein, which was also preventable by supplementing the cells with anti-oxidants. Our results thus indicate that Cx43 is a determinant in the regulation of cell susceptibility to PX-12 and that the upregulation of Cx43 may be an additional mechanism by which PX-12 exerts its anti-tumor actions.
Collapse
|
36
|
Liu Y, Wen Q, Chen XL, Yang SJ, Gao L, Gao L, Zhang C, Li JL, Xiang XX, Wan K, Chen XH, Zhang X, Zhong JF. All-trans retinoic acid arrests cell cycle in leukemic bone marrow stromal cells by increasing intercellular communication through connexin 43-mediated gap junction. J Hematol Oncol 2015; 8:110. [PMID: 26446715 PMCID: PMC4597383 DOI: 10.1186/s13045-015-0212-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/28/2015] [Indexed: 01/28/2023] Open
Abstract
Background Gap junctional intercellular communication (GJIC) is typically decreased in malignant tumors. Gap junction is not presented between hematopoietic cells but occurred in bone marrow stromal cells (BMSCs). Connexin 43 (Cx43) is the major gap junction (GJ) protein; our previous study revealed that Cx43 expression and GJIC were decreased in acute leukemic BMSCs. All-trans retinoic acid (ATRA) increases GJIC in a variety of cancer cells and has been used to treat acute promyelocytic leukemia, but the effects of ATRA on leukemic BMSCs is unknown. In this study, we evaluated the potential effects of ATRA on cell cycle, proliferation, and apoptosis of leukemic BMSCs. Effects of ATRA on Cx43 expression and GJIC were also examined. Methods Human BMSCs obtained from 25 patients with primary acute leukemia, and 10 normal healthy donors were cultured. Effects of ATRA on cell cycle, cell proliferation, and apoptosis were examined with or without co-treatment with amphotericin-B. Cx43 expression was examined at both the mRNA and protein expression levels. GJIC was examined by using a dye transfer assay and measuring the rate of fluorescence recovery after photobleaching (FRAP). Results ATRA arrested the cell cycle progression, inhibited cell growth, and increased apoptosis in leukemic BMSCs. Both Cx43 expression and GJIC function were increased by ATRA treatment. Most of the observed effects mediated by ATRA were abolished by amphotericin-B pretreatment. Conclusions ATRA arrests cell cycle progression in leukemic BMSCs, likely due to upregulating Cx43 expression and enhancing GJIC function. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0212-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yao Liu
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China. .,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - Qin Wen
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Xue-Lian Chen
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - Shi-Jie Yang
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Lei Gao
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Li Gao
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Cheng Zhang
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Jia-Li Li
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Xi-Xi Xiang
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Kai Wan
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Xing-Hua Chen
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, the Third Military Medical University, Xinqiao Street, Chongqing, 400037, China.
| | - Jiang-Fan Zhong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
37
|
Thuringer D, Berthenet K, Cronier L, Solary E, Garrido C. Primary tumor- and metastasis-derived colon cancer cells differently modulate connexin expression and function in human capillary endothelial cells. Oncotarget 2015; 6:28800-15. [PMID: 26320187 PMCID: PMC4745693 DOI: 10.18632/oncotarget.4894] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/24/2015] [Indexed: 12/30/2022] Open
Abstract
A gradual loss of functional gap junction between tumor cells has been reported with colorectal cancer (CRC) progression. Here, we explored if colon cancer cells could also affect gap junctions in blood capillary cells. Human microvascular endothelial cells (HMEC) were cultured with two CRC cell lines established from a unique patient. SW480 cells, derived from the primary tumor, migrate much faster across HMEC monolayer than SW620 cells derived from a metastatic site. The motile SW480 cells highly express and release HSP27 that increases gap junction formation with HMEC. Soluble HSP27 phosphorylates the connexin Cx43 on serine residues and induces its interaction with the oncoprotein 14-3-3, which promotes Cx43 delivery at the plasma membrane. The factors secreted by less motile SW620 cells do not affect Cx43 expression but up-regulate the expression of the connexin Cx32 through an activation of the chemokine receptor CXCR2. In turn, SW620 secreted factors induce tubulogenesis and ATP release. Altogether, cell lines derived from CRC primary tumor and metastasis differentially adapt endothelial cell functions by modulating connexin expression through released mediators.
Collapse
Affiliation(s)
| | | | - Laurent Cronier
- CNRS ERL7368, STIM Lab, University of Poitiers, 86022 Poitiers, France
| | - Eric Solary
- INSERM, U1170, Institut Gustave Roussy, 94508 Villejuif, France
| | - Carmen Garrido
- INSERM, U866 Faculty of Medecine, 21000 Dijon, France
- University of Bourgogne-Franche-Comté, 21000 Dijon, France
- CGFL, 21000 Dijon, France
| |
Collapse
|
38
|
Qiu X, Cheng JC, Zhao J, Chang HM, Leung PCK. Transforming growth factor-β stimulates human ovarian cancer cell migration by up-regulating connexin43 expression via Smad2/3 signaling. Cell Signal 2015; 27:1956-62. [PMID: 26186970 DOI: 10.1016/j.cellsig.2015.07.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 07/13/2015] [Indexed: 11/24/2022]
Abstract
Reduced connexin43 (Cx43) expression is frequently detected in different types of human cancer. Cx43 has been shown to regulate cancer cell migration in a cell-type dependent manner. In both primary and recurrent human ovarian cancer, overexpression of TGF-β ligand and its receptors have been detected. TGF-β can regulate Cx43 expression in other cell types and stimulate human ovarian cancer cell migration. However, whether Cx43 can be regulated by TGF-β and is involved in TGF-β-stimulated cell migration in human ovarian cancer cells remain unknown. In this study, we demonstrate that TGF-β up-regulates Cx43 in two human ovarian cancer cell lines, SKOV3 and OVCAR4. The stimulatory effect of TGF-β on Cx43 expression is blocked by inhibition of TGF-β receptor. Treatment with TGF-β activates Smad2 and Smad3 signaling pathways in both ovarian cancer cell lines. In addition, siRNA-mediated knockdown of Smad2 or Smd3 abolishes TGF-β-induced up-regulation of Cx43 expression. Moreover, knockdown of Cx43 attenuates TGF-β-stimulated cell migration. This study demonstrates an important role for Cx43 in mediating the effects of TGF-β on human ovarian cancer cell migration.
Collapse
Affiliation(s)
- Xin Qiu
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Jung-Chien Cheng
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Jianfang Zhao
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada.
| |
Collapse
|
39
|
Schalper KA, Carvajal-Hausdorf D, Oyarzo MP. Possible role of hemichannels in cancer. Front Physiol 2014; 5:237. [PMID: 25018732 PMCID: PMC4073485 DOI: 10.3389/fphys.2014.00237] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 06/09/2014] [Indexed: 12/12/2022] Open
Abstract
In humans, connexins (Cxs) and pannexins (Panxs) are the building blocks of hemichannels. These proteins are frequently altered in neoplastic cells and have traditionally been considered as tumor suppressors. Alteration of Cxs and Panxs in cancer cells can be due to genetic, epigenetic and post-transcriptional/post-translational events. Activated hemichannels mediate the diffusional membrane transport of ions and small signaling molecules. In the last decade hemichannels have been shown to participate in diverse cell processes including the modulation of cell proliferation and survival. However, their possible role in tumor growth and expansion remains largely unexplored. Herein, we hypothesize about the possible role of hemichannels in carcinogenesis and tumor progression. To support this theory, we summarize the evidence regarding the involvement of hemichannels in cell proliferation and migration, as well as their possible role in the anti-tumor immune responses. In addition, we discuss the evidence linking hemichannels with cancer in diverse models and comment on the current technical limitations for their study.
Collapse
Affiliation(s)
- Kurt A Schalper
- Servicio Anatomía Patológica, Clínica Alemana de Santiago, Facultad de Medicina Clinica Alemana Universidad del Desarrollo Santiago, Chile ; Department of Pathology, Yale School of Medicine New Haven, CT, USA
| | | | - Mauricio P Oyarzo
- Servicio Anatomía Patológica, Clínica Alemana de Santiago, Facultad de Medicina Clinica Alemana Universidad del Desarrollo Santiago, Chile
| |
Collapse
|
40
|
Ryszawy D, Sarna M, Rak M, Szpak K, Kędracka-Krok S, Michalik M, Siedlar M, Zuba-Surma E, Burda K, Korohoda W, Madeja Z, Czyż J. Functional links between Snail-1 and Cx43 account for the recruitment of Cx43-positive cells into the invasive front of prostate cancer. Carcinogenesis 2014; 35:1920-30. [PMID: 24503443 DOI: 10.1093/carcin/bgu033] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Suppressive function of connexin(Cx)43 in carcinogenesis was recently contested by reports that showed a multifaceted function of Cx43 in cancer progression. These studies did not attempt to model the dynamics of intratumoral heterogeneity involved in the metastatic cascade. An unorthodox look at the phenotypic heterogeneity of prostate cancer cells in vitro enabled us to identify links between Cx43 functions and Snail-1-regulated functional speciation of invasive cells. Incomplete Snail-1-dependent phenotypic shifts accounted for the formation of phenotypically stable subclones of AT-2 cells. These subclones showed diverse predilection for invasive behavior. High Snail-1 and Cx43 levels accompanied high motility and nanomechanical elasticity of the fibroblastoid AT-2_Fi2 subclone, which determined its considerable invasiveness. Transforming growth factor-β and ectopic Snail-1 overexpression induced invasiveness and Cx43 expression in epithelioid AT-2 subclones and DU-145 cells. Functional links between Snail-1 function and Cx43 expression were confirmed by Cx43 downregulation and phenotypic shifts in AT-2_Fi2, DU-145 and MAT-LyLu cells upon Snail-1 silencing. Corresponding morphological changes and Snail-1 downregulation were seen upon Cx43 silencing in AT-2_Fi2 cells. This indicates that feedback loops between both proteins regulate cell invasive behavior. We demonstrate that Cx43 may differentially predispose prostate cancer cells for invasion in a coupling-dependent and coupling-independent manner. When extrapolated to in vivo conditions, these data show the complexity of Cx43 functions during the metastatic cascade of prostate cancer. They may explain how Cx43 confers a selective advantage during cooperative invasion of clonally evolving, invasive prostate cancer cell subpopulations.
Collapse
Affiliation(s)
- Damian Ryszawy
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland, Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland, Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Michał Sarna
- Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland
| | - Monika Rak
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland, Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland, Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Katarzyna Szpak
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland, Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland, Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Sylwia Kędracka-Krok
- Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and
| | - Marta Michalik
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland, Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland, Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland, Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland, Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Kvetoslava Burda
- Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland
| | - Włodzimierz Korohoda
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland, Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland, Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland, Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland, Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Jarosław Czyż
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland, Department of Medical Physics and Biophysics, AGH University of Science and Technology, 30-059 Kraków, Poland, Department of Physical Biochemistry, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland and Department of Clinical Immunology, Polish-American Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| |
Collapse
|
41
|
Defamie N, Chepied A, Mesnil M. Connexins, gap junctions and tissue invasion. FEBS Lett 2014; 588:1331-8. [PMID: 24457198 DOI: 10.1016/j.febslet.2014.01.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 12/14/2022]
Abstract
Formation of metastases negatively impacts the survival prognosis of cancer patients. Globally, if the various steps involved in their formation are relatively well identified, the molecular mechanisms responsible for the emergence of invasive cancer cells are still incompletely resolved. Elucidating what are the mechanisms that allow cancer cells to evade from the tumor is a crucial point since it is the first step of the metastatic potential of a solid tumor. In order to be invasive, cancer cells have to undergo transformations such as down-regulation of cell-cell adhesions, modification of cell-matrix adhesions and acquisition of proteolytic properties. These transformations are accompanied by the capacity to "activate" stromal cells, which may favor the motility of the invasive cells through the extracellular matrix. Since modulation of gap junctional intercellular communication is known to be involved in cancer, we were interested to consider whether these different transformations necessary for the acquisition of invasive phenotype are related with gap junctions and their structural proteins, the connexins. In this review, emerging roles of connexins and gap junctions in the process of tissue invasion are proposed.
Collapse
Affiliation(s)
- Norah Defamie
- Team IP2C, STIM laboratory, University of Poitiers, CNRS ERL 7368, 1 rue Georges Bonnet, B36, 86073 Poitiers Cedex9, France.
| | - Amandine Chepied
- Team IP2C, STIM laboratory, University of Poitiers, CNRS ERL 7368, 1 rue Georges Bonnet, B36, 86073 Poitiers Cedex9, France.
| | - Marc Mesnil
- Team IP2C, STIM laboratory, University of Poitiers, CNRS ERL 7368, 1 rue Georges Bonnet, B36, 86073 Poitiers Cedex9, France.
| |
Collapse
|
42
|
Zhou JZ, Jiang JX. Gap junction and hemichannel-independent actions of connexins on cell and tissue functions--an update. FEBS Lett 2014; 588:1186-92. [PMID: 24434539 DOI: 10.1016/j.febslet.2014.01.001] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 12/20/2013] [Accepted: 01/02/2014] [Indexed: 10/25/2022]
Abstract
Connexins, a family of transmembrane proteins, are components of both gap junction channels and hemichannels, which mediate the exchange of ions and small molecules between adjacent cells, and between the inside and outside of the cell, respectively. Substantial advancements have been made in the comprehension of the role of gap junctions and hemichannels in coordinating cellular events. In recent years, a plethora of studies demonstrate a role of connexin proteins in the regulation of tissue homeostasis that occurs independently of their channel activities. This is shown in the context of cell growth, adhesion, migration, apoptosis, and signaling. The major mechanisms of these channel-independent activities still remain to be discovered. In this review, we provide an updated overview on the current knowledge of gap junction- and hemichannel-independent functions of connexins, in particular, their effects on tumorigenesis, neurogenesis and disease development.
Collapse
Affiliation(s)
- Jade Z Zhou
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Jean X Jiang
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA.
| |
Collapse
|
43
|
Du G, Yang Y, Yang Y, Zhang Y, Sun T, Liu W, Wang Y, Li J, Zhang H. Thrombocytosis and immunohistochemical expression of connexin 43 at diagnosis predict survival in advanced non-small-cell lung cancer treated with cisplatin-based chemotherapy. Cancer Chemother Pharmacol 2013; 71:893-904. [PMID: 23355038 DOI: 10.1007/s00280-013-2080-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 01/08/2013] [Indexed: 02/04/2023]
Abstract
PURPOSE Patients with advanced non-small-cell lung cancer (NSCLC) have poor survival, and platinum-based chemotherapy agents are the standard first-line chemotherapy agents for advanced NSCLC. This study aimed to identify predictive factors associated with the response to chemotherapy and survival in 258 patients with advanced NSCLC treated with platinum-based chemotherapy. METHODS Stage IIIA-IV NSCLC patients diagnosed in Kaifeng second people's hospital (Henan, China) between March 2002 and September 2011 were retrospectively reviewed. All of the patients had received platinum-based chemotherapy, and patients were followed up to date of death or last follow-up to obtain data of response to chemotherapy and survival. Potential prognostic factors such as gender, age, tumor size, tumor type, histologic stage, anemia, calcium levels, ECOG performance status (PS), thrombocytosis, TTF-1, p63, and connexin 43 were analyzed. Response to chemotherapy, overall survival (OS) and progression-free survival (PFS) were calculated by the Kaplan-Meier method and Cox regression model. RESULTS A univariate analysis indicated that thrombocytosis and connexin 43 were found to be significant prognostic factors (p < 0.001) and ECOG PS, Hb levels, and p63 presented a tendency toward association with survival. Kaplan-Meier survival showed that the mean OS and PFS in chemotherapy responders with connexin 43 ≥ +2 were significantly longer than in chemotherapy responders with connexin 43 ≤ 1+. In contrast, thrombocytosis was associated with increased mortality and resistance to chemotherapy in chemotherapy responders. In addition, all 21 patients of the 5-year OS were from chemotherapy responders with connexin 43 ≥ +2 or non-thrombocytosis. CONCLUSIONS Thrombocytosis and connexin 43 absence may be reliable surrogate markers for the prediction of chemotherapy response and prognosis for patients with advanced NSCLC, and assessment of these factors may identify a population of patients with advanced NSCLC that is likely to have a prolonged life expectancy.
Collapse
Affiliation(s)
- Gangjun Du
- Institute of Pharmacy, Pharmacy College of Henan University, Jinming street, Kaifeng, 475004 Henan, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Current World Literature. Curr Opin Support Palliat Care 2012; 6:402-16. [DOI: 10.1097/spc.0b013e3283573126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Li X, Pan JH, Song B, Xiong EQ, Chen ZW, Zhou ZS, Su YP. Suppression of CX43 expression by miR-20a in the progression of human prostate cancer. Cancer Biol Ther 2012; 13:890-8. [PMID: 22785209 DOI: 10.4161/cbt.20841] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The aberrant expression of microRNAs (miRNAs) has been found in various types of cancer. The present study found miR-20a to be significantly upregulated in prostate cancer compared with normal prostate tissues. The proliferation and colony formation assays revealed that the downregulation of miR-20a by miR-20a inhibitor suppresses the proliferation of MDA-PCa-2b cells in vitro and also inhibits tumor growth in vivo. Furthermore, a gap junction protein, α 1 (CX43), was identified as a direct target gene of miR-20a. The upregulation of CX43 was detected in MDA-PCa-2b cells after treatment with miR-20a inhibitor both in vitro and in vivo. In conclusion, the findings show that miR-20a significantly contributes to the progression of prostate cancer by targeting CX43.
Collapse
Affiliation(s)
- Xin Li
- Urologic Institute of PLA, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Czyż J, Szpak K, Madeja Z. The role of connexins in prostate cancer promotion and progression. Nat Rev Urol 2012; 9:274-82. [PMID: 22349655 DOI: 10.1038/nrurol.2012.14] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate cancer is a prevalent disease that is characterized by a presumably long latency period and a moderate propensity to metastasize. Although a range of mechanisms have been implicated in prostate carcinogenesis, the factors determining the initiation of metastasis remain obscure. The synchronized function of prostate cells depends on their metabolic and electrical coupling; disturbance of these functions has long been suggested to be integral to prostate carcinogenesis. However, although connexins form intercellular channels involved in gap-junction-mediated intercellular coupling (GJIC), whether these proteins also have GJIC-independent roles in cancer progression and metastasis remains a matter of debate. Some data indicate a correlation between connexin expression and the invasive potential of prostate cancer cells, which points to stage-specific functions of connexins during prostate cancer development. For example, restoration of connexin expression seems to be crucial for the formation of invasive cell subsets within heterogeneous prostate cancer cell populations that have undergone aberrant differentiation. Consequently, the clinical application of therapeutic and prophylactic approaches focused on the modulation of connexin expression in prostate cancer cells should be reconsidered.
Collapse
Affiliation(s)
- Jarosław Czyż
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland.
| | | | | |
Collapse
|