1
|
da Fonseca IIM, Nagamine MK, Sato A, Rossatto-Jr CA, Yeh ES, Dagli MLZ. Inhibitory Effects of Alpha-Connexin Carboxyl-Terminal Peptide on Canine Mammary Epithelial Cells: A Study on Benign and Malignant Phenotypes. Cancers (Basel) 2024; 16:820. [PMID: 38398211 PMCID: PMC10887206 DOI: 10.3390/cancers16040820] [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] [Received: 01/08/2024] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Mammary cancer is highly prevalent in non-castrated female dogs. Cell-to-cell communication is an important mechanism to maintain homeostasis, and connexins are proteins that assemble to form the communicating gap junctions. In many cancers, communication capacity is reduced; several approaches are being tested in order to increase the communication capacity in cancer cells and, therefore, alter their viability. This study analyzed the effects of the alpha-connexin carboxyl-terminal peptide (αCT1) on canine mammary non-neoplastic and neoplastic epithelial cells. Seven canine epithelial mammary cell lines were used. Among these, one was a normal canine epithelial mammary cell line (LOEC-NMG), two canine mammary adenomas (LOEC-MAd1 and LOEC-MAd2), and four canine mammary adenocarcinomas (LOEC-MCA1, LOEC-MCA2, LOEC-MCA3 and CF41). The αCT1 corresponds to a short Cx43 C-terminal sequence linked to an internalization sequence called the antennapedia. After 24 h of incubation, the medium containing different αCT1 peptide concentrations was added to the cells, and only the culture medium was used for control. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test was used to quantify cell viability before treatment and 48, 72, and 96 h after the treatment. Results showed that the normal mammary epithelial cell line (LOEC-NMG) was resistant to treatment with αCT1, which is consistent with a previous study on human mammary cell lines. One of the adenoma cell lines (LOEC-MAd2) was also resistant to treatment with αCT1, although the other (LOEC-MAd1) was susceptible to treatment, mostly at 72 h after treatment. Regarding the four canine adenocarcinoma cell lines, they differ regarding the susceptibility to the treatment with αCT1. Three cell lines, canine mixed adenocarcinoma (LOEC-MCA1), canine complex adenocarcinoma (LOEC-MCA2), and commercial canine mammary adenocarcinoma cell line CF41, were susceptible to treatment with αCT1, while one canine mammary adenocarcinoma cell line (LOEC-MCA3) was resistant to treatment. In most αCT1 treated cell lines, Cx43 was strongly detected in cell membranes by immunofluorescence. We propose that αCT1 restored the cell-to-cell communication capacity of neoplastic cells and induced inhibitory effects on cell viability.
Collapse
Affiliation(s)
- Ivone Izabel Mackowiak da Fonseca
- Laboratory of Experimental and Comparative Oncology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-900, Brazil; (I.I.M.d.F.); (M.K.N.); (A.S.); (C.A.R.-J.)
| | - Marcia Kazumi Nagamine
- Laboratory of Experimental and Comparative Oncology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-900, Brazil; (I.I.M.d.F.); (M.K.N.); (A.S.); (C.A.R.-J.)
| | - Ayami Sato
- Laboratory of Experimental and Comparative Oncology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-900, Brazil; (I.I.M.d.F.); (M.K.N.); (A.S.); (C.A.R.-J.)
- Institute of Life Innovation Studies, Toyo University, Tokyo 374-0193, Japan
| | - Carlos Alberto Rossatto-Jr
- Laboratory of Experimental and Comparative Oncology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-900, Brazil; (I.I.M.d.F.); (M.K.N.); (A.S.); (C.A.R.-J.)
| | - Elizabeth Shinmay Yeh
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Maria Lucia Zaidan Dagli
- Laboratory of Experimental and Comparative Oncology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-900, Brazil; (I.I.M.d.F.); (M.K.N.); (A.S.); (C.A.R.-J.)
| |
Collapse
|
2
|
Mulchandani V, Banerjee A, Vadlamannati AV, Kumar S, Das Sarma J. Connexin 43 trafficking and regulation of gap junctional intercellular communication alters ovarian cancer cell migration and tumorigenesis. Biomed Pharmacother 2023; 159:114296. [PMID: 36701988 DOI: 10.1016/j.biopha.2023.114296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
Ovarian cancer persists to be the most lethal gynecological malignancy, demanding rigorous treatments involving radio-chemotherapy that trigger toxicity and consequently mortality among patients. An improved understanding of the disease progression may pioneer curative therapies. Mouse epithelial ovarian cancer cell lines, ID8 and ID8-VEGF (overexpressing VEGF) were intraperitoneally injected in C57BL/6 female mice to develop a Syngeneic Ovarian cancer mouse model. It was observed that ID8-VEGF cells were able to induce aggressive tumor growth in mice compared to ID8 cells. Furthermore, results of the current in vitro study comparing ID8 and ID8-VEGF demonstrated that highly tumorigenic ID8-VEGF had reduced gap junctional intercellular communication (GJIC) due to intracellular Connexin 43 (Cx43) expression. Additionally, ID8 cells with reduced tumorigenic capability expressed significant GJIC. Furthermore, loss of GJIC in ID8-VEGF cells induced shorter tunneling nanotube formations, while ID8 cells develops longer tunneling nanotube to maintain cellular crosstalk. The administration of a pharmacological drug 4-phenylbutyrate (4PBA) ensured the restoration of GJIC in both the ovarian cancer cell lines. Additionally, 4PBA treatment significantly inhibited the migration of ovarian cancer cell lines and tumor formation in ovarian cancer mice models. In summary, the 4PBA-mediated restoration of GJIC suppressed migration (in vitro) and tumorigenesis (in vivo) of ovarian cancer cells. The present study suggests that Cx43 assembled GJIC and its supportive signaling pathways are a prospective target for restricting ovarian cancer progression.
Collapse
Affiliation(s)
- Vaishali Mulchandani
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Anurag Banerjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Arunima Vijaya Vadlamannati
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Saurav Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India; Department of Ophthalmology, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
3
|
Takamura N, Yamaguchi Y. Involvement of caveolin-1 in skin diseases. Front Immunol 2022; 13:1035451. [PMID: 36532050 PMCID: PMC9748611 DOI: 10.3389/fimmu.2022.1035451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/17/2022] [Indexed: 12/02/2022] Open
Abstract
The skin is the outermost layer and largest organ in the human body. Since the skin interfaces with the environment, it has a variety of roles, including providing a protective barrier against external factors, regulating body temperature, and retaining water in the body. It is also involved in the immune system, interacting with immune cells residing in the dermis. Caveolin-1 (CAV-1) is essential for caveolae formation and has multiple functions including endocytosis, lipid homeostasis, and signal transduction. CAV-1 is known to interact with a variety of signaling molecules and receptors and may influence cell proliferation and migration. Several skin-related disorders, especially those of the inflammatory or hyperproliferative type such as skin cancers, psoriasis, fibrosis, and wound healing, are reported to be associated with aberrant CAV-1 expression. In this review, we have explored CAV-1 involvement in skin physiology and skin diseases.
Collapse
|
4
|
FABP6 Expression Correlates with Immune Infiltration and Immunogenicity in Colorectal Cancer Cells. J Immunol Res 2022; 2022:3129765. [PMID: 36033394 PMCID: PMC9403257 DOI: 10.1155/2022/3129765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have rapidly revolutionized colorectal cancer (CRC) treatment, but resistance caused by the heterogeneous tumor microenvironment (TME) still presents a challenge. Therefore, it is necessary to characterize TME immune infiltration and explore new targets to improve immunotherapy. Methods The compositions of 64 types of infiltrating immune cells and their relationships with CRC patient clinical characteristics were assessed. Differentially expressed genes (DEGs) between “hot” and “cold” tumors were used for functional analysis. A prediction model was constructed to explore the survival of CRC patients treated with and without immunotherapy. Finally, fatty acid-binding protein (FABP6) was selected for in vitro experiments, which revealed its roles in the proliferation, apoptosis, migration, and immunogenicity of CRC tissues and cell lines. Results The infiltration levels of several immune cells were associated with CRC tumor stage and prognosis. Different cell types showed the synergistic or antagonism infiltration patterns. Enrichment analysis of DEGs revealed that immune-related signaling was significantly activated in hot tumors, while metabolic process pathways were altered in cold tumors. In addition, the constructed model effectively predicted the survival of CRC patients treated with and without immunotherapy. FABP6 knockdown did not significantly alter tumor cell proliferation, apoptosis, and migration. FABP6 was negatively correlated with immune infiltration, and knockdown of FABP6 increased major histocompatibility complex (MHC) class 1 expression and promoted immune-related chemokine secretion, indicating the immunogenicity enhancement of tumor cells. Finally, knockdown of FABP6 could promote the recruitment of CD8+ T cells. Conclusion Collectively, we described the landscape of immune infiltration in CRC and identified FABP6 as a potential immunotherapeutic target for treatment.
Collapse
|
5
|
Liu S, Zhao Y, Liu H, Zhao X, Shen X. miR-301-3p directly regulates Cx43 to mediate the development of gastric cancer. J Int Med Res 2021; 49:3000605211033185. [PMID: 34590921 PMCID: PMC8489753 DOI: 10.1177/03000605211033185] [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: 01/18/2021] [Accepted: 06/24/2021] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Identifying novel biomarkers involved in the development of gastric cancer (GC) can provide potential therapeutic strategies and improve clinical prognosis. miR-301-3p and Cx43 are reportedly dysregulated in GC. miR-301-3p and Cx43 interaction, and their functions in GC progression, are still poorly understood. METHODS The expression levels of miR-301-3p and Cx43 in GC tissues and cell lines with various differentiation degrees were evaluated by RT-qPCR. The interaction between miR-301-3p and Cx43 was assessed by dual-luciferase reporter assays. CCK8 and Transwell assays were employed to assess the effects of the miR-301-3p-Cx43 axis on GC cell proliferation, migration, and invasion. RESULTS Cx43 was significantly downregulated in GC tissues and cell lines, while miR-301-3p expression was negatively correlated with Cx43 mRNA levels. The expression levels of Cx43 and miR-301-3p were closely associated with the differentiation, TNM stage, vascular invasion, and lymph node metastasis status of GC patients. Cx43 overexpression could suppress the proliferation, migration, and invasion of GC cells. Cx43 mRNA is a direct target of miR-301-3p, and transfection of an miR-301-3p mimic could reverse the inhibitory effects of Cx43. CONCLUSION The miR-301-3p-Cx43 axis is involved in the development and progression of GC by affecting the proliferation, migration, and invasion of GC cells.
Collapse
Affiliation(s)
- Shasha Liu
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Hebei, 067000, China
| | - Yang Zhao
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Hebei, 067000, China
| | - Huan Liu
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Hebei, 067000, China
| | - Xing Zhao
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Hebei, 067000, China
| | - Xingbin Shen
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Hebei, 067000, China
| |
Collapse
|
6
|
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
|
7
|
Sinyuk M, Mulkearns-Hubert EE, Reizes O, Lathia J. Cancer Connectors: Connexins, Gap Junctions, and Communication. Front Oncol 2018; 8:646. [PMID: 30622930 PMCID: PMC6308394 DOI: 10.3389/fonc.2018.00646] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022] Open
Abstract
Despite concerted clinical and research efforts, cancer is a leading cause of death worldwide. Surgery, radiation, and chemotherapy have remained the most common standard-of-care strategies against cancer for decades. However, the side effects of these therapies demonstrate the need to investigate adjuvant novel treatment modalities that minimize the harm caused to healthy cells and tissues. Normal and cancerous cells require communication amongst themselves and with their surroundings to proliferate and drive tumor growth. It is vital to understand how intercellular and external communication impacts tumor cell malignancy. To survive and grow, tumor cells, and their normal counterparts utilize cell junction molecules including gap junctions (GJs), tight junctions, and adherens junctions to provide contact points between neighboring cells and the extracellular matrix. GJs are specialized structures composed of a family of connexin proteins that allow the free diffusion of small molecules and ions directly from the cytoplasm of adjacent cells, without encountering the extracellular milieu, which enables rapid, and coordinated cellular responses to internal and external stimuli. Importantly, connexins perform three main cellular functions. They enable direct gap junction intercellular communication (GJIC) between cells, form hemichannels to allow cell communication with the extracellular environment, and serve as a site for protein-protein interactions to regulate signaling pathways. Connexins themselves have been found to promote tumor cell growth and invasiveness, contributing to the overall tumorigenicity and have emerged as attractive anti-tumor targets due to their functional diversity. However, connexins can also serve as tumor suppressors, and therefore, a complete understanding of the roles of the connexins and GJs in physiological and pathophysiological conditions is needed before connexin targeting strategies are applied. Here, we discuss how the three aspects of connexin function, namely GJIC, hemichannel formation, and connexin-protein interactions, function in normal cells, and contribute to tumor cell growth, proliferation, and death. Finally, we discuss the current state of anti-connexin therapies and speculate which role may be most amenable for the development of targeting strategies.
Collapse
Affiliation(s)
- Maksim Sinyuk
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Erin E. Mulkearns-Hubert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Ofer Reizes
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, United States
- Case Comprehensive Cancer Center, Case Western University, Cleveland, OH, United States
| | - Justin Lathia
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, United States
- Case Comprehensive Cancer Center, Case Western University, Cleveland, OH, United States
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
8
|
Gheida SF, Neinaa YMEH, Mohammed DAEA. Caveolin-1 expression in hyperproliferative skin disorders: A potential predictive marker of disease severity and progression. DERMATOL SIN 2018. [DOI: 10.1016/j.dsi.2018.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
9
|
Kam CY, Dubash AD, Magistrati E, Polo S, Satchell KJF, Sheikh F, Lampe PD, Green KJ. Desmoplakin maintains gap junctions by inhibiting Ras/MAPK and lysosomal degradation of connexin-43. J Cell Biol 2018; 217:3219-3235. [PMID: 29959233 PMCID: PMC6123000 DOI: 10.1083/jcb.201710161] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 04/26/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
Desmosomal mutations result in potentially deadly cardiocutaneous disease caused by electrical conduction defects and disruption of gap junctions. Kam et al. demonstrate a mechanism whereby loss of the intermediate filament anchoring protein desmoplakin stimulates Cx43 turnover by increasing K-Ras expression, marking Cx43 for lysosomal degradation through ERK1/2 phosphorylation. Desmoplakin (DP) is an obligate component of desmosomes, intercellular adhesive junctions that maintain the integrity of the epidermis and myocardium. Mutations in DP can cause cardiac and cutaneous disease, including arrhythmogenic cardiomyopathy (ACM), an inherited disorder that frequently results in deadly arrhythmias. Conduction defects in ACM are linked to the remodeling and functional interference with Cx43-based gap junctions that electrically and chemically couple cells. How DP loss impairs gap junctions is poorly understood. We show that DP prevents lysosomal-mediated degradation of Cx43. DP loss triggered robust activation of ERK1/2–MAPK and increased phosphorylation of S279/282 of Cx43, which signals clathrin-mediated internalization and subsequent lysosomal degradation of Cx43. RNA sequencing revealed Ras-GTPases as candidates for the aberrant activation of ERK1/2 upon loss of DP. Using a novel Ras inhibitor, Ras/Rap1-specific peptidase (RRSP), or K-Ras knockdown, we demonstrate restoration of Cx43 in DP-deficient cardiomyocytes. Collectively, our results reveal a novel mechanism for the regulation of the Cx43 life cycle by DP in cardiocutaneous models.
Collapse
Affiliation(s)
- Chen Yuan Kam
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Adi D Dubash
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Simona Polo
- Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy.,Dipartimento di Oncologia ed Emato-oncologia, Universita' degli Studi di Milano, Milan, Italy
| | - Karla J F Satchell
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Farah Sheikh
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Paul D Lampe
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Research Center, Seattle, WA
| | - Kathleen J Green
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL .,Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
10
|
James CC, Zeitz MJ, Calhoun PJ, Lamouille S, Smyth JW. Altered translation initiation of Gja1 limits gap junction formation during epithelial-mesenchymal transition. Mol Biol Cell 2018; 29:797-808. [PMID: 29467255 PMCID: PMC5905293 DOI: 10.1091/mbc.e17-06-0406] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is activated during development, wound healing, and pathologies including fibrosis and cancer metastasis. Hallmarks of EMT are remodeling of intercellular junctions and adhesion proteins, including gap junctions. The GJA1 mRNA transcript encoding the gap junction protein connexin43 (Cx43) has been demonstrated to undergo internal translation initiation, yielding truncated isoforms that modulate gap junctions. The PI3K/Akt/mTOR pathway is central to translation regulation and is activated during EMT, leading us to hypothesize that altered translation initiation would contribute to gap junction loss. Using TGF-β-induced EMT as a model, we find reductions in Cx43 gap junctions despite increased transcription and stabilization of Cx43 protein. Biochemical experiments reveal suppression of the internally translated Cx43 isoform, GJA1-20k in a Smad3 and ERK-dependent manner. Ectopic expression of GJA1-20k does not halt EMT, but is sufficient to rescue gap junction formation. GJA1-20k localizes to the Golgi apparatus, and using superresolution localization microscopy we find retention of GJA1-43k at the Golgi in mesenchymal cells lacking GJA1-20k. NativePAGE demonstrates that levels of GJA1-20k regulate GJA1-43k hexamer oligomerization, a limiting step in Cx43 trafficking. These findings reveal alterations in translation initiation as an unexplored mechanism by which the cell regulates Cx43 gap junction formation during EMT.
Collapse
Affiliation(s)
- Carissa C James
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| | - Michael J Zeitz
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016
| | - Patrick J Calhoun
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| | - Samy Lamouille
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016
| | - James W Smyth
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| |
Collapse
|
11
|
Karsch-Bluman A, Amoyav B, Friedman N, Shoval H, Schwob O, Ella E, Wald O, Benny O. High mobility group box 1 antagonist limits metastatic seeding in the lungs via reduction of cell-cell adhesion. Oncotarget 2018; 8:32706-32721. [PMID: 28415753 PMCID: PMC5464821 DOI: 10.18632/oncotarget.16188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/06/2017] [Indexed: 01/08/2023] Open
Abstract
Metastatic spread is the leading cause for cancer-related mortality, with the lungs being a major site for metastatic seeding. Available therapies for patients with metastatic disease are extremely limited. Therefore, there is a desperate need for new strategies to prevent or limit metastatic dissemination and treat existing metastases. The metastatic cascade is highly complex and is affected by multiple factors related to both tumor cells themselves and the microenvironment in the future site of metastasis. We hypothesized that modifying the lung microenvironment by blocking central ubiquitous signals may affect metastatic seeding in the lungs. Given the high basal levels of the Receptor for Advanced Glycation End products (RAGE) in the pulmonary tissue, and its pro-inflammatory properties, we investigated the consequences of interfering with its ligand; High Mobility Group Box 1 (HMGB1). To this end, we tested the effect of Carbenoxolone, an HMGB1 antagonist, on primary tumor growth and metastatic progression in several murine tumor models. We show that antagonizing HMGB1 prevents the adhesion and colonization of cancer cells in the lungs through the reduction of their adhesion and cell–cell interaction both in vitro and in vivo. We demonstrated that these activities are mediated by downregulation of the adhesion molecule Intercellular Adhesion Molecule 1 (ICAM1) and ultimately result in reduced metastatic burden. Carbenoxolone decreases significantly lung metastases formation and can be used potentially as prophylactic therapy for metastatic diseases.
Collapse
Affiliation(s)
- Adi Karsch-Bluman
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Benzion Amoyav
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nethanel Friedman
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hila Shoval
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ouri Schwob
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ezra Ella
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ori Wald
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.,Division of General Thoracic Surgery, Michael E. DeBakey Department of General Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Ofra Benny
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
12
|
Nyffeler J, Chovancova P, Dolde X, Holzer AK, Purvanov V, Kindinger I, Kerins A, Higton D, Silvester S, van Vugt-Lussenburg BMA, Glaab E, van der Burg B, Maclennan R, Legler DF, Leist M. A structure-activity relationship linking non-planar PCBs to functional deficits of neural crest cells: new roles for connexins. Arch Toxicol 2017; 92:1225-1247. [PMID: 29164306 DOI: 10.1007/s00204-017-2125-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 11/15/2017] [Indexed: 12/14/2022]
Abstract
Migration of neural crest cells (NCC) is a fundamental developmental process, and test methods to identify interfering toxicants have been developed. By examining cell function endpoints, as in the 'migration-inhibition of NCC (cMINC)' assay, a large number of toxicity mechanisms and protein targets can be covered. However, the key events that lead to the adverse effects of a given chemical or group of related compounds are hard to elucidate. To address this issue, we explored here, whether the establishment of two overlapping structure-activity relationships (SAR)-linking chemical structure on the one hand to a phenotypic test outcome, and on the other hand to a mechanistic endpoint-was useful as strategy to identify relevant toxicity mechanisms. For this purpose, we chose polychlorinated biphenyls (PCB) as a large group of related, but still toxicologically and physicochemically diverse structures. We obtained concentration-dependent data for 26 PCBs in the cMINC assay. Moreover, the test chemicals were evaluated by a new high-content imaging method for their effect on cellular re-distribution of connexin43 and for their capacity to inhibit gap junctions. Non-planar PCBs inhibited NCC migration. The potency (1-10 µM) correlated with the number of ortho-chlorine substituents; non-ortho-chloro (planar) PCBs were non-toxic. The toxicity to NCC partially correlated with gap junction inhibition, while it fully correlated (p < 0.0004) with connexin43 cellular re-distribution. Thus, our double-SAR strategy revealed a mechanistic step tightly linked to NCC toxicity of PCBs. Connexin43 patterns in NCC may be explored as a new endpoint relevant to developmental toxicity screening.
Collapse
Affiliation(s)
- Johanna Nyffeler
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany.,Research Training Group RTG1331, 78457, Konstanz, Germany
| | - Petra Chovancova
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), 78457, Konstanz, Germany
| | - Xenia Dolde
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), 78457, Konstanz, Germany
| | - Anna-Katharina Holzer
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany
| | - Vladimir Purvanov
- Biotechnology Institute Thurgau at the University of Konstanz, 8280, Kreuzlingen, Switzerland
| | - Ilona Kindinger
- Biotechnology Institute Thurgau at the University of Konstanz, 8280, Kreuzlingen, Switzerland
| | - Anna Kerins
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | - David Higton
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | - Steve Silvester
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | | | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362, Esch-sur-Alzette, Luxembourg
| | - Bart van der Burg
- BioDetection Systems bv, Science Park 406, 1098XH, Amsterdam, The Netherlands
| | - Richard Maclennan
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | - Daniel F Legler
- Research Training Group RTG1331, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), 78457, Konstanz, Germany.,Biotechnology Institute Thurgau at the University of Konstanz, 8280, Kreuzlingen, Switzerland
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany. .,Research Training Group RTG1331, 78457, Konstanz, Germany. .,Konstanz Research School Chemical Biology (KoRS-CB), 78457, Konstanz, Germany.
| |
Collapse
|
13
|
Föll MC, Fahrner M, Gretzmeier C, Thoma K, Biniossek ML, Kiritsi D, Meiss F, Schilling O, Nyström A, Kern JS. Identification of tissue damage, extracellular matrix remodeling and bacterial challenge as common mechanisms associated with high-risk cutaneous squamous cell carcinomas. Matrix Biol 2017; 66:1-21. [PMID: 29158163 DOI: 10.1016/j.matbio.2017.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 01/03/2023]
Abstract
In this study we used a genetic extracellular matrix (ECM) disease to identify mechanisms associated with aggressive behavior of cutaneous squamous cell carcinoma (cSCC). cSCC is one of the most common malignancies and usually has a good prognosis. However, some cSCCs recur or metastasize and cause significant morbidity and mortality. Known factors that are associated with aggressiveness of cSCCs include tumor grading, size, localization and microinvasive behavior. To investigate molecular mechanisms that influence biologic behavior we used global proteomic and histologic analyses of formalin-fixed paraffin-embedded tissue of primary human cSCCs. We compared three groups: non-recurring, non-metastasizing low-risk sporadic cSCCs; metastasizing sporadic cSCCs; and cSCCs from patients with recessive dystrophic epidermolysis bullosa (RDEB). RDEB is a genetic skin blistering and ECM disease caused by collagen VII deficiency. Patients commonly suffer from high-risk early onset cSCCs that frequently metastasize. The results indicate that different processes are associated with formation of RDEB cSCCs compared to sporadic cSCCs. Sporadic cSCCs show signs of UV damage, whereas RDEB cSCCs have higher mutational rates and display tissue damage, inflammation and subsequent remodeling of the dermal ECM as tumor initiating factors. Interestingly the two high-risk groups - high-risk metastasizing sporadic cSCCs and RDEB cSCCs - are both associated with tissue damage and ECM remodeling in gene-ontology enrichment and Search Tool for the Retrieval of Interacting Genes/Proteins analyses. In situ histologic analyses validate these results. The high-risk cSCCs also show signatures of enhanced bacterial challenge. Histologic analyses confirm correlation of bacterial colonization with worse prognosis. Collectively, this unbiased study - performed directly on human patient material - reveals that common microenvironmental alterations linked to ECM remodeling and increased bacterial challenges are denominators of high-risk cSCCs. The proteins identified here could serve as potential diagnostic markers and therapeutic targets in high-risk cSCCs.
Collapse
Affiliation(s)
- Melanie C Föll
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Germany
| | - Matthias Fahrner
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Germany
| | - Christine Gretzmeier
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Käthe Thoma
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Germany
| | - Dimitra Kiritsi
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Frank Meiss
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Germany.
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany.
| | - Johannes S Kern
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Germany; Department of Dermatology, Royal Melbourne Hospital, Parkville and Box Hill Hospital - Monash University, Eastern Health Clinical School, Box Hill, Victoria, Australia
| |
Collapse
|
14
|
Leithe E, Mesnil M, Aasen T. The connexin 43 C-terminus: A tail of many tales. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:48-64. [PMID: 28526583 DOI: 10.1016/j.bbamem.2017.05.008] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/10/2017] [Accepted: 05/12/2017] [Indexed: 10/19/2022]
Abstract
Connexins are chordate gap junction channel proteins that, by enabling direct communication between the cytosols of adjacent cells, create a unique cell signalling network. Gap junctional intercellular communication (GJIC) has important roles in controlling cell growth and differentiation and in tissue development and homeostasis. Moreover, several non-canonical connexin functions unrelated to GJIC have been discovered. Of the 21 members of the human connexin family, connexin 43 (Cx43) is the most widely expressed and studied. The long cytosolic C-terminus (CT) of Cx43 is subject to extensive post-translational modifications that modulate its intracellular trafficking and gap junction channel gating. Moreover, the Cx43 CT contains multiple domains involved in protein interactions that permit crosstalk between Cx43 and cytoskeletal and regulatory proteins. These domains endow Cx43 with the capacity to affect cell growth and differentiation independently of GJIC. Here, we review the current understanding of the regulation and unique functions of the Cx43 CT, both as an essential component of full-length Cx43 and as an independent signalling hub. We highlight the complex regulatory and signalling networks controlled by the Cx43 CT, including the extensive protein interactome that underlies both gap junction channel-dependent and -independent functions. We discuss these data in relation to the recent discovery of the direct translation of specific truncated forms of Cx43. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, University of Oslo, NO-0424 Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, NO-0424 Oslo, Norway
| | - Marc Mesnil
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, France
| | - Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, 08035 Barcelona, Spain.
| |
Collapse
|
15
|
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
|
16
|
Fu P, Chen F, Pan Q, Zhao X, Zhao C, Cho WCS, Chen H. The different functions and clinical significances of caveolin-1 in human adenocarcinoma and squamous cell carcinoma. Onco Targets Ther 2017; 10:819-835. [PMID: 28243118 PMCID: PMC5317307 DOI: 10.2147/ott.s123912] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Caveolin-1 (Cav-1), a major structural protein of caveolae, is an integral membrane protein which plays an important role in the progression of carcinoma. However, whether Cav-1 acts as a tumor promoter or a tumor suppressor still remains controversial. For example, the tumor-promoting function of Cav-1 has been found in renal cancer, prostate cancer, tongue squamous cell carcinoma (SCC), lung SCC and bladder SCC. In contrast, Cav-1 also plays an inhibitory role in esophagus adenocarcinoma, lung adenocarcinoma and cutaneous SCC. The role of Cav-1 is still controversial in thyroid cancer, hepatocellular carcinoma, gastric adenocarcinoma, colon adenocarcinoma, breast cancer, pancreas cancer, oral SCC, laryngeal SCC, head and neck SCC, esophageal SCC and cervical SCC. Besides, it has been reported that the loss of stromal Cav-1 might predict poor prognosis in breast cancer, gastric cancer, pancreas cancer, prostate cancer, oral SCC and esophageal SCC. However, the accumulation of stromal Cav-1 has been found to be promoted by the progression of tongue SCC. Taken together, Cav-1 seems playing a different role in different cancer subtypes even of the same organ, as well as acting differently in the same cancer subtype of different organs. Thus, we hereby explore the functions of Cav-1 in human adenocarcinoma and SCC from the perspective of clinical significances and pathogenesis. We envision that novel targets may come with the further investigation of Cav-1 in carcinogenesis.
Collapse
Affiliation(s)
- Pin Fu
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | - Fuchun Chen
- Department of Thoracosurgery, Traditional Chinese Medical Hospital of Wenling, Wenling, Zhejiang
| | - Qi Pan
- Department of Thoracosurgery, Traditional Chinese Medical Hospital of Wenling, Wenling, Zhejiang
| | - Xianda Zhao
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | - Chen Zhao
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | | | - Honglei Chen
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan; Department of Pathology, Maternal and Child Health Hospital of Hubei, Wuhan, People's Republic of China
| |
Collapse
|
17
|
Ai XL, Chi Q, Qiu Y, Li HY, Li DJ, Wang JX, Wang ZY. Gap junction protein connexin43 deregulation contributes to bladder carcinogenesis via targeting MAPK pathway. Mol Cell Biochem 2017; 428:109-118. [PMID: 28074341 DOI: 10.1007/s11010-016-2921-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022]
Abstract
High expression of connexins was found in a variety of cancers, but their role is still controversial. We investigated whether connexin43 (Cx43) contributed to bladder carcinogenesis through MAPK activation. In this study, we found that Cx43 expression was significantly increased in bladder cancer tissues and cell line. Overexpression of Cx43 in bladder cancer 5637 cells increased cell proliferation, promoted cell cycle progression, and inhibited apoptosis. Western blot showed that JNK and ERK pathways were dramatically activated in Cx43-overexpressed cells. Conversely, knockdown of Cx43 inhibited cell proliferation by increasing apoptosis and causing cell cycle arrest, concomitant with inhibition of JNK and ERK signaling. In addition, JNK and ERK pathways were also activated in bladder cancer tissues. In conclusion, abnormal high expression and cytoplasmic localization of Cx43 contributed to bladder cancer. Inhibition of Cx43 activity could be a potential therapeutic strategy for preventing the progression of bladder cancer.
Collapse
Affiliation(s)
- Xiao-Lin Ai
- Department of Urology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Qiang Chi
- Department of Urology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yu Qiu
- Department of Urology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Hong-Yang Li
- Department of Urology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Dong-Jie Li
- Department of Urology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Jia-Xu Wang
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Zhi-Yong Wang
- Department of Urology, Affiliated Hospital of Chengde Medical University, Chengde, China.
| |
Collapse
|
18
|
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
|
19
|
Colangelo T, Polcaro G, Muccillo L, D'Agostino G, Rosato V, Ziccardi P, Lupo A, Mazzoccoli G, Sabatino L, Colantuoni V. Friend or foe? The tumour microenvironment dilemma in colorectal cancer. Biochim Biophys Acta Rev Cancer 2016; 1867:1-18. [PMID: 27864070 DOI: 10.1016/j.bbcan.2016.11.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/21/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
The network of bidirectional homotypic and heterotypic interactions established among parenchymal tumour cells and surrounding mesenchymal stromal cells generates the tumour microenvironment (TME). These intricate crosstalks elicit both beneficial and adverse effects on tumour initiation and progression unbalancing the signals and responses from the neighbouring cells. Here, we highlight the structure, activities and evolution of TME cells considering a novel colorectal cancer (CRC) classification based on differential stromal composition and gene expression profiles. In this scenario, we scrutinise the molecular pathways that either change or become corrupted during CRC development and their relative prognostic value. Finally, we survey the therapeutic molecules directed against TME components currently available in clinical trials as well as those with stronger potential in preclinical studies. Elucidation of dynamic variations in the CRC TME cell composition and their relative contribution could provide novel diagnostic or prognostic biomarkers and allow more personalised therapeutic strategies.
Collapse
Affiliation(s)
- Tommaso Colangelo
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy; present address: Institute for Stem-cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Casa Sollievo della Sofferenza-IRCCS, 71013 San Giovanni Rotondo (FG), Italy
| | - Giovanna Polcaro
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Livio Muccillo
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Giovanna D'Agostino
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Valeria Rosato
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Pamela Ziccardi
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Angelo Lupo
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo (FG), Italy
| | - Lina Sabatino
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | - Vittorio Colantuoni
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| |
Collapse
|
20
|
Abstract
Communication among cells via direct cell-cell contact by connexin gap junctions, or between cell and extracellular environment via pannexin channels or connexin hemichannels, is a key factor in cell function and tissue homeostasis. Upon malignant transformation in different cancer types, the dysregulation of these connexin and pannexin channels and their effect in cellular communication, can either enhance or suppress tumorigenesis and metastasis. In this review, we will highlight the latest reports on the role of the well characterized connexin family and its ability to form gap junctions and hemichannels in cancer. We will also introduce the more recently discovered family of pannexin channels and our current knowledge about their involvement in cancer progression.
Collapse
Affiliation(s)
- Jean X Jiang
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, N6A5C1, Canada.
| |
Collapse
|
21
|
Connexin 43, breast cancer tumor suppressor: Missed connections? Cancer Lett 2016; 374:117-126. [DOI: 10.1016/j.canlet.2016.02.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 12/21/2022]
|
22
|
Leithe E. Regulation of connexins by the ubiquitin system: Implications for intercellular communication and cancer. Biochim Biophys Acta Rev Cancer 2016; 1865:133-46. [DOI: 10.1016/j.bbcan.2016.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/15/2016] [Accepted: 02/04/2016] [Indexed: 12/31/2022]
|
23
|
Interaction of Cx43 with Hsc70 regulates G1/S transition through CDK inhibitor p27. Sci Rep 2015; 5:15365. [PMID: 26481195 PMCID: PMC4612729 DOI: 10.1038/srep15365] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 09/22/2015] [Indexed: 01/26/2023] Open
Abstract
Connexin 43 (Cx43) functions as a cell growth suppressor. We have demonstrated that Cx43 interacts with heat shock cognate protein 70 (Hsc70) for regulating cell proliferation. Hsc70 interacts with CDK inhibitor p27, which regulates the assembly and subcellular localization of cyclin D1-CDK4-p27 complex. However, the involvement of p27 with Cx43-mediated cell cycle suppression is still poorly understood. Here, we report that nuclear accumulation of p27 is reduced by overexpression of Cx43, and that this reduction is restored by co-overexpression with Hsc70. We found that Cx43 competes with p27 for binding to Hsc70, and as a result, decreases the level of Hsc70 in cyclin D1-CDK4-p27 complex, leading to prevention of the nuclear translocation of the complex and the G1/S transition. Collectively, our findings suggest that, in Cx43 up-regulation, which is most likely an emergency measure, Cx43-Hsc70 interaction regulates cell cycle G1/S progression through a novel mechanism by which Cx43-Hsc70 interaction prevents the nuclear accumulation of p27 through controlling the nuclear translocation of cyclin D1-CDK4-p27 complex.
Collapse
|
24
|
Qiu X, Cheng JC, Klausen C, Chang HM, Fan Q, Leung PCK. EGF-Induced Connexin43 Negatively Regulates Cell Proliferation in Human Ovarian Cancer. J Cell Physiol 2015; 231:111-9. [DOI: 10.1002/jcp.25058] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 05/26/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Xin Qiu
- Department of Obstetrics and Gynaecology; Child & Family Research Institute; University of British Columbia; Vancouver British Columbia Canada
| | - Jung-Chien Cheng
- Department of Obstetrics and Gynaecology; Child & Family Research Institute; University of British Columbia; Vancouver British Columbia Canada
| | - Christian Klausen
- Department of Obstetrics and Gynaecology; Child & Family Research Institute; University of British Columbia; Vancouver British Columbia Canada
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology; Child & Family Research Institute; University of British Columbia; Vancouver British Columbia Canada
| | - Qianlan Fan
- Department of Obstetrics and Gynaecology; Child & Family Research Institute; University of British Columbia; Vancouver British Columbia Canada
| | - Peter C. K. Leung
- Department of Obstetrics and Gynaecology; Child & Family Research Institute; University of British Columbia; Vancouver British Columbia Canada
| |
Collapse
|
25
|
Poyet C, Buser L, Roudnicky F, Detmar M, Hermanns T, Mannhard D, Höhn A, Rüschoff J, Zhong Q, Sulser T, Moch H, Wild PJ. Connexin 43 expression predicts poor progression-free survival in patients with non-muscle invasive urothelial bladder cancer. J Clin Pathol 2015; 68:819-24. [PMID: 26251520 PMCID: PMC4602233 DOI: 10.1136/jclinpath-2015-202898] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/25/2015] [Indexed: 12/05/2022]
Abstract
Objectives To evaluate the protein expression of connexin 43 (Cx43) in primary urothelial bladder cancer and test its association with the histopathological characteristics and clinical outcome. Methods A tissue microarray containing 348 tissue samples from 174 patients with primary urothelial carcinomas of the bladder was immunohistochemically stained for Cx43. The intensity of staining was semiquantitatively evaluated (score 0, 1+, 2+), and the association with clinicopathological features was assessed. Univariable and multivariable analyses were performed to identify predictors for progression-free survival (PFS). Results Membranous Cx43 immunoreactivity was detected in 118 (67.8%) of 174 analysable urothelial carcinomas, of which 31 (17.8%) showed even a strong (score 2+) and mainly homogeneous staining. Strong expression levels of Cx43 (score 2+) were associated with higher tumour grade, multiplicity and increased proliferation (all p<0.05). In the subgroup of patients with stage pTa and pT1 bladder tumours (n=158), strong Cx43 expression (p<0.001), solid growth pattern (p<0.001) and increased Ki-67 proliferation fraction (p<0.05) were significantly associated with shorter PFS in an univariable Cox regression analysis. In multivariable Cox regression models, Cx43 immunoreactivity and histological growth pattern remained highly significant and adverse risk factors for PFS. Conclusions The expression levels of Cx43 are frequent in non-muscle invasive bladder cancer (NMIBC), with high expression levels being associated with poor prognosis. Routine assessment of Cx43 expression may improve the identification of high-risk NMIBC.
Collapse
Affiliation(s)
- Cédric Poyet
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Lorenz Buser
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Filip Roudnicky
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Doris Mannhard
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Andrej Höhn
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Jan Rüschoff
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Qing Zhong
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Holger Moch
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Peter J Wild
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
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
|
27
|
Tittarelli A, Guerrero I, Tempio F, Gleisner MA, Avalos I, Sabanegh S, Ortíz C, Michea L, López MN, Mendoza-Naranjo A, Salazar-Onfray F. Overexpression of connexin 43 reduces melanoma proliferative and metastatic capacity. Br J Cancer 2015; 113:259-67. [PMID: 26135897 PMCID: PMC4506378 DOI: 10.1038/bjc.2015.162] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/18/2015] [Accepted: 04/21/2015] [Indexed: 01/14/2023] Open
Abstract
Background: Alterations in connexin 43 (Cx43) expression and/or gap junction (GJ)-mediated intercellular communication are implicated in cancer pathogenesis. Herein, we have investigated the role of Cx43 in melanoma cell proliferation and apoptosis sensitivity in vitro, as well as metastatic capability and tumour growth in vivo. Methods: Connexin 43 expression levels, GJ coupling and proliferation rates were analysed in four different human melanoma cell lines. Furthermore, tumour growth and lung metastasis of high compared with low Cx43-expressing FMS cells were evaluated in vivo using a melanoma xenograft model. Results: Specific inhibition of Cx43 channel activity accelerated melanoma cell proliferation, whereas overexpression of Cx43 increased GJ coupling and reduced cell growth. Moreover, Cx43 overexpression in FMS cells increased basal and tumour necrosis factor-α-induced apoptosis and resulted in decreased melanoma tumour growth and lower number and size of metastatic foci in vivo. Conclusions: Our findings reveal an important role for Cx43 in intrinsically controlling melanoma growth, death and metastasis, and emphasise the potential use of compounds that selectively enhance Cx43 expression on melanoma in the future chemotherapy and/or immunotherapy protocols.
Collapse
Affiliation(s)
- A Tittarelli
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - I Guerrero
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - F Tempio
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - M A Gleisner
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - I Avalos
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - S Sabanegh
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - C Ortíz
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - L Michea
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| | - M N López
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile [3] Research Support Office, University of Chile Clinical Hospital, Santiago 8380453, Chile
| | - A Mendoza-Naranjo
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - F Salazar-Onfray
- 1] Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile [2] Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
28
|
Grek CL, Rhett JM, Bruce JS, Abt MA, Ghatnekar GS, Yeh ES. Targeting connexin 43 with α-connexin carboxyl-terminal (ACT1) peptide enhances the activity of the targeted inhibitors, tamoxifen and lapatinib, in breast cancer: clinical implication for ACT1. BMC Cancer 2015; 15:296. [PMID: 25881004 PMCID: PMC4407347 DOI: 10.1186/s12885-015-1229-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/19/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Treatment failure is a critical issue in breast cancer and identifying useful interventions that optimize current cancer therapies remains a critical unmet need. Expression and functional studies have identified connexins (Cxs), a family of gap junction proteins, as potential tumor suppressors. Studies suggest that Cx43 has a role in breast cancer cell proliferation, differentiation, and migration. Although pan-gap junction drugs are available, the lack of specificity of these agents increases the opportunity for off target effects. Consequently, a therapeutic agent that specifically modulates Cx43 would be beneficial and has not been tested in breast cancer. In this study, we now test an agent that specifically targets Cx43, called ACT1, in breast cancer. METHODS We evaluated whether direct modulation of Cx43 using a Cx43-directed therapeutic peptide, called ACT1, enhances Cx43 gap junctional activity in breast cancer cells, impairs breast cancer cell proliferation or survival, and enhances the activity of the targeted inhibitors tamoxifen and lapatinib. RESULTS Our results show that therapeutic modulation of Cx43 by ACT1 maintains Cx43 at gap junction sites between cell-cell membrane borders of breast cancer cells and augments gap junction activity in functional assays. The increase in Cx43 gap junctional activity achieved by ACT1 treatment impairs proliferation or survival of breast cancer cells but ACT1 has no effect on non-transformed MCF10A cells. Furthermore, treating ER+ breast cancer cells with a combination of ACT1 and tamoxifen or HER2+ breast cancer cells with ACT1 and lapatinib augments the activity of these targeted inhibitors. CONCLUSIONS Based on our findings, we conclude that modulation of Cx43 activity in breast cancer can be effectively achieved with the agent ACT1 to sustain Cx43-mediated gap junctional activity resulting in impaired malignant progression and enhanced activity of lapatinib and tamoxifen, implicating ACT1 as part of a combination regimen in breast cancer.
Collapse
Affiliation(s)
- Christina L Grek
- FirstString Research, Inc., 300 W. Coleman Blvd., Suite 203, Mount Pleasant, SC, USA.
| | - Joshua Matthew Rhett
- Department of Surgery, Division of General Surgery, Medical University of South Carolina, Charleston, SC, USA.
| | - Jaclynn S Bruce
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Ave, BSB358, MSC509, Charleston, SC, 29425, USA.
| | - Melissa A Abt
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Ave, BSB358, MSC509, Charleston, SC, 29425, USA.
| | - Gautam S Ghatnekar
- FirstString Research, Inc., 300 W. Coleman Blvd., Suite 203, Mount Pleasant, SC, USA.
| | - Elizabeth S Yeh
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Ave, BSB358, MSC509, Charleston, SC, 29425, USA.
| |
Collapse
|
29
|
Kelsey L, Katoch P, Ray A, Mitra S, Chakraborty S, Lin MF, Mehta PP. Vitamin D3 regulates the formation and degradation of gap junctions in androgen-responsive human prostate cancer cells. PLoS One 2014; 9:e106437. [PMID: 25188420 PMCID: PMC4154685 DOI: 10.1371/journal.pone.0106437] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 08/06/2014] [Indexed: 11/19/2022] Open
Abstract
1α-25(OH)2 vitamin D3 (1-25D), an active hormonal form of Vitamin D3, is a well-known chemopreventive and pro-differentiating agent. It has been shown to inhibit the growth of several prostate cancer cell lines. Gap junctions, formed of proteins called connexins (Cx), are ensembles of cell-cell channels, which permit the exchange of small growth regulatory molecules between adjoining cells. Cell-cell communication mediated by gap junctional channels is an important homeostatic control mechanism for regulating cell growth and differentiation. We have investigated the effect of 1-25D on the formation and degradation of gap junctions in an androgen-responsive prostate cancer cell line, LNCaP, which expresses retrovirally-introduced Cx32. Connexin32 is expressed by the luminal and well-differentiated cells of normal prostate and prostate tumors. Our results document that 1-25D enhances the expression of Cx32 and its subsequent assembly into gap junctions. Our results further show that 1-25D prevents androgen-regulated degradation of Cx32, post-translationally, independent of androgen receptor (AR)-mediated signaling. Finally, our findings document that formation of gap junctions sensitizes Cx32-expressing LNCaP cells to the growth inhibitory effects of 1-25D and alters their morphology. These findings suggest that the growth-inhibitory effects of 1-25D in LNCaP cells may be related to its ability to modulate the assembly of Cx32 into gap junctions.
Collapse
Affiliation(s)
- Linda Kelsey
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Parul Katoch
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Anuttoma Ray
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shalini Mitra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Souvik Chakraborty
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Parmender P. Mehta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
30
|
Connexin43 functions as a novel interacting partner of heat shock cognate protein 70. Sci Rep 2014; 3:2719. [PMID: 24056538 PMCID: PMC3779846 DOI: 10.1038/srep02719] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/04/2013] [Indexed: 01/12/2023] Open
Abstract
Regulation of connexin43 (Cx43) expression affects cell proliferation, differentiation and apoptosis in a gap junctional intercellular communication (GJIC)-independent manner. However, the underlying mechanisms of Cx43-mediated cell cycle suppression are still poorly understood. To elucidate the molecular mechanism of Cx43-mediated cell cycle suppression, we searched for Cx43 interacting proteins by using a proteomics approach. Here, we have identified a Cx43-interacting protein, heat shock cognate protein 70 (Hsc70). We confirmed that Hsc70 directly binds to the C-terminus of Cx43, whereas Hsc54, a splice variant of Hsc70, does not, that Cx43 competes with cyclin D1 for binding to Hsc70, and that the nuclear accumulation of cyclin D1 is reduced by overexpression of Cx43 in a GJIC-independent manner, which is restored by co-overexpression with Hsc70. As a result, the cell proliferation is regulated by Cx43. Our results suggest that Cx43-Hsc70 interaction probably plays a critical role during G1/S progression.
Collapse
|
31
|
Koval M, Molina SA, Burt JM. Mix and match: investigating heteromeric and heterotypic gap junction channels in model systems and native tissues. FEBS Lett 2014; 588:1193-204. [PMID: 24561196 DOI: 10.1016/j.febslet.2014.02.025] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 02/12/2014] [Accepted: 02/13/2014] [Indexed: 12/12/2022]
Abstract
This review is based in part on a roundtable discussion session: "Physiological roles for heterotypic/heteromeric channels" at the 2013 International Gap Junction Conference (IGJC 2013) in Charleston, South Carolina. It is well recognized that multiple connexins can specifically co-assemble to form mixed gap junction channels with unique properties as a means to regulate intercellular communication. Compatibility determinants for both heteromeric and heterotypic gap junction channel formation have been identified and associated with specific connexin amino acid motifs. Hetero-oligomerization is also a regulated process; differences in connexin quality control and monomer stability are likely to play integral roles to control interactions between compatible connexins. Gap junctions in oligodendrocyte:astrocyte communication and in the cardiovascular system have emerged as key systems where heterotypic and heteromeric channels have unique physiologic roles. There are several methodologies to study heteromeric and heterotypic channels that are best applied to either heterologous expression systems, native tissues or both. There remains a need to use and develop different experimental approaches in order to understand the prevalence and roles for mixed gap junction channels in human physiology.
Collapse
Affiliation(s)
- Michael Koval
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA, United States; Department of Cell Biology, Emory University, Atlanta, GA, United States.
| | - Samuel A Molina
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Janis M Burt
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Nelson TK, Sorgen PL, Burt JM. Carboxy terminus and pore-forming domain properties specific to Cx37 are necessary for Cx37-mediated suppression of insulinoma cell proliferation. Am J Physiol Cell Physiol 2013; 305:C1246-56. [PMID: 24133065 PMCID: PMC3882364 DOI: 10.1152/ajpcell.00159.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/15/2013] [Indexed: 11/22/2022]
Abstract
Connexin 37 (Cx37) suppresses cell proliferation when expressed in rat insulinoma (Rin) cells, an effect also manifest in vivo during vascular development and in response to tissue injury. Mutant forms of Cx37 with nonfunctional channels but normally localized, wild-type carboxy termini are not growth suppressive. Here we determined whether the carboxy-terminal (CT) domain is required for Cx37-mediated growth suppression and whether the Cx37 pore-forming domain can be replaced with the Cx43 pore-forming domain and still retain growth-suppressive properties. We show that despite forming functional gap junction channels and hemichannels, Cx37 with residues subsequent to 273 replaced with a V5-epitope tag (Cx37-273tr*V5) had no effect on the proliferation of Rin cells, did not facilitate G1-cell cycle arrest with serum deprivation, and did not prolong cell cycle time comparably to the wild-type protein. The chimera Cx43*CT37, comprising the pore-forming domain of Cx43 and CT of Cx37, also did not suppress proliferation, despite forming functional gap junctions with a permselective profile similar to wild-type Cx37. Differences in channel behavior of both Cx37-273tr*V5 and Cx43*CT37 relative to their wild-type counterparts and failure of the Cx37-CT to interact as the Cx43-CT does with the Cx43 cytoplasmic loop suggest that the Cx37-CT and pore-forming domains are both essential to growth suppression by Cx37.
Collapse
Affiliation(s)
- Tasha K Nelson
- Department of Physiology, University of Arizona, Tucson, Arizona; and
| | | | | |
Collapse
|
35
|
Haslam IS, Roubos EW, Mangoni ML, Yoshizato K, Vaudry H, Kloepper JE, Pattwell DM, Maderson PFA, Paus R. From frog integument to human skin: dermatological perspectives from frog skin biology. Biol Rev Camb Philos Soc 2013; 89:618-55. [DOI: 10.1111/brv.12072] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 10/03/2013] [Accepted: 10/22/2013] [Indexed: 12/15/2022]
Affiliation(s)
- Iain S. Haslam
- The Dermatology Centre, Salford Royal NHS Foundation Trust, Institute of Inflammation and Repair; University of Manchester; Oxford Road Manchester M13 9PT U.K
| | - Eric W. Roubos
- Department of Anatomy; Radboud University Medical Centre; Geert Grooteplein Noord 2, 6525 EZ, Nijmegen P.O. Box 9101, 6500 HB Nijmegen The Netherlands
| | - Maria Luisa Mangoni
- Department of Biochemical Sciences, Istituto Pasteur-Fondazione Cenci Bolognetti; La Sapienza University of Rome, Piazzale Aldo Moro, 5-00185; Rome Italy
| | - Katsutoshi Yoshizato
- Academic Advisors Office, Synthetic Biology Research Center; Osaka City University Graduate School of Medicine; Osaka Japan
- Phoenixbio Co. Ltd; 3-4-1, Kagamiyama; Higashihiroshima Hiroshima 739-0046 Japan
| | - Hubert Vaudry
- European Institute for Peptide Research; University of Rouen; Mont-Saint-Aignan Place Emile Blondel 76821 France
- INSERM U-982, CNRS; University of Rouen; Mont-Saint-Aignan Place Emile Blondel 76821 France
| | - Jennifer E. Kloepper
- Klinik für Dermatologie, Allergologie und Venerologie; Universitätsklinikum Schleswig-Holstein, Ratzeburger Allee 160; 23538 Lübeck Germany
| | - David M. Pattwell
- Leahurst Campus, Institute of Learning & Teaching; School of Veterinary Science, University of Liverpool; Neston CH64 7TE U.K
| | | | - Ralf Paus
- The Dermatology Centre, Salford Royal NHS Foundation Trust, Institute of Inflammation and Repair; University of Manchester; Oxford Road Manchester M13 9PT U.K
- Klinik für Dermatologie, Allergologie und Venerologie; Universitätsklinikum Schleswig-Holstein, Ratzeburger Allee 160; 23538 Lübeck Germany
| |
Collapse
|
36
|
Ableser MJ, Penuela S, Lee J, Shao Q, Laird DW. Connexin43 reduces melanoma growth within a keratinocyte microenvironment and during tumorigenesis in vivo. J Biol Chem 2013; 289:1592-603. [PMID: 24297173 DOI: 10.1074/jbc.m113.507228] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Connexins (Cx) have been identified as tumor suppressors or enhancers, a distinction that appears to be dependent on the type and stage of disease. However, the role of connexins in melanoma tumorigenesis and their status during cancer onset and progression remain controversial and unclear. Here, we show that the aggressive B16-BL6 mouse melanoma cell line expresses low basal levels of Cx26 and Cx43, rendering them gap junctional intercellular communication-deficient as elucidated by immunofluorescence, Western blotting, and dye transfer studies. Following ectopic expression of green fluorescent protein-tagged Cx26 and Cx43 in these connexin-deficient melanomas, punctate gap junction-like plaques were evident at sites of cell-cell apposition, and the incidence of dye transfer was significantly increased similar to connexin-rich keratinocytes. We found that the expression of Cx43, but not Cx26, significantly reduced cellular proliferation and anchorage-independent growth from control melanomas, whereas migration was unaffected. Additionally, melanomas expressing Cx43 displayed significantly reduced growth within the in situ-like microenvironment of keratinocytes, despite a lack of heterocellular gap junctional intercellular communication between the two cell types. Furthermore, when grown in vivo in the chicken chorioallantoic membrane, primary tumors derived from Cx43-expressing melanomas were significantly smaller than controls, whereas Cx26-expressing melanomas produced tumors similar to controls. Collectively, these results suggest that Cx43, and not Cx26, can act as a tumor suppressor during melanoma tumorigenesis.
Collapse
|
37
|
Tang B, Peng ZH, Yu PW, Yu G, Qian F, Zeng DZ, Zhao YL, Shi Y, Hao YX, Luo HX. Aberrant expression of Cx43 is associated with the peritoneal metastasis of gastric cancer and Cx43-mediated gap junction enhances gastric cancer cell diapedesis from peritoneal mesothelium. PLoS One 2013; 8:e74527. [PMID: 24040271 PMCID: PMC3770585 DOI: 10.1371/journal.pone.0074527] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 08/06/2013] [Indexed: 12/24/2022] Open
Abstract
The process of peritoneal metastasis involves the diapedesis of intra-abdominal exfoliated gastric cancer cells through the mesothelial cell monolayers; however, the related molecular mechanisms for this process are still unclear. Heterocellular gap-junctional intercellular communication (GJIC) between gastric cancer cells and mesothelial cells may play an active role during diapedesis. In this study we detected the expression of connexin 43 (Cx43) in primary gastric cancer tissues, intra-abdominal exfoliated cancer cells, and matched metastatic peritoneal tissues. We found that the expression of Cx43 in primary gastric cancer tissues was significantly decreased; the intra-abdominal exfoliated cancer cells and matched metastatic peritoneal tissues exhibited increasing expression compared with primary gastric cancer tissues. BGC-823 and SGC-7901 human gastric cancer cells were engineered to express Cx43 or Cx43T154A (a mutant protein that only couples gap junctions but provides no intercellular communication) and were co-cultured with human peritoneal mesothelial cells (HPMCs). Heterocellular GJIC and diapedesis through HPMC monolayers on matrigel-coated coverslips were investigated. We found that BGC-823 and SGC-7901 gastric cancer cells expressing Cx43 formed functional heterocellular gap junctions with HPMC monolayers within one hour. A significant increase in diapedesis was observed in engineered Cx43-expressing cells compared with Cx43T154A and control group cells, which suggested that the observed upregulation of diapedesis in Cx43-expressing cells required heterocellular GJIC. Further study revealed that the gastric cancer cells transmigrated through the intercellular space between the mesothelial cells via a paracellular route. Our results suggest that the abnormal expression of Cx43 plays an essential role in peritoneal metastasis and that Cx43-mediated heterocellular GJIC between gastric cancer cells and mesothelial cells may be an important regulatory step during metastasis. Finally, we observed that the diapedesis of exfoliated gastric cancer cells through mesothelial barriers is a viable route of paracellular migration.
Collapse
Affiliation(s)
- Bo Tang
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
| | - Zhi-hong Peng
- Institute of Digestive Diseases, Southwest Hospital, the Third Military Medical University, Chongqing, China
| | - Pei-Wu Yu
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
- * E-mail:
| | - Ge Yu
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
| | - Feng Qian
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
| | - Dong-zhu Zeng
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
| | - Yong-liang Zhao
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
| | - Yan Shi
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
| | - Ying-xue Hao
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
| | - Hua-xing Luo
- General Surgery Center of PLA, Southwest Hospital, the Third Military Medical University, Chongqing, China
| |
Collapse
|
38
|
Mancuso M, Leonardi S, Giardullo P, Pasquali E, Tanori M, De Stefano I, Casciati A, Naus CC, Pazzaglia S, Saran A. Oncogenic radiation abscopal effects in vivo: interrogating mouse skin. Int J Radiat Oncol Biol Phys 2013; 86:993-9. [PMID: 23755921 DOI: 10.1016/j.ijrobp.2013.04.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 04/16/2013] [Accepted: 04/19/2013] [Indexed: 11/26/2022]
Abstract
PURPOSE To investigate the tissue dependence in transmission of abscopal radiation signals and their oncogenic consequences in a radiosensitive mouse model and to explore the involvement of gap junction intercellular communication (GJIC) in mediating radiation tumorigenesis in off-target mouse skin. METHODS AND MATERIALS Patched1 heterozygous (Ptch1(+/-)) mice were irradiated at postnatal day 2 (P2) with 10 Gy of x-rays. Individual lead cylinders were used to protect the anterior two-thirds of the body, whereas the hindmost part was directly exposed to radiation. To test the role of GJICs and their major constituent connexin43 (Cx43), crosses between Ptch1(+/-) and Cx43(+/-) mice were similarly irradiated. These mouse groups were monitored for their lifetime, and skin basal cell carcinomas (BCCs) were counted and recorded. Early responses to DNA damage - Double Strand Breaks (DSBs) and apoptosis - were also evaluated in shielded and directly irradiated skin areas. RESULTS We report abscopal tumor induction in the shielded skin of Ptch1(+/-) mice after partial-body irradiation. Endpoints were induction of early nodular BCC-like tumors and macroscopic infiltrative BCCs. Abscopal tumorigenesis was significantly modulated by Cx43 status, namely, Cx43 reduction was associated with decreased levels of DNA damage and oncogenesis in out-of-field skin, suggesting a key role of GJIC in transmission of oncogenic radiation signals to unhit skin. CONCLUSIONS Our results further characterize the nature of abscopal responses and the implications they have on pathologic processes in different tissues, including their possible underlying mechanistic bases.
Collapse
Affiliation(s)
- Mariateresa Mancuso
- Laboratory of Radiation Biology and Biomedicine, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), Casaccia Research Centre, Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Clarke LA, Sousa L, Barreto C, Amaral MD. Changes in transcriptome of native nasal epithelium expressing F508del-CFTR and intersecting data from comparable studies. Respir Res 2013; 14:38. [PMID: 23537407 PMCID: PMC3637641 DOI: 10.1186/1465-9921-14-38] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 03/07/2013] [Indexed: 01/06/2023] Open
Abstract
Background Microarray studies related to cystic fibrosis (CF) airway gene expression have gone some way in clarifying the complex molecular background of CF lung diseases, but have made little progress in defining a robust “molecular signature” associated with mutant CFTR expression. Disparate methodological and statistical analyses complicate comparisons between independent studies of the CF transcriptome, and although each study may be valid in isolation, the conclusions reached differ widely. Methods We carried out a small-scale whole genome microarray study of gene expression in human native nasal epithelial cells from F508del-CFTR homozygotes in comparison to non-CF controls. We performed superficial comparisons with other microarray datasets in an attempt to identify a subset of regulated genes that could act as a signature of F508del-CFTR expression in native airway tissue samples. Results Among the alterations detected in CF, up-regulation of genes involved in cell proliferation, and down-regulation of cilia genes were the most notable. Other changes involved gene expression changes in calcium and membrane pathways, inflammation, defence response, wound healing and the involvement of estrogen signalling. Comparison of our data set with previously published studies allowed us to assess the consistency of independent microarray data sets, and shed light on the limitations of such snapshot studies in measuring a system as subtle and dynamic as the transcriptome. Comparison of in-vivo studies nevertheless yielded a small molecular CF signature worthy of future investigation. Conclusions Despite the variability among the independent studies, the current CF transcriptome meta-analysis identified subsets of differentially expressed genes in native airway tissues which provide both interesting clues to CF pathogenesis and a possible CF biomarker.
Collapse
Affiliation(s)
- Luka A Clarke
- BioFIG-Centre for Biodiversity, Functional and Integrative Genomics, FCUL-Faculty of Sciences, University of Lisboa, Lisboa 1749-016, Portugal.
| | | | | | | |
Collapse
|
40
|
Zhao JQ, Sun FJ, Liu SS, Yang J, Wu YQ, Li GS, Chen QY, Wang JX. Expression of Connexin 43 and E-cadherin Protein and mRNA in Non-small Cell Lung Cancers in Chinese Patients. Asian Pac J Cancer Prev 2013; 14:639-43. [DOI: 10.7314/apjcp.2013.14.2.639] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
41
|
Ke Q, Li L, Cai B, Liu C, Yang Y, Gao Y, Huang W, Yuan X, Wang T, Zhang Q, Harris AL, Tao L, Xiang AP. Connexin 43 is involved in the generation of human-induced pluripotent stem cells. Hum Mol Genet 2013; 22:2221-33. [PMID: 23420013 DOI: 10.1093/hmg/ddt074] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Although somatic cells can be successfully programmed to create pluripotent stem cells by ectopically expressing defined transcriptional factors, reprogramming efficiency is low and the reprogramming mechanism remains unclear. Previous reports have shown that almost all human connexin (CX) isoforms are expressed by human embryonic stem (hES) cells and that gap junctional intercellular communication (GJIC) is important for ES cell survival and differentiation. However, the CX expression profiles in human induced pluripotent stem (iPS) cells and the role of CXs in the process of reprogramming back to iPS cells remains unknown. Here, we determined the expression levels of most forms of CX in human embryonic fibroblasts (hEFs) and in the hEF-derived iPS cells. A scrape loading/dye transfer assay showed that human iPS cells contained functional gap junctions (GJs) that could be affected by pharmacological inhibitors of GJ function. We found that CX43 was the most dramatically upregulated CX following reprogramming. Most importantly, the ectopic expression of CX43 significantly enhanced the reprogramming efficiency, whereas shRNA-mediated knockdown of endogenous CX43 expression greatly reduced the efficiency. In addition, we found that CX43 overexpression or knockdown affected the expression of E-CADHERIN, a marker of the mesenchymal-to-epithelial transition (MET), during reprogramming. In conclusion, our data indicate that CX43 expression is important for reprogramming and may mediate the MET that is associated with the acquisition of pluripotency.
Collapse
Affiliation(s)
- Qiong Ke
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Stoletov K, Strnadel J, Zardouzian E, Momiyama M, Park FD, Kelber JA, Pizzo DP, Hoffman R, VandenBerg SR, Klemke RL. Role of connexins in metastatic breast cancer and melanoma brain colonization. J Cell Sci 2013; 126:904-13. [PMID: 23321642 DOI: 10.1242/jcs.112748] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Breast cancer and melanoma cells commonly metastasize to the brain using homing mechanisms that are poorly understood. Cancer patients with brain metastases display poor prognosis and survival due to the lack of effective therapeutics and treatment strategies. Recent work using intravital microscopy and preclinical animal models indicates that metastatic cells colonize the brain, specifically in close contact with the existing brain vasculature. However, it is not known how contact with the vascular niche promotes microtumor formation. Here, we investigate the role of connexins in mediating early events in brain colonization using transparent zebrafish and chicken embryo models of brain metastasis. We provide evidence that breast cancer and melanoma cells utilize connexin gap junction proteins (Cx43, Cx26) to initiate brain metastatic lesion formation in association with the vasculature. RNAi depletion of connexins or pharmacological blocking of connexin-mediated cell-cell communication with carbenoxolone inhibited brain colonization by blocking tumor cell extravasation and blood vessel co-option. Activation of the metastatic gene twist in breast cancer cells increased Cx43 protein expression and gap junction communication, leading to increased extravasation, blood vessel co-option and brain colonization. Conversely, inhibiting twist activity reduced Cx43-mediated gap junction coupling and brain colonization. Database analyses of patient histories revealed increased expression of Cx26 and Cx43 in primary melanoma and breast cancer tumors, respectively, which correlated with increased cancer recurrence and metastasis. Together, our data indicate that Cx43 and Cx26 mediate cancer cell metastasis to the brain and suggest that connexins might be exploited therapeutically to benefit cancer patients with metastatic disease.
Collapse
Affiliation(s)
- Konstantin Stoletov
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cav1 suppresses tumor growth and metastasis in a murine model of cutaneous SCC through modulation of MAPK/AP-1 activation. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 182:992-1004. [PMID: 23267770 DOI: 10.1016/j.ajpath.2012.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 10/03/2012] [Accepted: 11/08/2012] [Indexed: 01/17/2023]
Abstract
Caveolin-1 (Cav1) is a scaffolding protein that serves to regulate the activity of several signaling molecules. Its loss has been implicated in the pathogenesis of several types of cancer, but its role in the development and progression of cutaneous squamous cell carcinoma (cSCC) remains largely unexplored. Herein, we use the keratinocyte cell line PAM212, a murine model of cSCC, to determine the function of Cav1 in skin tumor biology. We first show that Cav1 overexpression decreases cell and tumor growth, whereas Cav1 knockdown increases these attributes in PAM212 cells. In addition, Cav1 knockdown increases the invasive ability and incidence of spontaneous lymph node metastasis. Finally, we demonstrate that Cav1 knockdown increases extracellular signaling-related kinase 1/2 mitogen-activated protein kinase/activator protein-1 pathway activation. We attribute the growth and invasive advantage conferred by Cav1 knockdown to increased expression of activator protein-1 transcriptional targets, including cyclin D1 and keratin 18, which show inverse expression in PAM212 based on the expression level of Cav1. In summary, we demonstrate that loss of Cav1 affects several characteristics associated with aggressive human skin tumors and that this protein may be an important modulator of tumor growth and invasion in cSCC.
Collapse
|
44
|
Cowan KN, Langlois S, Penuela S, Cowan BJ, Laird DW. Pannexin1 and Pannexin3 exhibit distinct localization patterns in human skin appendages and are regulated during keratinocyte differentiation and carcinogenesis. ACTA ACUST UNITED AC 2012; 19:45-53. [PMID: 22947051 DOI: 10.3109/15419061.2012.712575] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Having shown that Panx1 and Panx3 are expressed in the epidermis, we investigated their distribution in human skin adnexal structures and skin cancer. Both proteins were found in hair follicles, sebaceous and eccrine glands, as well as blood vessels. Panx1 was detected as punctate or diffuse intracellular labeling, while Panx3 was only observed as diffuse intracellular staining, suggesting different functions. We also identified the Panx3 immunoreactive ~70 kD species modulated during keratinocyte differentiation as Panx3. Since our data indicate that pannexins are regulated during keratinocyte differentiation, we assessed whether their levels are altered under circumstances in which keratinocyte differentiation is compromised. We found that Panx1 and Panx3 levels are highly reduced in human keratinocyte tumors, thus showing for the first time that both pannexins are dysregulated in human cancers. Altogether, these data suggest that Panx1 and Panx3 have distinct and unique functions within the skin in health and disease.
Collapse
Affiliation(s)
- Kyle N Cowan
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Canada.
| | | | | | | | | |
Collapse
|
45
|
Degradation of connexins through the proteasomal, endolysosomal and phagolysosomal pathways. J Membr Biol 2012; 245:389-400. [PMID: 22772442 DOI: 10.1007/s00232-012-9461-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 06/20/2012] [Indexed: 01/23/2023]
Abstract
Connexins comprise gap junction channels, which create a direct conduit between the cytoplasms of adjacent cells and provide for intercellular communication. Therefore, the level of total cellular connexin protein can have a direct influence on the level of intercellular communication. Control of connexin protein levels can occur through different mechanisms during the connexin life cycle, such as by regulation of connexin gene expression and turnover of existing protein. The degradation of connexins has been extensively studied, revealing proteasomal, endolysosomal and more recently autophagosomal degradation mechanisms that modulate connexin turnover and, subsequently, affect intercellular communication. Here, we review the current knowledge of connexin degradation pathways.
Collapse
|
46
|
SUN YAN, ZHAO XIAOPING, YAO YUTING, QI XINYI, YUAN YAOZONG, HU YU. Connexin 43 interacts with Bax to regulate apoptosis of pancreatic cancer through a gap junction-independent pathway. Int J Oncol 2012; 41:941-8. [DOI: 10.3892/ijo.2012.1524] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Accepted: 04/24/2012] [Indexed: 11/05/2022] Open
|
47
|
Yu SC, Xiao HL, Jiang XF, Wang QL, Li Y, Yang XJ, Ping YF, Duan JJ, Jiang JY, Ye XZ, Xu SL, Xin YH, Yao XH, Chen JH, Chu WH, Sun W, Wang B, Wang JM, Zhang X, Bian XW. Connexin 43 reverses malignant phenotypes of glioma stem cells by modulating E-cadherin. Stem Cells 2012; 30:108-20. [PMID: 22131169 DOI: 10.1002/stem.1685] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Malfunctioned gap junctional intercellular communication (GJIC) has been thought associated with malignant transformation of normal cells. However, the role of GJIC-related proteins such as connexins in sustaining the malignant behavior of cancer stem cells remains unclear. In this study, we obtained tumorspheres formed by glioma stem cells (GSCs) and adherent GSCs and then examined their GJIC. All GSCs showed reduced GJIC, and differentiated glioma cells had more gap junction-like structures than GSCs. GSCs expressed very low level of connexins, Cx43 in particular, which are key components of gap junction. We observed hypermethylation in the promoter of gap junction protein α1, which encodes Cx43 in GSCs. Reconstitution of Cx43 in GSCs inhibited their capacity of self-renewal, invasiveness, and tumorigenicity via influencing E-cadherin and its coding protein, which leads to changes in the expression of Wnt/β-catenin targeting genes. Our results suggest that GSCs require the low expression of Cx43 for maintaining their malignant phenotype, and upregulation of Cx43 might be a potential strategy for treatment of malignant glioma.
Collapse
Affiliation(s)
- Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lazarowski ER. Vesicular and conductive mechanisms of nucleotide release. Purinergic Signal 2012; 8:359-73. [PMID: 22528679 DOI: 10.1007/s11302-012-9304-9] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 01/21/2012] [Indexed: 12/22/2022] Open
Abstract
Extracellular nucleotides and nucleosides promote a vast range of physiological responses, via activation of cell surface purinergic receptors. Virtually all tissues and cell types exhibit regulated release of ATP, which, in many cases, is accompanied by the release of uridine nucleotides. Given the relevance of extracellular nucleotide/nucleoside-evoked responses, understanding how ATP and other nucleotides are released from cells is an important physiological question. By facilitating the entry of cytosolic nucleotides into the secretory pathway, recently identified vesicular nucleotide and nucleotide-sugar transporters contribute to the exocytotic release of ATP and UDP-sugars not only from endocrine/exocrine tissues, but also from cell types in which secretory granules have not been biochemically characterized. In addition, plasma membrane connexin hemichannels, pannexin channels, and less-well molecularly defined ATP conducting anion channels have been shown to contribute to the release of ATP (and UTP) under a variety of conditions.
Collapse
Affiliation(s)
- Eduardo R Lazarowski
- School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7248, USA.
| |
Collapse
|
49
|
Prade E, Tobiasch M, Hitkova I, Schäffer I, Lian F, Xing X, Tänzer M, Rauser S, Walch A, Feith M, Post S, Röcken C, Schmid RM, Ebert MPA, Burgermeister E. Bile acids down-regulate caveolin-1 in esophageal epithelial cells through sterol responsive element-binding protein. Mol Endocrinol 2012; 26:819-32. [PMID: 22474125 DOI: 10.1210/me.2011-1140] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bile acids are synthesized from cholesterol and are major risk factors for Barrett adenocarcinoma (BAC) of the esophagus. Caveolin-1 (Cav1), a scaffold protein of membrane caveolae, is transcriptionally regulated by cholesterol via sterol-responsive element-binding protein-1 (SREBP1). Cav1 protects squamous epithelia by controlling cell growth and stabilizing cell junctions and matrix adhesion. Cav1 is frequently down-regulated in human cancers; however, the molecular mechanisms that lead to this event are unknown. We show that the basal layer of the nonneoplastic human esophageal squamous epithelium expressed Cav1 mainly at intercellular junctions. In contrast, Cav1 was lost in 95% of tissue specimens from BAC patients (n = 100). A strong cytoplasmic expression of Cav1 correlated with poor survival in a small subgroup (n = 5) of BAC patients, and stable expression of an oncogenic Cav1 variant (Cav1-P132L) in the human BAC cell line OE19 promoted proliferation. Cav1 was also detectable in immortalized human squamous epithelial, Barrett esophagus (CPC), and squamous cell carcinoma cells (OE21), but was low in BAC cell lines (OE19, OE33). Mechanistically, bile acids down-regulated Cav1 expression by inhibition of the proteolytic cleavage of 125-kDa pre-SREBP1 from the endoplasmic reticulum/Golgi apparatus and nuclear translocation of active 68-kDa SREBP1. This block in SREBP1's posttranslational processing impaired transcriptional activation of SREBP1 response elements in the proximal human Cav1 promoter. Cav1 was also down-regulated in esophagi from C57BL/6 mice on a diet enriched with 1% (wt/wt) chenodeoxycholic acid. Mice deficient for Cav1 or the nuclear bile acid receptor farnesoid X receptor showed hyperplasia and hyperkeratosis of the basal cell layer of esophageal epithelia, respectively. These data indicate that bile acid-mediated down-regulation of Cav1 marks early changes in the squamous epithelium, which may contribute to onset of Barrett esophagus metaplasia and progression to BAC.
Collapse
Affiliation(s)
- Elke Prade
- Department of Chemistry, Klinikum rechts der Isar, Technische Universität München, D-81675 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
WANG XH, ZHENG YM, CUI YQ, LIU S, SUN HC, LI F. The Inhibition Effect of Caveolin-1 on PANC1 Human Pancreatic Tumor Growth <I>In vitro</I> and <I>In vivo</I>*. PROG BIOCHEM BIOPHYS 2012. [DOI: 10.3724/sp.j.1206.2011.00168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|