1
|
Pelaz SG, Flores-Hernández R, Vujic T, Schvartz D, Álvarez-Vázquez A, Ding Y, García-Vicente L, Belloso A, Talaverón R, Sánchez JC, Tabernero A. A proteomic approach supports the clinical relevance of TAT-Cx43 266-283 in glioblastoma. Transl Res 2024; 272:95-110. [PMID: 38876188 DOI: 10.1016/j.trsl.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/18/2024] [Accepted: 06/01/2024] [Indexed: 06/16/2024]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary brain cancer. The Src inhibitor, TAT-Cx43266-283, exerts antitumor effects in in vitro and in vivo models of GBM. Because addressing the mechanism of action is essential to translate these results to a clinical setting, in this study we carried out an unbiased proteomic approach. Data-independent acquisition mass spectrometry proteomics allowed the identification of 190 proteins whose abundance was modified by TAT-Cx43266-283. Our results were consistent with the inhibition of Src as the mechanism of action of TAT-Cx43266-283 and unveiled antitumor effectors, such as p120 catenin. Changes in the abundance of several proteins suggested that TAT-Cx43266-283 may also impact the brain microenvironment. Importantly, the proteins whose abundance was reduced by TAT-Cx43266-283 correlated with an improved GBM patient survival in clinical datasets and none of the proteins whose abundance was increased by TAT-Cx43266-283 correlated with shorter survival, supporting its use in clinical trials.
Collapse
Affiliation(s)
- Sara G Pelaz
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain.
| | - Raquel Flores-Hernández
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Tatjana Vujic
- Department of Medicine, University of Geneva, 1211, Geneva, Switzerland; University Center of Legal Medicine, Lausanne-Geneva, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne Geneva, Switzerland
| | - Domitille Schvartz
- Department of Medicine, University of Geneva, 1211, Geneva, Switzerland; University of Geneva, Faculty of Medicine, Proteomics Core Facility, Geneva, Switzerland
| | - Andrea Álvarez-Vázquez
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Yuxin Ding
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Laura García-Vicente
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Aitana Belloso
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Rocío Talaverón
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | | | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain.
| |
Collapse
|
2
|
Álvarez-Vázquez A, San-Segundo L, Cerveró-García P, Flores-Hernández R, Ollauri-Ibáñez C, Segura-Collar B, Hubert CG, Morrison G, Pollard SM, Lathia JD, Sánchez-Gómez P, Tabernero A. EGFR amplification and EGFRvIII predict and participate in TAT-Cx43266-283 antitumor response in preclinical glioblastoma models. Neuro Oncol 2024; 26:1230-1246. [PMID: 38507464 PMCID: PMC11226870 DOI: 10.1093/neuonc/noae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) commonly displays epidermal growth factor receptor (EGFR) alterations (mainly amplification and EGFRvIII) and TAT-Cx43266-283 is a Src-inhibitory peptide with antitumor properties in preclinical GBM models. Given the link between EGFR and Src, the aim of this study was to explore the role of EGFR in the antitumor effects of TAT-Cx43266-283. METHODS The effect of TAT-Cx43266-283, temozolomide (TMZ), and erlotinib (EGFR inhibitor) was studied in patient-derived GBM stem cells (GSCs) and murine neural stem cells (NSCs) with and without EGFR alterations, in vitro and in vivo. EGFR alterations were analyzed by western blot and fluorescence in situ hybridization in these cells, and compared with Src activity and survival in GBM samples from The Cancer Genome Atlas. RESULTS The effect of TAT-Cx43266-283 correlated with EGFR alterations in a set of patient-derived GSCs and was stronger than that exerted by TMZ and erlotinib. In fact, TAT-Cx43266-283 only affected NSCs with EGFR alterations, but not healthy NSCs. EGFR alterations correlated with Src activity and poor survival in GBM patients. Finally, tumors generated from NSCs with EGFR alterations showed a decrease in growth, invasiveness, and vascularization after treatment with TAT-Cx43266-283, which enhanced the survival of immunocompetent mice. CONCLUSIONS Clinically relevant EGFR alterations are predictors of TAT-Cx43266-283 response and part of its mechanism of action, even in TMZ- and erlotinib-resistant GSCs. TAT-Cx43266-283 targets NSCs with GBM-driver mutations, including EGFR alterations, in an immunocompetent GBM model in vivo, suggesting a promising effect on GBM recurrence. Together, this study represents an important step toward the clinical application of TAT-Cx43266-283.
Collapse
Affiliation(s)
- Andrea Álvarez-Vázquez
- Department of Biochemistry and Molecular Biology, Neuroscience Institute of Castilla y León (INCyL), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | | | - Pilar Cerveró-García
- Department of Biochemistry and Molecular Biology, Neuroscience Institute of Castilla y León (INCyL), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Raquel Flores-Hernández
- Department of Biochemistry and Molecular Biology, Neuroscience Institute of Castilla y León (INCyL), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Department of Biochemistry and Molecular Biology, Neuroscience Institute of Castilla y León (INCyL), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Berta Segura-Collar
- Instituto de investigaciones Biomédicas I+12 (Imas12), Hospital 12 de Octubre, Madrid, Spain
| | - Christopher G Hubert
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gillian Morrison
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Steven M Pollard
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Justin D Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Pilar Sánchez-Gómez
- Neuro-Oncology Unit, Instituto de Salud Carlos III (ISCIII-UFIEC), Madrid, Spain
| | - Arantxa Tabernero
- Department of Biochemistry and Molecular Biology, Neuroscience Institute of Castilla y León (INCyL), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| |
Collapse
|
3
|
Su Y, Verkhratsky A, Yi C. Targeting connexins: possible game changer in managing neuropathic pain? Trends Mol Med 2024; 30:642-659. [PMID: 38594094 DOI: 10.1016/j.molmed.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
Neuropathic pain is a chronic debilitating condition caused by nerve injury or a variety of diseases. At the core of neuropathic pain lies the aberrant neuronal excitability in the peripheral and/or central nervous system (PNS and CNS). Enhanced connexin expression and abnormal activation of connexin-assembled gap junctional channels are prominent in neuropathic pain along with reactive gliosis, contributing to neuronal hypersensitivity and hyperexcitability. In this review, we delve into the current understanding of how connexin expression and function contribute to the pathogenesis and pathophysiology of neuropathic pain and argue for connexins as potential therapeutic targets for neuropathic pain management.
Collapse
Affiliation(s)
- Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, Bilbao, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China; Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen, China.
| |
Collapse
|
4
|
Raji L, Tetteh A, Amin ARMR. Role of c-Src in Carcinogenesis and Drug Resistance. Cancers (Basel) 2023; 16:32. [PMID: 38201459 PMCID: PMC10778207 DOI: 10.3390/cancers16010032] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
The aberrant transformation of normal cells into cancer cells, known as carcinogenesis, is a complex process involving numerous genetic and molecular alterations in response to innate and environmental stimuli. The Src family kinases (SFK) are key components of signaling pathways implicated in carcinogenesis, with c-Src and its oncogenic counterpart v-Src often playing a significant role. The discovery of c-Src represents a compelling narrative highlighting groundbreaking discoveries and valuable insights into the molecular mechanisms underlying carcinogenesis. Upon oncogenic activation, c-Src activates multiple downstream signaling pathways, including the PI3K-AKT pathway, the Ras-MAPK pathway, the JAK-STAT3 pathway, and the FAK/Paxillin pathway, which are important for cell proliferation, survival, migration, invasion, metastasis, and drug resistance. In this review, we delve into the discovery of c-Src and v-Src, the structure of c-Src, and the molecular mechanisms that activate c-Src. We also focus on the various signaling pathways that c-Src employs to promote oncogenesis and resistance to chemotherapy drugs as well as molecularly targeted agents.
Collapse
Affiliation(s)
| | | | - A. R. M. Ruhul Amin
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV 25755, USA; (L.R.); (A.T.)
| |
Collapse
|
5
|
León-Fuentes IM, Salgado-Gil MG, Novoa MS, Retamal MA. Connexins in Cancer, the Possible Role of Connexin46 as a Cancer Stem Cell-Determining Protein. Biomolecules 2023; 13:1460. [PMID: 37892142 PMCID: PMC10604234 DOI: 10.3390/biom13101460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/15/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is a widespread and incurable disease caused by genetic mutations, leading to uncontrolled cell proliferation and metastasis. Connexins (Cx) are transmembrane proteins that facilitate intercellular communication via hemichannels and gap junction channels. Among them, Cx46 is found mostly in the eye lens. However, in pathological conditions, Cx46 has been observed in various types of cancers, such as glioblastoma, melanoma, and breast cancer. It has been demonstrated that elevated Cx46 levels in breast cancer contribute to cellular resistance to hypoxia, and it is an enhancer of cancer aggressiveness supporting a pro-tumoral role. Accordingly, Cx46 is associated with an increase in cancer stem cell phenotype. These cells display radio- and chemoresistance, high proliferative abilities, self-renewal, and differentiation capacities. This review aims to consolidate the knowledge of the relationship between Cx46, its role in forming hemichannels and gap junctions, and its connection with cancer and cancer stem cells.
Collapse
Affiliation(s)
| | | | | | - Mauricio A. Retamal
- Programa de Comunicación Celular en Cáncer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, República de Honduras 12740, Las Condes, Santiago 7610496, Chile; (I.M.L.-F.); (M.G.S.-G.); (M.S.N.)
| |
Collapse
|
6
|
Zhou M, Zheng M, Zhou X, Tian S, Yang X, Ning Y, Li Y, Zhang S. The roles of connexins and gap junctions in the progression of cancer. Cell Commun Signal 2023; 21:8. [PMID: 36639804 PMCID: PMC9837928 DOI: 10.1186/s12964-022-01009-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/03/2022] [Indexed: 01/15/2023] Open
Abstract
Gap junctions (GJs), which are composed of connexins (Cxs), provide channels for direct information exchange between cells. Cx expression has a strong spatial specificity; however, its influence on cell behavior and information exchange between cells cannot be ignored. A variety of factors in organisms can modulate Cxs and subsequently trigger a series of responses that have important effects on cellular behavior. The expression and function of Cxs and the number and function of GJs are in dynamic change. Cxs have been characterized as tumor suppressors in the past, but recent studies have highlighted the critical roles of Cxs and GJs in cancer pathogenesis. The complex mechanism underlying Cx and GJ involvement in cancer development is a major obstacle to the evolution of therapy targeting Cxs. In this paper, we review the post-translational modifications of Cxs, the interactions of Cxs with several chaperone proteins, and the effects of Cxs and GJs on cancer. Video Abstract.
Collapse
Affiliation(s)
- Mingming Zhou
- grid.265021.20000 0000 9792 1228Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121 People’s Republic of China
| | - Xinyue Zhou
- grid.265021.20000 0000 9792 1228Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Shifeng Tian
- grid.265021.20000 0000 9792 1228Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Xiaohui Yang
- grid.216938.70000 0000 9878 7032Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yidi Ning
- grid.216938.70000 0000 9878 7032Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yuwei Li
- grid.417031.00000 0004 1799 2675Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121 People’s Republic of China
| |
Collapse
|
7
|
Sedovy MW, Leng X, Leaf MR, Iqbal F, Payne LB, Chappell JC, Johnstone SR. Connexin 43 across the Vasculature: Gap Junctions and Beyond. J Vasc Res 2022; 60:101-113. [PMID: 36513042 PMCID: PMC11073551 DOI: 10.1159/000527469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/26/2022] [Indexed: 12/15/2022] Open
Abstract
Connexin 43 (Cx43) is essential to the function of the vasculature. Cx43 proteins form gap junctions that allow for the exchange of ions and molecules between vascular cells to facilitate cell-to-cell signaling and coordinate vasomotor activity. Cx43 also has intracellular signaling functions that influence vascular cell proliferation and migration. Cx43 is expressed in all vascular cell types, although its expression and function vary by vessel size and location. This includes expression in vascular smooth muscle cells (vSMC), endothelial cells (EC), and pericytes. Cx43 is thought to coordinate homocellular signaling within EC and vSMC. Cx43 gap junctions also function as conduits between different cell types (heterocellular signaling), between EC and vSMC at the myoendothelial junction, and between pericyte and EC in capillaries. Alterations in Cx43 expression, localization, and post-translational modification have been identified in vascular disease states, including atherosclerosis, hypertension, and diabetes. In this review, we discuss the current understanding of Cx43 localization and function in healthy and diseased blood vessels across all vascular beds.
Collapse
Affiliation(s)
- Meghan W. Sedovy
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Translational Biology, Medicine, And Health Graduate Program, Virginia Tech, Blacksburg, VA, USA
| | - Xinyan Leng
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - Melissa R. Leaf
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Farwah Iqbal
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Laura Beth Payne
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - John C. Chappell
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - Scott R. Johnstone
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
8
|
Pelaz SG, Tabernero A. Src: coordinating metabolism in cancer. Oncogene 2022; 41:4917-4928. [PMID: 36217026 PMCID: PMC9630107 DOI: 10.1038/s41388-022-02487-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/08/2022]
Abstract
Metabolism must be tightly regulated to fulfil the dynamic requirements of cancer cells during proliferation, migration, stemness and differentiation. Src is a node of several signals involved in many of these biological processes, and it is also an important regulator of cell metabolism. Glucose uptake, glycolysis, the pentose-phosphate pathway and oxidative phosphorylation are among the metabolic pathways that can be regulated by Src. Therefore, this oncoprotein is in an excellent position to coordinate and finely tune cell metabolism to fuel the different cancer cell activities. Here, we provide an up-to-date summary of recent progress made in determining the role of Src in glucose metabolism as well as the link of this role with cancer cell metabolic plasticity and tumour progression. We also discuss the opportunities and challenges facing this field.
Collapse
Affiliation(s)
- Sara G Pelaz
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain
| | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Calle Pintor Fernando Gallego 1, Salamanca, 37007, Spain.
| |
Collapse
|
9
|
King DR, Sedovy MW, Leng X, Xue J, Lamouille S, Koval M, Isakson BE, Johnstone SR. Mechanisms of Connexin Regulating Peptides. Int J Mol Sci 2021; 22:ijms221910186. [PMID: 34638526 PMCID: PMC8507914 DOI: 10.3390/ijms221910186] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/22/2022] Open
Abstract
Gap junctions (GJ) and connexins play integral roles in cellular physiology and have been found to be involved in multiple pathophysiological states from cancer to cardiovascular disease. Studies over the last 60 years have demonstrated the utility of altering GJ signaling pathways in experimental models, which has led to them being attractive targets for therapeutic intervention. A number of different mechanisms have been proposed to regulate GJ signaling, including channel blocking, enhancing channel open state, and disrupting protein-protein interactions. The primary mechanism for this has been through the design of numerous peptides as therapeutics, that are either currently in early development or are in various stages of clinical trials. Despite over 25 years of research into connexin targeting peptides, the overall mechanisms of action are still poorly understood. In this overview, we discuss published connexin targeting peptides, their reported mechanisms of action, and the potential for these molecules in the treatment of disease.
Collapse
Affiliation(s)
- D. Ryan King
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
| | - Meghan W. Sedovy
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, VA 24061, USA
| | - Xinyan Leng
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
| | - Jianxiang Xue
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (J.X.); (B.E.I.)
| | - Samy Lamouille
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA 24016, USA
| | - Michael Koval
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (J.X.); (B.E.I.)
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Scott R. Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060, USA
- Correspondence:
| |
Collapse
|
10
|
Orellana VP, Tittarelli A, Retamal MA. Connexins in melanoma: Potential role of Cx46 in its aggressiveness. Pigment Cell Melanoma Res 2021; 34:853-868. [PMID: 33140904 DOI: 10.1111/pcmr.12945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022]
Abstract
Melanoma is the most aggressive skin cancer, and in metastatic advanced states, it is completely refractory to chemotherapy. Therefore, it is relevant to understand the molecular bases that rule their aggressiveness. Connexins (Cxs) are proteins that under normal physiological conditions participate in intercellular communication, via the exchange of signaling molecules between the cytoplasm and extracellular milieu and the exchange of ions/second messengers between the cytoplasm of contacting cells. These proteins have shown important roles in cancer progression, chemo- and radiotherapy resistance, and metastasis. Accordingly, Cx26 and Cx43 seem to play important roles in melanoma progression and metastasis. On the other hand, Cx46 is typically expressed in the eye lens, where it seems to be associated with oxidative stress protection in fiber lens cells. However, in the last decade, Cx46 expression has been associated with breast and brain cancers, due to its role in potentiation of both extracellular vesicle release and cancer stem cell-like properties. In this review, we analyzed a potential role of Cx46 as a new biomarker and therapeutic target in melanoma.
Collapse
Affiliation(s)
- Viviana P Orellana
- Universidad del Desarrollo. Centro de Fisiología Celular e Integrativa, Clinica Alemana Facultad de Medicina, Santiago, Chile
- Universidad del Desarrollo. Programa de Comunicación Celular en Cáncer, Clínica Alemana Facultad de Medicina, Santiago, Chile
| | - Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación (PIDi), Universidad Tecnológica Metropolitana (UTEM), Santiago, Chile
| | - Mauricio A Retamal
- Universidad del Desarrollo. Centro de Fisiología Celular e Integrativa, Clinica Alemana Facultad de Medicina, Santiago, Chile
- Universidad del Desarrollo. Programa de Comunicación Celular en Cáncer, Clínica Alemana Facultad de Medicina, Santiago, Chile
| |
Collapse
|
11
|
Pelaz SG, Ollauri-Ibáñez C, Lillo C, Tabernero A. Impairment of Autophagic Flux Participates in the Antitumor Effects of TAT-Cx43 266-283 in Glioblastoma Stem Cells. Cancers (Basel) 2021; 13:cancers13174262. [PMID: 34503072 PMCID: PMC8428230 DOI: 10.3390/cancers13174262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/13/2021] [Accepted: 08/21/2021] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Autophagy is a process in which the cell recycles components that are not needed at that moment and uses the resulting elements to satisfy more urgent needs. Depending on the specific context, this can be beneficial or detrimental for tumor development. We found that in glioblastoma, the most lethal brain tumor, autophagy is upregulated and contributes to glioblastoma stem cell survival under starvation. Importantly, the antitumor peptide TAT-Cx43266-283 blocks autophagy flux, contributing to the death of glioblastoma stem cells. This peptide induces glioblastoma stem cell death in nutrient-deprived and complete environments, while the effect of other unsuccessful drugs for glioblastoma depends on nutrient context, supporting the potential of TAT-Cx43266-283 as a treatment to improve the lives of glioblastoma patients. Abstract Autophagy is a physiological process by which various damaged or non-essential cytosolic components are recycled, contributing to cell survival under stress conditions. In cancer, autophagy can have antitumor or protumor effects depending on the developmental stage. Here, we use Western blotting, immunochemistry, and transmission electron microscopy to demonstrate that the antitumor peptide TAT-Cx43266-283, a c-Src inhibitor, blocks autophagic flux in glioblastoma stem cells (GSCs) under basal and nutrient-deprived conditions. Upon nutrient deprivation, GSCs acquired a dormant-like phenotype that was disrupted by inhibition of autophagy with TAT-Cx43266-283 or chloroquine (a classic autophagy inhibitor), leading to GSC death. Remarkably, dasatinib, a clinically available c-Src inhibitor, could not replicate TAT-Cx43266-283 effect on dormant GSCs, revealing for the first time the possible involvement of pathways other than c-Src in TAT-Cx43266-283 effect. TAT-Cx43266-283 exerts an antitumor effect both in nutrient-complete and nutrient-deprived environments, which constitutes an advantage over chloroquine and dasatinib, whose effects depend on nutrient environment. Finally, our analysis of the levels of autophagy-related proteins in healthy and glioma donors suggests that autophagy is upregulated in glioblastoma, further supporting the interest in inhibiting this process in the most aggressive brain tumor and the potential use of TAT-Cx43266-283 as a therapy for this type of cancer.
Collapse
Affiliation(s)
- Sara G. Pelaz
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, 37007 Salamanca, Spain; (S.G.P.); (C.O.-I.); (C.L.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª Planta, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, 37007 Salamanca, Spain; (S.G.P.); (C.O.-I.); (C.L.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª Planta, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
| | - Concepción Lillo
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, 37007 Salamanca, Spain; (S.G.P.); (C.O.-I.); (C.L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª Planta, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, 37007 Salamanca, Spain; (S.G.P.); (C.O.-I.); (C.L.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª Planta, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
- Correspondence:
| |
Collapse
|
12
|
Wu DP, Zhou Y, Hou LX, Zhu XX, Yi W, Yang SM, Lin TY, Huang JL, Zhang B, Yin XX. Cx43 deficiency confers EMT-mediated tamoxifen resistance to breast cancer via c-Src/PI3K/Akt pathway. Int J Biol Sci 2021; 17:2380-2398. [PMID: 34326682 PMCID: PMC8315014 DOI: 10.7150/ijbs.55453] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/30/2021] [Indexed: 12/25/2022] Open
Abstract
Tamoxifen (TAM) resistance has indicated a significant challenge during endocrine therapy for hormone-sensitive breast cancer. Thus, it is significant to elucidate the molecular events endowing TAM resistance to endocrine therapy. In this study, we found that epithelial-mesenchymal transition (EMT) was an important event to confer TAM resistance, and attenuating EMT by elevating connexin (Cx) 43 expression could reverse TAM resistance. Specifically, Cx43 overexpression improved TAM sensitivity, while Cx43 depletion facilitated TAM insensitivity by modulating EMT in T47D TAM-resistant and -sensitive cells, and transplanted xenografts. Importantly, we found a novel reciprocal regulation between Cx43 and c-Src/PI3K/Akt pathway contributing to EMT and TAM resistance in breast cancer. Moreover, we identified that Cx43 deficiency was significantly correlated with poor relapse-free survival in patients undergoing TAM treatment. Therefore, Cx43 represents a prognostic marker and an attractive target for breast cancer treatments. Therapeutic strategies designed to increase or maintain Cx43 function may be beneficial to overcome TAM resistance.
Collapse
Affiliation(s)
- Deng-Pan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, P.R. China
- Department of Pharmacology, Pharmacy School of Xuzhou Medical University, 221004, Xuzhou City, Jiangsu Province, P.R. China
| | - Yan Zhou
- Clinical Pharmacy, Jingjiang People's Hospital, 214500, Jingjiang City, Jiangsu Province, P.R. China
| | - Li-Xiang Hou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, P.R. China
| | - Xiao-Xiao Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, P.R. China
| | - Wen Yi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, P.R. China
| | - Si-Man Yang
- Scientific research center of traditional Chinese medicine, Guangxi University of Chinese Medicine, Nanning City, Guangxi Zhuang Autonomous Region, P.R. China
| | - Tian-Yu Lin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, P.R. China
| | - Jin-Lan Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, P.R. China
- Department of Pharmacology, Pharmacy School of Xuzhou Medical University, 221004, Xuzhou City, Jiangsu Province, P.R. China
| | - Bei Zhang
- Department of gynaecology and obstetrics, Xuzhou Central Hospital, 221009, Xuzhou City, Jiangsu Province, P.R. China
| | - Xiao-Xing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, P.R. China
| |
Collapse
|
13
|
Jaraíz-Rodríguez M, Talaverón R, García-Vicente L, Pelaz SG, Domínguez-Prieto M, Álvarez-Vázquez A, Flores-Hernández R, Sin WC, Bechberger J, Medina JM, Naus CC, Tabernero A. Connexin43 peptide, TAT-Cx43266-283, selectively targets glioma cells, impairs malignant growth, and enhances survival in mouse models in vivo. Neuro Oncol 2021; 22:493-504. [PMID: 31883012 PMCID: PMC7158688 DOI: 10.1093/neuonc/noz243] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Malignant gliomas are the most frequent primary brain tumors and remain among the most incurable cancers. Although the role of the gap junction protein, connexin43 (Cx43), has been deeply investigated in malignant gliomas, no compounds have been reported with the ability to recapitulate the tumor suppressor properties of this protein in in vivo glioma models. Methods TAT-Cx43266–283 a cell-penetrating peptide which mimics the effect of Cx43 on c-Src inhibition, was studied in orthotopic immunocompetent and immunosuppressed models of glioma. The effects of this peptide in brain cells were also analyzed. Results While glioma stem cell malignant features were strongly affected by TAT-Cx43266–283, these properties were not significantly modified in neurons and astrocytes. Intraperitoneally administered TAT-Cx43266–283 decreased the invasion of intracranial tumors generated by GL261 mouse glioma cells in immunocompetent mice. When human glioma stem cells were intracranially injected with TAT-Cx43266–283 into immunodeficient mice, there was reduced expression of the stemness markers nestin and Sox2 in human glioma cells at 7 days post-implantation. Consistent with the role of Sox2 as a transcription factor required for tumorigenicity, TAT-Cx43266–283 reduced the number and stemness of human glioma cells at 30 days post-implantation. Furthermore, TAT-Cx43266–283 enhanced the survival of immunocompetent mice bearing gliomas derived from murine glioma stem cells. Conclusion TAT-Cx43266–283 reduces the growth, invasion, and progression of malignant gliomas and enhances the survival of glioma-bearing mice without exerting toxicity in endogenous brain cells, which suggests that this peptide could be considered as a new clinical therapy for high-grade gliomas.
Collapse
Affiliation(s)
- Myriam Jaraíz-Rodríguez
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Rocío Talaverón
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Laura García-Vicente
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Sara G Pelaz
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Marta Domínguez-Prieto
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Andrea Álvarez-Vázquez
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Raquel Flores-Hernández
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Wun Chey Sin
- Department of Cellular and Physiological Sciences, The Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - John Bechberger
- Department of Cellular and Physiological Sciences, The Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - José M Medina
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Christian C Naus
- Department of Cellular and Physiological Sciences, The Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arantxa Tabernero
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| |
Collapse
|
14
|
Zhang S, Gong Y, Wang H, Li Z, Huang Y, Fu X, Xiang P, Fan T. AS602801 sensitizes glioma cells to temozolomide and vincristine by blocking gap junction communication between glioma cells and astrocytes. J Cell Mol Med 2021; 25:4062-4072. [PMID: 33609076 PMCID: PMC8051707 DOI: 10.1111/jcmm.16375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/28/2022] Open
Abstract
Previous studies showed that the chemotherapeutic effect of temozolomide (TMZ) and vincristine (VCR) against glioma might be blunted by the co‐culture with astrocytes, and connexin‐43 (CX43) was thought to play a vital role in the communication between glioma cells and astrocytes. In this study, we aimed to investigate the combined chemotherapeutic effect of AS602801 and TMZ/ VCR in glioma cells both. Dye transfer assay was used to evaluate the gap junction activity between U251 cells and astrocytes. Western blot and immunohistochemistry were carried out to analyse the expression of p‐JNK, CX43 and CASP‐3 proteins treated under different conditions. AS602801 significantly suppressed the gap junction activity between U251 cells and astrocytes. The expression of p‐JNK and CX43 was remarkably inhibited by AS602801. TMZ/VCR‐induced apoptosis of glioma cells was effectively enhanced by AS602801 treatment. Accordingly, the inhibitory role of TMZ/VCR in the expression of p‐JNK, CX43 and CASP‐3 in glioma cells was notably restored by AS602801. Furthermore, in a glioma cell xenograft, AS602801 showed an apparent capability to enhance TMZ/VCR‐induced tumour cell apoptosis through altering the expression of p‐JNK, CX43 and CASP‐3. The findings of this study demonstrated that the co‐culture of glioma cells with astrocytes blunted the tumour killing effect of TMZ and VCR. AS602801 down‐regulated CX43 expression by inhibiting JNK. And AS602801 also sensitized glioma cells to TMZ/VCR by blocking the gap junction communication between glioma cells and astrocytes via down‐regulating CX43, indicating its potential role as a novel adjuvant chemotherapeutic agent in the treatment of glioma.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Neurosurgery, ChangSha Central Hospital, Changsha, China
| | - Yong Gong
- Department of Neurosurgery, ChangSha Central Hospital, Changsha, China
| | - Hongxin Wang
- Department of Neurosurgery, ChangSha Central Hospital, Changsha, China
| | - Zhongfan Li
- Department of Neurosurgery, ChangSha Central Hospital, Changsha, China
| | - Yunfeng Huang
- Department of Neurosurgery, ChangSha Central Hospital, Changsha, China
| | - Xing Fu
- Department of Neurosurgery, ChangSha Central Hospital, Changsha, China
| | - Peng Xiang
- Department of Neurosurgery, ChangSha Central Hospital, Changsha, China
| | - TianYu Fan
- Department of Neurosurgery, ChangSha Central Hospital, Changsha, China
| |
Collapse
|
15
|
Sheng Z. Connexin 43 peptidic medicine for glioblastoma stem cells. EBioMedicine 2021; 64:103205. [PMID: 33493796 PMCID: PMC7823208 DOI: 10.1016/j.ebiom.2020.103205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 01/03/2023] Open
Affiliation(s)
- Zhi Sheng
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, United States; Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, United States; Faculty of Health Science, Virginia Tech, Blacksburg, VA 24061, United States.
| |
Collapse
|
16
|
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
|
17
|
Pelaz SG, Jaraíz-Rodríguez M, Álvarez-Vázquez A, Talaverón R, García-Vicente L, Flores-Hernández R, Gómez de Cedrón M, Tabernero M, Ramírez de Molina A, Lillo C, Medina JM, Tabernero A. Targeting metabolic plasticity in glioma stem cells in vitro and in vivo through specific inhibition of c-Src by TAT-Cx43 266-283. EBioMedicine 2020; 62:103134. [PMID: 33254027 PMCID: PMC7708820 DOI: 10.1016/j.ebiom.2020.103134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 10/24/2020] [Accepted: 11/02/2020] [Indexed: 12/22/2022] Open
Abstract
Background Glioblastoma is the most aggressive primary brain tumour and has a very poor prognosis. Inhibition of c-Src activity in glioblastoma stem cells (GSCs, responsible for glioblastoma lethality) and primary glioblastoma cells by the peptide TAT-Cx43266–283 reduces tumorigenicity, and boosts survival in preclinical models. Because c-Src can modulate cell metabolism and several reports revealed poor clinical efficacy of various antitumoral drugs due to metabolic rewiring in cancer cells, here we explored the inhibition of advantageous GSC metabolic plasticity by the c-Src inhibitor TAT-Cx43266-283. Methods Metabolic impairment induced by the c-Src inhibitor TAT-Cx43266-283 in vitro was assessed by fluorometry, western blotting, immunofluorescence, qPCR, enzyme activity assays, electron microscopy, Seahorse analysis, time-lapse imaging, siRNA, and MTT assays. Protein expression in tumours from a xenograft orthotopic glioblastoma mouse model was evaluated by immunofluorescence. Findings TAT-Cx43266–283 decreased glucose uptake in human GSCs and reduced oxidative phosphorylation without a compensatory increase in glycolysis, with no effect on brain cell metabolism, including rat neurons, human and rat astrocytes, and human neural stem cells. TAT-Cx43266-283 impaired metabolic plasticity, reducing GSC growth and survival under different nutrient environments. Finally, GSCs intracranially implanted with TAT-Cx43266–283 showed decreased levels of important metabolic targets for cancer therapy, such as hexokinase-2 and GLUT-3. Interpretation The reduced ability of TAT-Cx43266-283–treated GSCs to survive in metabolically challenging settings, such as those with restricted nutrient availability or the ever-changing in vivo environment, allows us to conclude that the advantageous metabolic plasticity of GSCs can be therapeutically exploited through the specific and cell-selective inhibition of c-Src by TAT-Cx43266-283. Funding Spanish Ministerio de Economía y Competitividad (FEDER BFU2015-70040-R and FEDER RTI2018-099873-B-I00), Fundación Ramón Areces. Fellowships from the Junta de Castilla y León, European Social Fund, Ministerio de Ciencia and Asociación Española Contra el Cáncer (AECC).
Collapse
Affiliation(s)
- Sara G Pelaz
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Departamento de Bioquímica y Biología Celular, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain
| | - Myriam Jaraíz-Rodríguez
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Departamento de Bioquímica y Biología Celular, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain
| | - Andrea Álvarez-Vázquez
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Departamento de Bioquímica y Biología Celular, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain
| | - Rocío Talaverón
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Departamento de Bioquímica y Biología Celular, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain
| | - Laura García-Vicente
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Departamento de Bioquímica y Biología Celular, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain
| | - Raquel Flores-Hernández
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Departamento de Bioquímica y Biología Celular, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain
| | - Marta Gómez de Cedrón
- Precision Nutrition and Cancer Program, Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Canto Blanco 8 E, Madrid 28049, Spain
| | - María Tabernero
- Precision Nutrition and Cancer Program, Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Canto Blanco 8 E, Madrid 28049, Spain
| | - Ana Ramírez de Molina
- Precision Nutrition and Cancer Program, Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Canto Blanco 8 E, Madrid 28049, Spain
| | - Concepción Lillo
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain
| | - José M Medina
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Departamento de Bioquímica y Biología Celular, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain
| | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Calle Pintor Fernando Gallego 1, Salamanca 37007, Spain; Departamento de Bioquímica y Biología Celular, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca 37007, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Virgen de la Vega, 10ª planta, Paseo de San Vicente, 58-182, Salamanca 37007, Spain.
| |
Collapse
|
18
|
Talaverón R, Matarredona ER, Herrera A, Medina JM, Tabernero A. Connexin43 Region 266-283, via Src Inhibition, Reduces Neural Progenitor Cell Proliferation Promoted by EGF and FGF-2 and Increases Astrocytic Differentiation. Int J Mol Sci 2020; 21:ijms21228852. [PMID: 33238452 PMCID: PMC7700635 DOI: 10.3390/ijms21228852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Neural progenitor cells (NPCs) are self-renewing cells that give rise to the major cells in the nervous system and are considered to be the possible cell of origin of glioblastoma. The gap junction protein connexin43 (Cx43) is expressed by NPCs, exerting channel-dependent and -independent roles. We focused on one property of Cx43—its ability to inhibit Src, a key protein in brain development and oncogenesis. Because Src inhibition is carried out by the sequence 266–283 of the intracellular C terminus in Cx43, we used a cell-penetrating peptide containing this sequence, TAT-Cx43266–283, to explore its effects on postnatal subventricular zone NPCs. Our results show that TAT-Cx43266–283 inhibited Src activity and reduced NPC proliferation and survival promoted by epidermal growth factor (EGF) and fibroblast growth factor-2 (FGF-2). In differentiation conditions, TAT-Cx43266–283 increased astrocyte differentiation at the expense of neuronal differentiation, which coincided with a reduction in Src activity and β-catenin expression. We propose that Cx43, through the region 266–283, reduces Src activity, leading to disruption of EGF and FGF-2 signaling and to down-regulation of β-catenin with effects on proliferation and differentiation. Our data indicate that the inhibition of Src might contribute to the complex role of Cx43 in NPCs and open new opportunities for further research in gliomagenesis.
Collapse
Affiliation(s)
- Rocío Talaverón
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, 37007 Salamanca, Spain; (R.T.); (J.M.M.)
| | | | - Alejandro Herrera
- Departamento de Fisiología, Universidad de Sevilla, 41012 Sevilla, Spain; (E.R.M.); (A.H.)
| | - José M. Medina
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, 37007 Salamanca, Spain; (R.T.); (J.M.M.)
| | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, 37007 Salamanca, Spain; (R.T.); (J.M.M.)
- Correspondence: ; Tel.: +34-923-29-45-00 (ext. 5311)
| |
Collapse
|
19
|
Cell-penetrating peptides in oncologic pharmacotherapy: A review. Pharmacol Res 2020; 162:105231. [PMID: 33027717 DOI: 10.1016/j.phrs.2020.105231] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/23/2020] [Accepted: 09/30/2020] [Indexed: 01/10/2023]
Abstract
Cancer is the second leading cause of death in the world and its treatment is extremely challenging, mainly due to its complexity. Cell-Penetrating Peptides (CPPs) are peptides that can transport into the cell a wide variety of biologically active conjugates (or cargoes), and are, therefore, promising in the treatment and in the diagnosis of several types of cancer. Some notable examples are TAT and Penetratin, capable of penetrating the central nervous system (CNS) and, therefore, acting in cancers of this system, such as Glioblastoma Multiforme (GBM). These above-mentioned peptides, conjugated with traditional chemotherapeutic such as Doxorubicin (DOX) and Paclitaxel (PTX), have also been shown to induce apoptosis of breast and liver cancer cells, as well as in lung cancer cells, respectively. In other cancers, such as esophageal cancer, the attachment of Magainin 2 (MG2) to Bombesin (MG2B), another CPP, led to pronounced anticancer effects. Other examples are CopA3, that selectively decreased the viability of gastric cancer cells, and the CPP p28. Furthermore, in preclinical tests, the anti-tumor efficacy of this peptide was evaluated on human breast cancer, prostate cancer, ovarian cancer, and melanoma cells in vitro, leading to high expression of p53 and promoting cell cycle arrest. Despite the numerous in vitro and in vivo studies with promising results, and the increasing number of clinical trials using CPPs, few treatments reach the expected clinical efficacy. Usually, their clinical application is limited by its poor aqueous solubility, immunogenicity issues and dose-limiting toxicity. This review describes the most recent advances and innovations in the use of CPPs in several types of cancer, highlighting their crucial importance for various purposes, from therapeutic to diagnosis. Further clinical trials with these peptides are warranted to examine its effects on various types of cancer.
Collapse
|
20
|
Giaume C, Naus CC, Sáez JC, Leybaert L. Glial Connexins and Pannexins in the Healthy and Diseased Brain. Physiol Rev 2020; 101:93-145. [PMID: 32326824 DOI: 10.1152/physrev.00043.2018] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Over the past several decades a large amount of data have established that glial cells, the main cell population in the brain, dynamically interact with neurons and thus impact their activity and survival. One typical feature of glia is their marked expression of several connexins, the membrane proteins forming intercellular gap junction channels and hemichannels. Pannexins, which have a tetraspan membrane topology as connexins, are also detected in glial cells. Here, we review the evidence that connexin and pannexin channels are actively involved in dynamic and metabolic neuroglial interactions in physiological as well as in pathological situations. These features of neuroglial interactions open the way to identify novel non-neuronal aspects that allow for a better understanding of behavior and information processing performed by neurons. This will also complement the "neurocentric" view by facilitating the development of glia-targeted therapeutic strategies in brain disease.
Collapse
Affiliation(s)
- Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christian C Naus
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Juan C Sáez
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Luc Leybaert
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
21
|
Sánchez OF, Rodríguez AV, Velasco-España JM, Murillo LC, Sutachan JJ, Albarracin SL. Role of Connexins 30, 36, and 43 in Brain Tumors, Neurodegenerative Diseases, and Neuroprotection. Cells 2020; 9:E846. [PMID: 32244528 PMCID: PMC7226843 DOI: 10.3390/cells9040846] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/15/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023] Open
Abstract
Gap junction (GJ) channels and their connexins (Cxs) are complex proteins that have essential functions in cell communication processes in the central nervous system (CNS). Neurons, astrocytes, oligodendrocytes, and microglial cells express an extraordinary repertory of Cxs that are important for cell to cell communication and diffusion of metabolites, ions, neurotransmitters, and gliotransmitters. GJs and Cxs not only contribute to the normal function of the CNS but also the pathological progress of several diseases, such as cancer and neurodegenerative diseases. Besides, they have important roles in mediating neuroprotection by internal or external molecules. However, regulation of Cx expression by epigenetic mechanisms has not been fully elucidated. In this review, we provide an overview of the known mechanisms that regulate the expression of the most abundant Cxs in the central nervous system, Cx30, Cx36, and Cx43, and their role in brain cancer, CNS disorders, and neuroprotection. Initially, we focus on describing the Cx gene structure and how this is regulated by epigenetic mechanisms. Then, the posttranslational modifications that mediate the activity and stability of Cxs are reviewed. Finally, the role of GJs and Cxs in glioblastoma, Alzheimer's, Parkinson's, and Huntington's diseases, and neuroprotection are analyzed with the aim of shedding light in the possibility of using Cx regulators as potential therapeutic molecules.
Collapse
Affiliation(s)
- Oscar F. Sánchez
- Department of Nutrition and Biochemistry, Pontificia Universidad Javeriana, 110911 Bogota, Colombia; (A.V.R.); (J.M.V.-E.); (L.C.M.); (J.-J.S.)
| | | | | | | | | | - Sonia-Luz Albarracin
- Department of Nutrition and Biochemistry, Pontificia Universidad Javeriana, 110911 Bogota, Colombia; (A.V.R.); (J.M.V.-E.); (L.C.M.); (J.-J.S.)
| |
Collapse
|
22
|
Kirichenko EY, Salah M M S, Goncharova ZA, Nikitin AG, Filippova SY, Todorov SS, Akimenko MA, Logvinov AK. Ultrastructural evidence for presenсe of gap junctions in rare case of pleomorphic xanthoastrocytoma. Ultrastruct Pathol 2020; 44:227-236. [PMID: 32148147 DOI: 10.1080/01913123.2020.1737609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The phenomenon of unstable expression of gap junction's proteins connexins remains a "visiting card" of astrocytic tumors with various degrees of malignancy. At the same time, it stays unclear what is detected by the positive expression of connexins in astrocytic tumors: gap junctions, hemi-channels, or connexin proteins in cytosol. In the present work, for the first time, we demonstrate an ultrastructural evidence of gap junctions in pleomorphic xanthoastrocytoma, a rare primary brain tumor, the intercellular characteristics of which are poorly studied and remain very discursive and controversial. The primary tumor mass was resected during craniotomy from a 57-old patient diagnosed with pleomorphic xanthoastrocytoma Grade II based on the histopathological analysis. The immunohistochemical study was conducted with primary antibodies: Neurofilament, Myelin basic protein, Glial fibrillary acidic protein, and Synaptophysin. For electron microscopic examination fragments of tumor tissue were fixed in a glutaraldehyde, postfixed in a 1% OsO4, dehydrated and embedded into resin. After the detailed clinical, histological, and immunohistochemical study we revealed some ultrastructural characteristics of the tumor, as well as the first evidence of direct intercellular connection between the tumor cells via gap junctions. Regularly arranged gap junctions connected the somas of xanthastrocytes with dark cytoplasm containing lipid drops. Besides the localization between the cell bodies, from one to several gap junctions were found between the branches of xanthoastrocytoma in tumor intercellular space in close proximity to tumor cell. Our results may indicate gap junctions as a possible structure for intercellular communication between pleomorphic xanthoastrocytoma cells.
Collapse
Affiliation(s)
| | | | | | - Aleksei G Nikitin
- Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies FMBA of Russia, Moscow, Russian Federation
| | | | | | | | | |
Collapse
|
23
|
Wang J, Yang ZY, Guo YF, Kuang JY, Bian XW, Yu SC. Targeting different domains of gap junction protein to control malignant glioma. Neuro Oncol 2019; 20:885-896. [PMID: 29106645 DOI: 10.1093/neuonc/nox207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A rational treatment strategy for glioma, the most common primary central nervous system tumor, should focus on early invasive growth and resistance to current therapeutics. Connexin 43 (Cx43), a gap junction protein, plays important roles not only in the development of the central nervous system and but also in the progression of glioma. The different structural domains of Cx43, including extracellular loops, transmembrane domains, and an intracellular carboxyl terminal, have distinct functions in the invasion and proliferation of gliomas. Targeting these domains of Cx43, which is expressed in distinct patterns in the heterogeneous glioma cell population, can inhibit tumor cell invasion and new tumor formation. Thus, this review summarizes the structural characteristics of Cx43, the effects of regulating different Cx43 domains on the biological characteristics of glioma cells, intervention strategies targeting different domains of Cx43, and future research directions.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Ze-Yu Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Yu-Feng Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Jing-Ya Kuang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| |
Collapse
|
24
|
Aasen T, Leithe E, Graham SV, Kameritsch P, Mayán MD, Mesnil M, Pogoda K, Tabernero A. Connexins in cancer: bridging the gap to the clinic. Oncogene 2019; 38:4429-4451. [PMID: 30814684 PMCID: PMC6555763 DOI: 10.1038/s41388-019-0741-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/26/2019] [Accepted: 01/26/2019] [Indexed: 02/08/2023]
Abstract
Gap junctions comprise arrays of intercellular channels formed by connexin proteins and provide for the direct communication between adjacent cells. This type of intercellular communication permits the coordination of cellular activities and plays key roles in the control of cell growth and differentiation and in the maintenance of tissue homoeostasis. After more than 50 years, deciphering the links among connexins, gap junctions and cancer, researchers are now beginning to translate this knowledge to the clinic. The emergence of new strategies for connexin targeting, combined with an improved understanding of the molecular bases underlying the dysregulation of connexins during cancer development, offers novel opportunities for clinical applications. However, different connexin isoforms have diverse channel-dependent and -independent functions that are tissue and stage specific. This can elicit both pro- and anti-tumorigenic effects that engender significant challenges in the path towards personalised medicine. Here, we review the current understanding of the role of connexins and gap junctions in cancer, with particular focus on the recent progress made in determining their prognostic and therapeutic potential.
Collapse
Affiliation(s)
- Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, Barcelona, Spain.
| | - Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital and K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Sheila V Graham
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Petra Kameritsch
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), University of A Coruña, A Coruña, Spain
| | - Marc Mesnil
- STIM Laboratory, Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers, France
| | - Kristin Pogoda
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany
| | - Arantxa Tabernero
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca, Spain.
| |
Collapse
|
25
|
Freitas-Andrade M, Wang N, Bechberger JF, De Bock M, Lampe PD, Leybaert L, Naus CC. Targeting MAPK phosphorylation of Connexin43 provides neuroprotection in stroke. J Exp Med 2019; 216:916-935. [PMID: 30872361 PMCID: PMC6446879 DOI: 10.1084/jem.20171452] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 03/31/2018] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
This study demonstrates that astrocytic connexin43 gap junction hemichannels are largely controlled by four C-terminal tail–located serine residues and provides mechanistic insight on how phosphorylation of these residues affects recovery from stroke. Connexin43 (Cx43) function is influenced by kinases that phosphorylate specific serine sites located near its C-terminus. Stroke is a powerful inducer of kinase activity, but its effect on Cx43 is unknown. We investigated the impact of wild-type (WT) and knock-in Cx43 with serine to alanine mutations at the protein kinase C (PKC) site Cx43S368A, the casein kinase 1 (CK1) sites Cx43S325A/328Y/330A, and the mitogen-activated protein kinase (MAPK) sites Cx43S255/262/279/282A (MK4) on a permanent middle cerebral artery occlusion (pMCAO) stroke model. We demonstrate that MK4 transgenic animals exhibit a significant decrease in infarct volume that was associated with improvement in behavioral performance. An increase in astrocyte reactivity with a concomitant decrease in microglial reactivity was observed in MK4 mice. In contrast to WT, MK4 astrocytes displayed reduced Cx43 hemichannel activity. Pharmacological blockade of Cx43 hemichannels with TAT-Gap19 also significantly decreased infarct volume in WT animals. This study provides novel molecular insights and charts new avenues for therapeutic intervention associated with Cx43 function.
Collapse
Affiliation(s)
- Moises Freitas-Andrade
- Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nan Wang
- Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - John F Bechberger
- Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marijke De Bock
- Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Paul D Lampe
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Luc Leybaert
- Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Christian C Naus
- Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Beckmann A, Hainz N, Tschernig T, Meier C. Facets of Communication: Gap Junction Ultrastructure and Function in Cancer Stem Cells and Tumor Cells. Cancers (Basel) 2019; 11:cancers11030288. [PMID: 30823688 PMCID: PMC6468480 DOI: 10.3390/cancers11030288] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/28/2022] Open
Abstract
Gap junction proteins are expressed in cancer stem cells and non-stem cancer cells of many tumors. As the morphology and assembly of gap junction channels are crucial for their function in intercellular communication, one focus of our review is to outline the data on gap junction plaque morphology available for cancer cells. Electron microscopic studies and freeze-fracture analyses on gap junction ultrastructure in cancer are summarized. As the presence of gap junctions is relevant in solid tumors, we exemplarily outline their role in glioblastomas and in breast cancer. These were also shown to contain cancer stem cells, which are an essential cause of tumor onset and of tumor transmission into metastases. For these processes, gap junctional communication was shown to be important and thus we summarize, how the expression of gap junction proteins and the resulting communication between cancer stem cells and their surrounding cells contributes to the dissemination of cancer stem cells via blood or lymphatic vessels. Based on their importance for tumors and metastases, future cancer-specific therapies are expected to address gap junction proteins. In turn, gap junctions also seem to contribute to the unattainability of cancer stem cells by certain treatments and might thus contribute to therapeutic resistance.
Collapse
Affiliation(s)
- Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Nadine Hainz
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
27
|
Zhang Q, Jia GJ, Zhang GB, Wang L, Wu Z, Jia W, Hao SY, Ni M, Li D, Wang K, Zhang JT. A Logistic Regression Model for Detecting the Presence of Malignant Progression in Atypical Meningiomas. World Neurosurg 2019; 126:e392-e401. [PMID: 30822595 DOI: 10.1016/j.wneu.2019.02.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To develop a method to distinguish atypical meningiomas (AMs) with malignant progression (MP) from primary AMs without a clinical history. METHODS The clinical, radiologic, and pathologic data of 33 previously Simpson grade I resected (if any) as well as no radiotherapy treated intracranial AMs between January 2008 and December 2015 were reviewed. Immunohistochemical staining for connexin 43 (Cx43) and Ki-67 was performed. Descriptive analysis and univariate and multivariate logistic regression analyses were used to explore independent predictors of MP. A multivariable logistic model was developed to estimate the risk of MP, and its diagnostic value was determined from a receiver operating characteristic curve. RESULTS There were 11 AMs (33.3%) with histopathologically confirmed MP from benign meningiomas. The other 22 (66.7%) were initially diagnosed AMs with no histopathologically confirmed MP during a median 60.5 months (range, 42-126 months) of follow-up. Univariate and multivariate logistic analyses showed that irregular tumor shape (P = 0.010) and low Cx43 expression (P = 0.010) were independent predictors of the presence of MP, and the predicted probability was calculated by the following formula: P = 1/[1+exp.{1.218-(3.202×Shape)+(3.814×Cx43)}]. P > 0.5 for an irregularly shaped (score 1) AM with low Cx43 expression (score 0) indicated a high probability of MP. The sensitivity, specificity, positive predictive value, negative predictive value, and overall predictive accuracy were 63.6, 95.6, 87.5, 84.0, and 84.8%, respectively. CONCLUSIONS Low Cx43 expression and irregular tumor shape were independent predictors of the presence of MP. The relevant logistic regression model was found to be effective in distinguishing MP-AMs from primary AMs.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Gui-Jun Jia
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Guo-Bin Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Shu-Yu Hao
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Ming Ni
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Da Li
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Jun-Ting Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China.
| |
Collapse
|
28
|
Connexin43 Suppresses Lung Cancer Stem Cells. Cancers (Basel) 2019; 11:cancers11020175. [PMID: 30717421 PMCID: PMC6406368 DOI: 10.3390/cancers11020175] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 02/07/2023] Open
Abstract
Alterations in gap junctions and their protein components, connexins, have been associated with neoplastic transformation and drug resistance, and more recently have been shown to play important roles in cancer stem cells (CSCs). However, there is less knowledge of connexins and gap junctions in lung CSCs. To address this, Connexin43 (Cx43), the major human lung epithelial gap junction protein, was expressed ectopically in poorly expressing National Cancer Institute-125 (NCI-H125) metastatic human lung adenocarcinoma cells, and phenotypic characteristics of malignant cells and abundance of CSCs were evaluated. The ectopic expression of Cx43 resulted in the formation of functional gap junctions; a more epithelial morphology; reduced proliferation, invasion, colony formation, tumorsphere formation, pluripotency marker expression, and percentage of aldehyde dehydrogenase (ALDH)-positive cells; and increased cisplatin sensitivity. Similarly, in NCI-H522 (human lung adenocarcinoma) and NCI-H661 (human lung large cell carcinoma) cell lines, which express Cx43 and functional gap junctions endogenously, the Cx43 content was lower in tumorspheres and ALDH-positive cells than in bulk cells. These results demonstrate that Cx43 can reverse several neoplastic characteristics and reduce the abundance of human lung CSCs.
Collapse
|
29
|
Shi G, Zheng X, Wu X, Wang S, Wang Y, Xing F. All-trans retinoic acid reverses epithelial-mesenchymal transition in paclitaxel-resistant cells by inhibiting nuclear factor kappa B and upregulating gap junctions. Cancer Sci 2018; 110:379-388. [PMID: 30375704 PMCID: PMC6317959 DOI: 10.1111/cas.13855] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 10/18/2018] [Accepted: 10/23/2018] [Indexed: 01/12/2023] Open
Abstract
Paclitaxel is a widely used chemotherapy drug, but development of resistance leads to treatment failure. Tumor cells that are treated with a sublethal dose of paclitaxel for a long period of time show the epithelial‐mesenchymal transition (EMT) phenotype, which leads to metastasis and resistance. All‐trans retinoic acid (ATRA) is always used in combination with paclitaxel and can reverse EMT in many types of cancer cells. The ability of ATRA to reverse EMT in chemoresistant cells is still unknown. In the present study, the ability of ATRA to reverse EMT in paclitaxel‐resistant cells was investigated. Three colorectal cancer cell lines, HCT116, LoVo and CT26, were treated with sublethal doses of paclitaxel to create resistant cell lines. Western blotting, immunocytochemistry, and “parachute” dye‐coupling assays showed that ATRA reverses EMT, inhibits nuclear factor kappa B (NF‐κΒ), and upregulates gap junctions in paclitaxel‐resistant cells. Scratch wound‐healing and Transwell assays showed that ATRA decreases the migration and invasion abilities of paclitaxel‐resistant cells. In addition, the CT26 cell line was used in the Balb/c pulmonary metastasis model to show that ATRA reduces metastasis of paclitaxel‐resistant cells in vivo. Given these data, ATRA may reverse EMT by inhibiting NF‐κΒ and upregulating gap junctions in paclitaxel‐resistant cells.
Collapse
Affiliation(s)
- Guiling Shi
- Tianjin Union Medical Center, Tianjin, China
| | | | - Xiaojing Wu
- Tianjin Union Medical Center, Tianjin, China
| | - Siqi Wang
- Tianjin Union Medical Center, Tianjin, China
| | - Yijia Wang
- Tianjin Union Medical Center, Tianjin, China.,State Key Laboratory of Medicinal Chemical Biology, NanKai University, Tianjin, China
| | - Fei Xing
- School of Physics and Optoelectronic Engineering, Shandong University of Technology, Zibo, China
| |
Collapse
|
30
|
Li K, Zhou H, Zhan L, Shi Z, Sun W, Liu D, Liu L, Liang D, Tan Y, Xu W, Xu E. Hypoxic Preconditioning Maintains GLT-1 Against Transient Global Cerebral Ischemia Through Upregulating Cx43 and Inhibiting c-Src. Front Mol Neurosci 2018; 11:344. [PMID: 30323740 PMCID: PMC6172853 DOI: 10.3389/fnmol.2018.00344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 09/03/2018] [Indexed: 01/06/2023] Open
Abstract
Transient global cerebral ischemia (tGCI) causes excessive release of glutamate from neurons. Astrocytic glutamate transporter-1 (GLT-1) and glutamine synthetase (GS) together play a predominant role in maintaining glutamate at normal extracellular concentrations. Though our previous studies reported the alleviation of tGCI-induced neuronal death by hypoxic preconditioning (HPC) in hippocampal Cornu Ammonis 1 (CA1) of adult rats, the underlying mechanism has not yet been fully elaborated. In this study, we aimed to investigate the roles of GLT-1 and GS in the neuroprotection mediated by HPC against tGCI and to ascertain whether these roles can be regulated by connexin 43 (Cx43) and cellular-Src (c-Src) activity. We found that HPC decreased the level of extracellular glutamate in CA1 after tGCI via maintenance of GLT-1 expression and GS activity. Inhibition of GLT-1 expression with dihydrokainate (DHK) or inhibition of GS activity with methionine sulfoximine (MSO) abolished the neuroprotection induced by HPC. Also, HPC markedly upregulated Cx43 and inhibited p-c-Src expression in CA1 after tGCI, whereas inhibition of Cx43 with Gap26 dramatically reversed this effect. Furthermore, inhibition of p-c-Src with 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo (3, 4-d) pyrimidine (PP2) decreased c-Src activity, increased protein levels of GLT-1 and Cx43, enhanced GS activity, and thus reduced extracellular glutamate level in CA1 after tGCI. Collectively, our data demonstrated that reduced extracellular glutamate induced by HPC against tGCI through preventing the reduction of GLT-1 expression and maintaining GS activity in hippocampal CA1, which was mediated by upregulating Cx43 expression and inhibiting c-Src activity.
Collapse
Affiliation(s)
- Kongping Li
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Huarong Zhou
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Lixuan Zhan
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Zhe Shi
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Weiwen Sun
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Dandan Liu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Liu Liu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Donghai Liang
- Department of Environmental Health Sciences, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Yafu Tan
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China.,Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wensheng Xu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - En Xu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| |
Collapse
|
31
|
Kuang JY, Guo YF, Chen Y, Wang J, Duan JJ, He XL, Li L, Yu SC, Bian XW. Connexin 43 C-terminus directly inhibits the hyperphosphorylation of Akt/ERK through protein-protein interactions in glioblastoma. Cancer Sci 2018; 109:2611-2622. [PMID: 29931708 PMCID: PMC6113504 DOI: 10.1111/cas.13707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/05/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
Although the deregulation of epidermal growth factor receptor (EGFR) is one of the most common molecular mechanisms of glioblastoma (GBM) pathogenesis, the efficacy of anti-EGFR therapy is limited. Additionally, response to anti-EGFR therapy is not solely dependent on EGFR expression and is more promising in patients with reduced activity of EGFR downstream signaling pathways. Thus, there is considerable interest in identifying the compensatory regulatory factors of the EGFR signaling pathway to improve the efficacy of anti-EGFR therapies for GBM. In this study, we confirmed the low efficacy of EGFR inhibitors in GBM patients by meta-analysis. We then identified a negative correlation between connexin 43 (Cx43) expression and Akt/ERK activation, which was caused by the direct interactions between Akt/ERK and Cx43. By comparing the interactions between Akt/ERK and Cx43 using a series of truncated and mutated Cx43 variants, we revealed that the residues T286/A305/Q308/Y313 and S272/S273 at the carboxy terminus of Cx43 are critical for its binding with Akt and ERK, respectively. In addition, Kaplan-Meier survival analysis using data from The Cancer Genome Atlas datasets indicated that the expression of Cx43 significantly improved the prognosis of GBM patients who express EGFR. Together, our results suggested that Cx43 acts as an inhibitory regulator of the activation of growth factor receptor downstream signaling pathways, indicating the potential of Cx43 as a marker for predicting the efficacy of EGFR inhibitor treatments for GBM. Targeting the interaction between the carboxy terminus of Cx43 and Akt/ERK could be an effective therapeutic strategy against GBM.
Collapse
Affiliation(s)
- Jing-Ya Kuang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Yu-Feng Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Ying Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Jun Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Jiang-Jie Duan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Xiao-Li He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Lin Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| |
Collapse
|
32
|
Iyyathurai J, Wang N, D'hondt C, Jiang JX, Leybaert L, Bultynck G. The SH3-binding domain of Cx43 participates in loop/tail interactions critical for Cx43-hemichannel activity. Cell Mol Life Sci 2018; 75:2059-2073. [PMID: 29218600 PMCID: PMC6318120 DOI: 10.1007/s00018-017-2722-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022]
Abstract
Connexin 43 (Cx43) hemichannels establish local signaling networks via the release of ATP and other molecules, but their excessive opening may result in cell death. Hence, the activity of Cx43-hemichannels ought to be critically controlled. This involves interactions between the C-terminal tail (CT) and the cytoplasmic loop (CL), more particularly the L2 domain within CL. Previous work revealed an important role for the last nine amino acids of the Cx43 CT by targeting the L2 domain, as these nine amino acids were sufficient to restore the activity of CT-truncated Cx43-hemichannels. However, we discovered that deletion of the last 19 amino acids of the CT only partially lowered the binding to the L2 domain, indicating that a second L2-binding region is present in the CT. We here provide evidence that the SH3-binding domain is another CT region that targets the L2 domain. At the functional level, the SH3-binding domain was able to restore the activity of CT-truncated Cx43-hemichannels and alleviate the inhibition of full-length Cx43-hemichannels by high intracellular Ca2+ concentration ([Ca2+]i) as demonstrated by various approaches including patch clamp studies of unitary Cx43-hemichannel activity. Finally, we show that in full-length Cx43-hemichannels, deletion of either the SH3-binding domain or the CT9 region suppresses the hemichannel activity, while deletion of both domains completely annihilates the hemichannel activity. These results demonstrate that the Cx43 SH3-binding domain, in addition to the CT9 region, critically controls hemichannel activity at high [Ca2+]i, which may be involved in pathological hemichannel opening.
Collapse
Affiliation(s)
- Jegan Iyyathurai
- Laboratory of Molecular and Cellular Signaling, Department Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O/N-1 Bus 802, Herestraat 49, 3000, Louvain, Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, 9000, Ghent, Belgium
| | - Catheleyne D'hondt
- Laboratory of Molecular and Cellular Signaling, Department Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O/N-1 Bus 802, Herestraat 49, 3000, Louvain, Belgium
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, 9000, Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O/N-1 Bus 802, Herestraat 49, 3000, Louvain, Belgium.
| |
Collapse
|
33
|
Grek CL, Sheng Z, Naus CC, Sin WC, Gourdie RG, Ghatnekar GG. Novel approach to temozolomide resistance in malignant glioma: connexin43-directed therapeutics. Curr Opin Pharmacol 2018; 41:79-88. [PMID: 29803991 DOI: 10.1016/j.coph.2018.05.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/30/2018] [Accepted: 05/03/2018] [Indexed: 01/03/2023]
Abstract
Resistance of malignant glioma, including glioblastoma (GBM), to the chemotherapeutic temozolomide (TMZ) remains a key obstacle in treatment strategies. The gap junction protein connexin43 (Cx43) has complex roles in the establishment, progression, and persistence of malignant glioma. Recent findings demonstrate that connexins play an important role in the microenvironment of malignant glioma and that Cx43 is capable of conferring chemotherapeutic resistance to GBM cells. Carboxyl-terminal Cx43 peptidomimetics show therapeutic promise in overcoming TMZ resistance via mechanisms that may include modulating junctional activity between tumor cells and peritumoral cells and/or downstream molecular signaling events mediated by Cx43 protein binding. High levels of intra-tumor and inter-tumor heterogeneity make it difficult to clearly define specific populations for Cx43-targeted therapy; hence, development of in vitro models that better mimic the microenvironment of malignant glioma, and the incorporation of patient-derived stem cells, could provide opportunities for patient-specific drug screening. This review summarizes recent advances in understanding the roles of Cx43 in malignant glioma, with a special focus on tumor microenvironment, TMZ resistance, and therapeutic opportunity offered by Cx43 peptidomimetics.
Collapse
Affiliation(s)
| | - Zhi Sheng
- Virginia Tech Carilion Research Institute, Roanoke, VA, USA; Faculty of Health Science, Virginia Tech, Blacksburg, VA, USA; Department of Biological Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA; Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Christian C Naus
- Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Wun Chey Sin
- Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Robert G Gourdie
- Virginia Tech Carilion Research Institute, Roanoke, VA, USA; Faculty of Health Science, Virginia Tech, Blacksburg, VA, USA; Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, VA, USA; Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | | |
Collapse
|
34
|
González-Sánchez A, Jaraíz-Rodríguez M, Domínguez-Prieto M, Herrero-González S, Medina JM, Tabernero A. Connexin43 recruits PTEN and Csk to inhibit c-Src activity in glioma cells and astrocytes. Oncotarget 2018; 7:49819-49833. [PMID: 27391443 PMCID: PMC5226550 DOI: 10.18632/oncotarget.10454] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 06/26/2016] [Indexed: 11/30/2022] Open
Abstract
Connexin43 (Cx43), the major protein forming gap junctions in astrocytes, is reduced in high-grade gliomas, where its ectopic expression exerts important effects, including the inhibition of the proto-oncogene tyrosine-protein kinase Src (c-Src). In this work we aimed to investigate the mechanism responsible for this effect. The inhibition of c-Src requires phosphorylation at tyrosine 527 mediated by C-terminal Src kinase (Csk) and dephosphorylation at tyrosine 416 mediated by phosphatases, such as phosphatase and tensin homolog (PTEN). Our results showed that the antiproliferative effect of Cx43 is reduced when Csk and PTEN are silenced in glioma cells, suggesting the involvement of both enzymes. Confocal microscopy and immunoprecipitation assays confirmed that Cx43, in addition to c-Src, binds to PTEN and Csk in glioma cells transfected with Cx43 and in astrocytes. Pull-down assays showed that region 266–283 in Cx43 is sufficient to recruit c-Src, PTEN and Csk and to inhibit the oncogenic activity of c-Src. As a result of c-Src inhibition, PTEN was increased with subsequent inactivation of Akt and reduction of proliferation of human glioblastoma stem cells. We conclude that the recruitment of Csk and PTEN to the region between residues 266 and 283 within the C-terminus of Cx43 leads to c-Src inhibition.
Collapse
Affiliation(s)
- Ana González-Sánchez
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, Spain
| | - Myriam Jaraíz-Rodríguez
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, Spain
| | - Marta Domínguez-Prieto
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, Spain
| | - Sandra Herrero-González
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, Spain
| | - José M Medina
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, Spain
| | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
35
|
Jaraíz-Rodríguez M, González-Sánchez A, García-Vicente L, Medina JM, Tabernero A. Biotinylated Cell-penetrating Peptides to Study Intracellular Protein-protein Interactions. J Vis Exp 2017:56457. [PMID: 29286477 PMCID: PMC5755618 DOI: 10.3791/56457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Here we present a protocol to study intracellular protein-protein interactions that is based on the widely used biotin-avidin pull-down system. The modification presented includes the combination of this technique with cell-penetrating sequences. We propose to design cell-penetrating baits that can be incubated with living cells instead of cell lysates and therefore the interactions found will reflect those that occur within the intracellular context. Connexin43 (Cx43), a protein that forms gap junction channels and hemichannels is down-regulated in high-grade gliomas. The Cx43 region comprising amino acids 266-283 is responsible for the inhibition of the oncogenic activity of c-Src in glioma cells. Here we use TAT as the cell-penetrating sequence, biotin as the pull-down tag and the region of Cx43 comprised between amino acids 266-283 as the target to find intracellular interactions in the hard-to-transfect human glioma stem cells. One of the limitations of the proposed method is that the molecule used as bait could fail to fold properly and, consequently, the interactions found could not be associated with the effect. However, this method can be especially interesting for the interactions involved in signal transduction pathways because they are usually carried out by intrinsically disordered regions and, therefore, they do not require an ordered folding. In addition, one of the advantages of the proposed method is that the relevance of each residue on the interaction can be easily studied. This is a modular system; therefore, other cell-penetrating sequences, other tags, and other intracellular targets can be employed. Finally, the scope of this protocol is far beyond protein-protein interaction because this system can be applied to other bioactive cargoes such as RNA sequences, nanoparticles, viruses or any molecule that can be transduced with cell-penetrating sequences and fused to pull-down tags to study their intracellular mechanism of action.
Collapse
Affiliation(s)
- Myriam Jaraíz-Rodríguez
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca
| | - Ana González-Sánchez
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca; Centre for Cancer Research & Cell Biology (CCRCB), School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast
| | - Laura García-Vicente
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca
| | - Jose M Medina
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca
| | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca;
| |
Collapse
|
36
|
Gangoso E, Talaverón R, Jaraíz-Rodríguez M, Domínguez-Prieto M, Ezan P, Koulakoff A, Medina JM, Giaume C, Tabernero A. A c-Src Inhibitor Peptide Based on Connexin43 Exerts Neuroprotective Effects through the Inhibition of Glial Hemichannel Activity. Front Mol Neurosci 2017; 10:418. [PMID: 29326548 PMCID: PMC5737028 DOI: 10.3389/fnmol.2017.00418] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 12/01/2017] [Indexed: 12/29/2022] Open
Abstract
The non-receptor tyrosine kinase c-Src is an important mediator in several signaling pathways related to neuroinflammation. Our previous study showed that cortical injection of kainic acid (KA) promoted a transient increase in c-Src activity in reactive astrocytes surrounding the neuronal lesion. As a cell-penetrating peptide based on connexin43 (Cx43), specifically TAT-Cx43266–283, inhibits Src activity, we investigated the effect of TAT-Cx43266–283 on neuronal death promoted by cortical KA injections in adult mice. As expected, KA promoted neuronal death, estimated by the reduction in NeuN-positive cells and reactive gliosis, characterized by the increase in glial fibrillary acidic protein (GFAP) expression. Interestingly, TAT-Cx43266–283 injected with KA diminished neuronal death and reactive gliosis compared to KA or KA+TAT injections. In order to gain insight into the neuroprotective mechanism, we used in vitro models. In primary cultured neurons, TAT-Cx43266–283 did not prevent neuronal death promoted by KA, but when neurons were grown on top of astrocytes, TAT-Cx43266–283 prevented neuronal death promoted by KA. These observations demonstrate the participation of astrocytes in the neuroprotective effect of TAT-Cx43266–283. Furthermore, the neuroprotective effect was also present in non-contact co-cultures, suggesting the contribution of soluble factors released by astrocytes. As glial hemichannel activity is associated with the release of several factors, such as ATP and glutamate, that cause neuronal death, we explored the participation of these channels on the neuroprotective effect of TAT-Cx43266–283. Our results confirmed that inhibitors of ATP and NMDA receptors prevented neuronal death in co-cultures treated with KA, suggesting the participation of astrocyte hemichannels in neurotoxicity. Furthermore, TAT-Cx43266–283 reduced hemichannel activity promoted by KA in neuron-astrocyte co-cultures as assessed by ethidium bromide (EtBr) uptake assay. In fact, TAT-Cx43266–283 and dasatinib, a potent c-Src inhibitor, strongly reduced the activation of astrocyte hemichannels. In conclusion, our results suggest that TAT-Cx43266–283 exerts a neuroprotective effect through the reduction of hemichannel activity likely mediated by c-Src in astrocytes. These data unveil a new role of c-Src in the regulation of Cx43-hemichannel activity that could be part of the mechanism by which astroglial c-Src participates in neuroinflammation.
Collapse
Affiliation(s)
- Ester Gangoso
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Collège de France, Université Pierre et Marie Curie, Paris, France
| | - Rocío Talaverón
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca, Spain
| | - Myriam Jaraíz-Rodríguez
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca, Spain
| | - Marta Domínguez-Prieto
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca, Spain
| | - Pascal Ezan
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Collège de France, Université Pierre et Marie Curie, Paris, France
| | - Annette Koulakoff
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Collège de France, Université Pierre et Marie Curie, Paris, France
| | - José M Medina
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca, Spain
| | - Christian Giaume
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Collège de France, Université Pierre et Marie Curie, Paris, France
| | - Arantxa Tabernero
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
37
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
38
|
Dong H, Zhou XW, Wang X, Yang Y, Luo JW, Liu YH, Mao Q. Complex role of connexin 43 in astrocytic tumors and possible promotion of glioma‑associated epileptic discharge (Review). Mol Med Rep 2017; 16:7890-7900. [PMID: 28983585 DOI: 10.3892/mmr.2017.7618] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 06/19/2017] [Indexed: 02/05/2023] Open
Abstract
Connexin (Cx)43 is a multifunction protein which forms gap junction channels and hemi‑channels. It also contains abundant binding domains which possess the ability to interact with certain Cx43‑associated proteins and therefore serve a fundamental role in various physiological and pathological functions. However, the understanding of the association between cancer and Cx43 along with Cx43‑gap junctions (GJ) remains unclear. All available data illustrate that Cx43 and its associated GJ serve important functions in cancers. The expression levels of Cx43 demonstrate a downward trend and an increase in the levels of malignancy, particularly in astrocytomas. The GJ intercellular communication activity in glioma cells can be adjusted via Cx43 phosphorylation and through the combination of Cx43 and its associated protein. Available evidence reveals Cx43 as a tumor‑inhibiting factor that suppresses glioma growth and proliferation. However, its mechanism is also regarded as complicated and ambiguous. Furthermore, it is apparent that Cx43‑GJ and the carboxyl tail may contribute to glioma growth and proliferation too. However, this valuable role could be weakened by its effects on migration and invasiveness. The detailed mechanism remains unclear and full of controversies. Cx43 can enhance the motor ability and invasiveness of astrocytic glioma cells. It is also able to influence glioma cells to detach from the tumor core to the peritumoral neocortex. This peritumoral region has recently been regarded as the basic focus of glioma‑associated seizure. Thus, Cx43 may take part in the onset and development of glioma‑associated epileptic discharge. In addition, change and increase of Cx43 expression in GJs has been observed in seizure perilesional tissue, which is associated with brain tumors. Cx43 or GJ/hemi‑channels exert enduring effects in the promotion of glioma‑associated epileptic release through direct mass effects and change of the tumor microenvironment. However, there are still a number of issues concerning this aspect that require further exploration. Cx43, as a potential treatment target against this incurable disease and its common symptom of epilepsy, requires further investigation.
Collapse
Affiliation(s)
- Hui Dong
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xing-Wang Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiang Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuan Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jie-Wen Luo
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan-Hui Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qing Mao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
39
|
Kou Y, Ji L, Wang H, Wang W, Zheng H, Zou J, Liu L, Qi X, Liu Z, Du B, Lu L. Connexin 43 upregulation by dioscin inhibits melanoma progression via suppressing malignancy and inducing M1 polarization. Int J Cancer 2017; 141:1690-1703. [DOI: 10.1002/ijc.30872] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/21/2017] [Accepted: 06/22/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Yu Kou
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
- Department of Pathology; School of Basic Medical Sciences, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Liyan Ji
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Haojia Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Wensheng Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Hongming Zheng
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Juan Zou
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Linxin Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Xiaoxiao Qi
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Biaoyan Du
- Department of Pathology; School of Basic Medical Sciences, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| | - Linlin Lu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine; Guangzhou 510006 China
| |
Collapse
|
40
|
Jaraíz-Rodríguez M, Tabernero MD, González-Tablas M, Otero A, Orfao A, Medina JM, Tabernero A. A Short Region of Connexin43 Reduces Human Glioma Stem Cell Migration, Invasion, and Survival through Src, PTEN, and FAK. Stem Cell Reports 2017; 9:451-463. [PMID: 28712848 PMCID: PMC5549880 DOI: 10.1016/j.stemcr.2017.06.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/13/2017] [Accepted: 06/14/2017] [Indexed: 12/22/2022] Open
Abstract
Connexin43 (CX43), a protein that forms gap junction channels and hemichannels in astrocytes, is downregulated in high-grade gliomas. Its relevance for glioma therapy has been thoroughly explored; however, its positive effects on proliferation are counterbalanced by its effects on migration and invasion. Here, we show that a cell-penetrating peptide based on CX43 (TAT-Cx43266-283) inhibited c-Src and focal adhesion kinase (FAK) and upregulated phosphatase and tensin homolog in glioma stem cells (GSCs) derived from patients. Consequently, TAT-Cx43266-283 reduced GSC motility, as analyzed by time-lapse microscopy, and strongly reduced their invasive ability. Interestingly, we investigated the effects of TAT-Cx43266-283 on freshly removed surgical specimens as undissociated glioblastoma blocks, which revealed a dramatic reduction in the growth, migration, and survival of these cells. In conclusion, a region of CX43 (amino acids 266–283) exerts an important anti-tumor effect in patient-derived glioblastoma models that includes impairment of GSC migration and invasion. TAT-Cx43266-283 exerts anti-tumor effects in patient-derived glioblastoma models TAT-Cx43266-283 targets Src, PTEN, and FAK TAT-Cx43266-283 inhibits glioma stem cell migration and invasion
Collapse
Affiliation(s)
- Myriam Jaraíz-Rodríguez
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, C/ Pintor Fernando Gallego 1, 37007 Salamanca, Spain
| | - Ma Dolores Tabernero
- Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL), Departamento de Medicina Universidad de Salamanca, 37007 Salamanca, Spain
| | - María González-Tablas
- Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL), Departamento de Medicina Universidad de Salamanca, 37007 Salamanca, Spain
| | - Alvaro Otero
- Neurosurgery Service, Hospital Universitario de Salamanca and IBSAL, 37007 Salamanca, Spain
| | - Alberto Orfao
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL), Departamento de Medicina Universidad de Salamanca, 37007 Salamanca, Spain
| | - Jose M Medina
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, C/ Pintor Fernando Gallego 1, 37007 Salamanca, Spain
| | - Arantxa Tabernero
- Instituto de Neurociencias de Castilla y León (INCYL), Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, C/ Pintor Fernando Gallego 1, 37007 Salamanca, Spain.
| |
Collapse
|
41
|
Rhett JM, Calder BW, Fann SA, Bainbridge H, Gourdie RG, Yost MJ. Mechanism of action of the anti-inflammatory connexin43 mimetic peptide JM2. Am J Physiol Cell Physiol 2017; 313:C314-C326. [PMID: 28701358 PMCID: PMC5625091 DOI: 10.1152/ajpcell.00229.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/31/2022]
Abstract
Connexin-based therapeutics have shown the potential for therapeutic efficacy in improving wound healing. Our previous work demonstrated that the connexin43 (Cx43) mimetic peptide juxtamembrane 2 (JM2) reduced the acute inflammatory response to a submuscular implant model by inhibiting purinergic signaling. Given the prospective application in improving tissue-engineered construct tolerance that these results indicated, we sought to determine the mechanism of action for JM2 in the present study. Using confocal microscopy, a gap-FRAP cell communication assay, and an ethidium bromide uptake assay of hemichannel function we found that the peptide reduced cell surface Cx43 levels, Cx43 gap junction (GJ) size, GJ communication, and hemichannel activity. JM2 is based on the sequence of the Cx43 microtubule binding domain, and microtubules have a confirmed role in intracellular trafficking of Cx43 vesicles. Therefore, we tested the effect of JM2 on Cx43-microtubule interaction and microtubule polymerization. We found that JM2 enhanced Cx43-microtubule interaction and that microtubule polymerization was significantly enhanced. Taken together, these data suggest that JM2 inhibits trafficking of Cx43 to the cell surface by promoting irrelevant microtubule polymerization and thereby reduces the number of hemichannels in the plasma membrane available to participate in proinflammatory purinergic signaling. Importantly, this work indicates that JM2 may have therapeutic value in the treatment of proliferative diseases such as cancer. We conclude that the targeted action of JM2 on Cx43 channels may improve the tolerance of implanted tissue-engineered constructs against the innate inflammatory response.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina;
| | - Bennett W Calder
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Stephen A Fann
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Heather Bainbridge
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, Roanoke, Virginia; and
| | - Michael J Yost
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina.,Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
42
|
Mesnil M, Aasen T, Boucher J, Chépied A, Cronier L, Defamie N, Kameritsch P, Laird DW, Lampe PD, Lathia JD, Leithe E, Mehta PP, Monvoisin A, Pogoda K, Sin WC, Tabernero A, Yamasaki H, Yeh ES, Dagli MLZ, Naus CC. An update on minding the gap in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:237-243. [PMID: 28655619 DOI: 10.1016/j.bbamem.2017.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 01/08/2023]
Abstract
This article is a report of the "International Colloquium on Gap junctions: 50Years of Impact on Cancer" that was held 8-9 September 2016, at the Amphitheater "Pôle Biologie Santé" of the University of Poitiers (Poitiers, France). The colloquium was organized by M Mesnil (Université de Poitiers, Poitiers, France) and C Naus (University of British Columbia, Vancouver, Canada) to celebrate the 50th anniversary of the seminal work published in 1966 by Loewenstein and Kanno [Intercellular communication and the control of tissue growth: lack of communication between cancer cells, Nature, 116 (1966) 1248-1249] which initiated studies on the involvement of gap junctions in carcinogenesis. During the colloquium, 15 participants presented reviews or research updates in the field which are summarized below.
Collapse
Affiliation(s)
- Marc Mesnil
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France.
| | - Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, 08035 Barcelona, Spain
| | - Jonathan Boucher
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Amandine Chépied
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Laurent Cronier
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Norah Defamie
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Petra Kameritsch
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario N6A 5C1, Canada
| | - Paul D Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Justin D Lathia
- Cleveland Clinic, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, and Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, NO-0424 Oslo, Norway
| | - Parmender P Mehta
- Department of Biochemistry and Molecular Biology, University of Nebraska, Medical Center, Omaha, NE 68198, USA
| | - Arnaud Monvoisin
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, cedex 09, France
| | - Kristin Pogoda
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany
| | - Wun-Chey Sin
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Arantxa Tabernero
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca 37007, Spain
| | | | - Elizabeth S Yeh
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29412, USA
| | - Maria Lucia Zaidan Dagli
- Laboratory of Experimental and Comparative Oncology, School of Veterinary Medicine and Animal Science of the University of São Paulo, São Paulo, SP CEP 05508-900, Brazil
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
43
|
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
|
44
|
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
|
45
|
Naus CC, Giaume C. Bridging the gap to therapeutic strategies based on connexin/pannexin biology. J Transl Med 2016; 14:330. [PMID: 27899102 PMCID: PMC5129631 DOI: 10.1186/s12967-016-1089-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/18/2016] [Indexed: 01/02/2023] Open
Abstract
A unique workshop was recently held focusing on enhancing collaborations leading to identify and update the development of therapeutic strategies targeting connexin/pannexin large pore channels. Basic scientists exploring the functions of these channels in various pathologies gathered together with leading pharma companies which are targeting gap junction proteins for specific therapeutic applications. This highlights how paths of discovery research can converge with therapeutic strategies in innovative ways to enhance target identification and validation.
Collapse
Affiliation(s)
- Christian C Naus
- Department of Cellular and Physiological Sciences, Faculty of Medicine, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Christian Giaume
- ICIRB, CNRS UMR7241/INSERM U1050, Collège de France, Paris Cedex 05, France
| |
Collapse
|
46
|
Han Y, Zhou L, Wu T, Huang Y, Cheng Z, Li X, Sun T, Zhou Y, Du Z. Downregulation of lncRNA-MALAT1 Affects Proliferation and the Expression of Stemness Markers in Glioma Stem Cell Line SHG139S. Cell Mol Neurobiol 2016; 36:1097-107. [PMID: 26649728 DOI: 10.1007/s10571-015-0303-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 11/07/2015] [Indexed: 10/22/2022]
Abstract
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is among the most abundant and highly conserved lncRNAs, which has been detected in a wide variety of human tumors, including gastric cancer, gallbladder cancer, and so on. Previous research has showed that MALAT1 can activate LTBP3 gene in mesenchymal stem cells. However, the specific roles of MALAT1 in glioma stem cells (GSCs) remain unclear. In this study, we aimed to identify the effects of MALAT1 on proliferation and the expression of stemness markers on glioma stem cell line SHG139S. Our results showed that downregulation of MALAT1 suppressed the expression of Sox2 and Nestin which are related to stemness, while downregulation of MALAT1 promoted the proliferation in SHG139S. Further research on the underlying mechanism showed that the effects of MALAT1 downregulation on SHG139S were through regulating ERK/MAPk signaling activity. And we also found that downregulation of MALAT1 could activate ERK/MAPK signaling and promoted proliferation in SHG139 cells. These findings show that MALAT1 plays an important role in regulating the expression of stemness markers and proliferation of SHG139S, and provide a new research direction to target the progression of GSCs.
Collapse
Affiliation(s)
- Yong Han
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Liang Zhou
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Tingfeng Wu
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Yulun Huang
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Zhe Cheng
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Xuetao Li
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Ting Sun
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Youxin Zhou
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China.
| | - Ziwei Du
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, People's Republic of China
| |
Collapse
|
47
|
Peng H, Park JK, Katsnelson J, Kaplan N, Yang W, Getsios S, Lavker RM. microRNA-103/107 Family Regulates Multiple Epithelial Stem Cell Characteristics. Stem Cells 2016; 33:1642-56. [PMID: 25639731 DOI: 10.1002/stem.1962] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 01/14/2015] [Indexed: 12/28/2022]
Abstract
The stem cell niche is thought to affect cell cycle quiescence, proliferative capacity, and communication between stem cells and their neighbors. How these activities are controlled is not completely understood. Here we define a microRNA family (miRs-103/107) preferentially expressed in the stem cell-enriched limbal epithelium that regulates and integrates these stem cell characteristics. miRs-103/107 target the ribosomal kinase p90RSK2, thereby arresting cells in G0/G1 and contributing to a slow-cycling phenotype. Furthermore, miRs-103/107 increase the proliferative capacity of keratinocytes by targeting Wnt3a, which enhances Sox9 and YAP1 levels and thus promotes a stem cell phenotype. This miRNA family also regulates keratinocyte cell-cell communication by targeting: (a) the scaffolding protein NEDD9, preserving E-cadherin-mediated cell adhesion; and (b) the tyrosine phosphatase PTPRM, which negatively regulates connexin 43-based gap junctions. We propose that such regulation of cell communication and adhesion molecules maintains the integrity of the stem cell niche ultimately preserving self-renewal, a hallmark of epithelial stem cells.
Collapse
Affiliation(s)
- Han Peng
- Department of Dermatology, Northwestern University, Chicago, Illinois, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Upregulation of connexin43 contributes to PX-12-induced oxidative cell death. Tumour Biol 2015; 37:7535-46. [PMID: 26684802 DOI: 10.1007/s13277-015-4620-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/10/2015] [Indexed: 01/24/2023] Open
Abstract
Thioredoxin (Trx) is a small redox protein that underlies aggressive tumor growth and resistance to chemotherapy. Inhibition of Trx with the chemical inhibitor PX-12 suppresses tumor growth and induces cell apoptosis. Currently, the mechanism underlying the therapeutic actions of PX-12 and the molecules influencing cell susceptibility to PX-12 are incompletely understood. Given that connexin43 (Cx43), a tumor suppressor, regulates tumor cell susceptibility to chemotherapy, we examined the possible involvement of Cx43 in PX-12-induced cell death. Exposure of cells to PX-12 led to a loss of cell viability, which was associated with the activation of oxidative sensitive c-Jun N-terminal kinase (JNK). Inhibition of JNK or supplement of cells with anti-oxidants prevented the cell-killing action of PX-12. The forced expression of Cx43 in normal and tumor cells increased cell sensitivity to PX-12-induced JNK activation and cell death. In contrast, the downregulation of Cx43 with siRNA or the suppression of gap junctions with chemical inhibitors attenuated JNK activation and enhanced cell resistance to PX-12. Further analysis revealed that PX-12 at low concentrations induced a JNK-dependent elevation in the Cx43 protein, which was also preventable by supplementing the cells with anti-oxidants. Our results thus indicate that Cx43 is a determinant in the regulation of cell susceptibility to PX-12 and that the upregulation of Cx43 may be an additional mechanism by which PX-12 exerts its anti-tumor actions.
Collapse
|
49
|
Zhang F, Xu CL, Liu CM. Drug delivery strategies to enhance the permeability of the blood-brain barrier for treatment of glioma. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2089-100. [PMID: 25926719 PMCID: PMC4403597 DOI: 10.2147/dddt.s79592] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gliomas are amongst the most insidious and destructive types of brain cancer and are associated with a poor prognosis, frequent recurrences, and extremely high lethality despite combination treatment of surgery, radiotherapy, and chemotherapy. The existence of the blood–brain barrier (BBB) restricts the delivery of therapeutic molecules into the brain and offers the clinical efficacy of many pharmaceuticals that have been demonstrated to be effective for other kinds of tumors. This challenge emphasizes the need to be able to deliver drugs effectively across the BBB to reach the brain parenchyma. Enhancement of the permeability of the BBB and being able to transport drugs across it has been shown to be a promising strategy to improve drug absorption and treatment efficacy. This review highlights the innovative technologies that have been introduced to enhance the permeability of the BBB and to obtain an optimal distribution and concentration of drugs in the brain to treat gliomas, such as nanotechniques, hyperthermia techniques, receptor-mediated transport, cell-penetrating peptides, and cell-mediated delivery.
Collapse
Affiliation(s)
- Fang Zhang
- School of Pharmacy, National First-Class Key Discipline for Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Chun-Lei Xu
- School of Pharmacy, National First-Class Key Discipline for Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Chun-Mei Liu
- School of Pharmacy, National First-Class Key Discipline for Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| |
Collapse
|
50
|
Li Y, Wang H, Sun T, Chen J, Guo L, Shen H, Du Z, Zhou Y. Biological characteristics of a new human glioma cell line transformed into A2B5(+) stem cells. Mol Cancer 2015; 14:75. [PMID: 25879429 PMCID: PMC4392480 DOI: 10.1186/s12943-015-0343-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/13/2015] [Indexed: 11/30/2022] Open
Abstract
Objective The new glioma cell line SHG-139 was established and its phenotype, tumorigenicity, pathological characteristics, derived stem cells SHG139S were studied. Methods Immunohistochemistry was used to assess expressions in the patient and mouse tumor tissues, SHG-139 and SHG-139S. Primary SHG-139 culture was performed, cell proliferation, cell cycle and genetic characteristics were assessed. MiRNA (Micro RNA) and LncRNA (Long non-coding RNA) microarray was performed. Results We found that the glioma tissue was positive for A2B5 (Glial precursors ganglioside), GFAP (Glial fibrillary acidic protein), S-100 (Acid calcium bingding protein), VEGF (Vascular endothelial growth factor), VEGFR (Vascular endothelial growth factor receptor) and negative for Ki-67 (Nuclcar- associated antigen). SHG-139 proliferated significantly within 24h; its total number of chromosomes was 68; ratios of SHG-139 and SHG-139S cells in G1 phase were highest. SHG-139 cells were positive for A2B5, GalC (Galactocerebrosides), GFAP, S-100 and Vimentin, while SHG-139S cells were positive for A2B5, Nestin, and NG2 (Neuron-glia antigen2), and negative for Vimentin and IDHR132H (Isocitrate dehydrogenase); cells rarely stained for CD133 (Cluster of differentiation133). SHG-139 intracranial xenografts expressed GFAP, but no overt oligodendroglioma was observed. In SHG-139S xenografts, GFAP and S-100 were expressed, while CD133 was not detected; a few A2B5+ cells were found at tumor edges, and typical oligodendroglioma were obtained. In addition, SHG-139S xenograft tumors were more aggressive than those of SHG-139. Anti-mouse CD31 (Cluster of differentiation31) staining revealed murine vessels at the border between xenograft tumor and normal brain tissue; Anti-human CD34 (Cluster of differentiation34) staining was negative. Biochip technology of SHG139S showed several miRNA and lncRNA were differently expressed in SHG139 and SHG139S. Conclusions SHG-139 was an astroglioma cell line which yielded stem cells SHG-139S. SHG-139S cells constituted an A2B5+/CD133− GSC subgroup. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0343-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanyan Li
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Hangzhou Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Ting Sun
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Jinming Chen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Haitao Shen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Ziwei Du
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Youxin Zhou
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, China.
| |
Collapse
|