1
|
Yasarbas SS, Inal E, Yildirim MA, Dubrac S, Lamartine J, Mese G. Connexins in epidermal health and diseases: insights into their mutations, implications, and therapeutic solutions. Front Physiol 2024; 15:1346971. [PMID: 38827992 PMCID: PMC11140265 DOI: 10.3389/fphys.2024.1346971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/15/2024] [Indexed: 06/05/2024] Open
Abstract
The epidermis, the outermost layer of the skin, serves as a protective barrier against external factors. Epidermal differentiation, a tightly regulated process essential for epidermal homeostasis, epidermal barrier formation and skin integrity maintenance, is orchestrated by several players, including signaling molecules, calcium gradient and junctional complexes such as gap junctions (GJs). GJ proteins, known as connexins facilitate cell-to-cell communication between adjacent keratinocytes. Connexins can function as either hemichannels or GJs, depending on their interaction with other connexons from neighboring keratinocytes. These channels enable the transport of metabolites, cAMP, microRNAs, and ions, including Ca2+, across cell membranes. At least ten distinct connexins are expressed within the epidermis and mutations in at least five of them has been linked to various skin disorders. Connexin mutations may cause aberrant channel activity by altering their synthesis, their gating properties, their intracellular trafficking, and the assembly of hemichannels and GJ channels. In addition to mutations, connexin expression is dysregulated in other skin conditions including psoriasis, chronic wound and skin cancers, indicating the crucial role of connexins in skin homeostasis. Current treatment options for conditions with mutant or altered connexins are limited and primarily focus on symptom management. Several therapeutics, including non-peptide chemicals, antibodies, mimetic peptides and allele-specific small interfering RNAs are promising in treating connexin-related skin disorders. Since connexins play crucial roles in maintaining epidermal homeostasis as shown with linkage to a range of skin disorders and cancer, further investigations are warranted to decipher the molecular and cellular alterations within cells due to mutations or altered expression, leading to abnormal proliferation and differentiation. This would also help characterize the roles of each isoform in skin homeostasis, in addition to the development of innovative therapeutic interventions. This review highlights the critical functions of connexins in the epidermis and the association between connexins and skin disorders, and discusses potential therapeutic options.
Collapse
Affiliation(s)
- S. Suheda Yasarbas
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Izmir, Turkiye
| | - Ece Inal
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Izmir, Turkiye
| | - M. Azra Yildirim
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Izmir, Turkiye
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jérôme Lamartine
- Skin Functional Integrity Group, Laboratory for Tissue Biology and Therapeutics Engineering (LBTI) CNRS UMR5305, University of Lyon, Lyon, France
| | - Gulistan Mese
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Izmir, Turkiye
| |
Collapse
|
2
|
Montgomery J, Richardson WJ, Marsh S, Rhett JM, Bustos F, Degen K, Ghatnekar GS, Grek CL, Jourdan LJ, Holmes JW, Gourdie RG. The connexin 43 carboxyl terminal mimetic peptide αCT1 prompts differentiation of a collagen scar matrix in humans resembling unwounded skin. FASEB J 2021; 35:e21762. [PMID: 34246197 DOI: 10.1096/fj.202001881r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 06/02/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022]
Abstract
Phase II clinical trials have reported that acute treatment of surgical skin wounds with the therapeutic peptide alpha Connexin Carboxy-Terminus 1 (αCT1) improves cutaneous scar appearance by 47% 9-month postsurgery. While Cx43 and ZO-1 have been identified as molecular targets of αCT1, the mode-of-action of the peptide in scar mitigation at cellular and tissue levels remains to be further characterized. Scar histoarchitecture in αCT1 and vehicle-control treated skin wounds within the same patient were compared using biopsies from a Phase I clinical trial at 29-day postwounding. The sole effect on scar structure of a range of epidermal and dermal variables examined was that αCT1-treated scars had less alignment of collagen fibers relative to control wounds-a characteristic that resembles unwounded skin. The with-in subject effect of αCT1 on scar collagen order observed in Phase I testing in humans was recapitulated in Sprague-Dawley rats and the IAF hairless guinea pig. Transient increase in histologic collagen density in response to αCT1 was also observed in both animal models. Mouse NIH 3T3 fibroblasts and primary human dermal fibroblasts treated with αCT1 in vitro showed more rapid closure in scratch wound assays, with individual cells showing decreased directionality in movement. An agent-based computational model parameterized with fibroblast motility data predicted collagen alignments in simulated scars consistent with that observed experimentally in human and the animal models. In conclusion, αCT1 prompts decreased directionality of fibroblast movement and the generation of a 3D collagen matrix postwounding that is similar to unwounded skin-changes that correlate with long-term improvement in scar appearance.
Collapse
Affiliation(s)
- Jade Montgomery
- Fralin Biomedical Research Institute, Center for Vascular and Heart Research, Virginia Tech Carilion, Roanoke, VA, USA.,Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | | | - Spencer Marsh
- Fralin Biomedical Research Institute, Center for Vascular and Heart Research, Virginia Tech Carilion, Roanoke, VA, USA
| | - J Matthew Rhett
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Francis Bustos
- Fralin Biomedical Research Institute, Center for Vascular and Heart Research, Virginia Tech Carilion, Roanoke, VA, USA.,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Katherine Degen
- Fralin Biomedical Research Institute, Center for Vascular and Heart Research, Virginia Tech Carilion, Roanoke, VA, USA.,Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | | | | | - L Jane Jourdan
- Fralin Biomedical Research Institute, Center for Vascular and Heart Research, Virginia Tech Carilion, Roanoke, VA, USA
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Robert G Gourdie
- Fralin Biomedical Research Institute, Center for Vascular and Heart Research, Virginia Tech Carilion, Roanoke, VA, USA.,Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| |
Collapse
|
3
|
Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int J Mol Sci 2021; 22:ijms22094413. [PMID: 33922534 PMCID: PMC8122935 DOI: 10.3390/ijms22094413] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Collapse
|
4
|
Varela-Eirín M, Carpintero-Fernández P, Sánchez-Temprano A, Varela-Vázquez A, Paíno CL, Casado-Díaz A, Continente AC, Mato V, Fonseca E, Kandouz M, Blanco A, Caeiro JR, Mayán MD. Senolytic activity of small molecular polyphenols from olive restores chondrocyte redifferentiation and promotes a pro-regenerative environment in osteoarthritis. Aging (Albany NY) 2020; 12:15882-15905. [PMID: 32745074 PMCID: PMC7485729 DOI: 10.18632/aging.103801] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022]
Abstract
Articular cartilage and synovial tissue from patients with osteoarthritis (OA) show an overactivity of connexin43 (Cx43) and accumulation of senescent cells associated with disrupted tissue regeneration and disease progression. The aim of this study was to determine the effect of oleuropein on Cx43 and cellular senescence for tissue engineering and regenerative medicine strategies for OA treatment. Oleuropein regulates Cx43 promoter activity and enhances the propensity of hMSCs to differentiate into chondrocytes and bone cells, reducing adipogenesis. This small molecule reduce Cx43 levels and decrease Twist-1 activity in osteoarthritic chondrocytes (OACs), leading to redifferentiation, restoring the synthesis of cartilage ECM components (Col2A1 and proteoglycans), and reducing the inflammatory and catabolic factors mediated by NF-kB (IL-1ß, IL-6, COX-2 and MMP-3), in addition to lowering cellular senescence in OACs, synovial and bone cells. Our in vitro results demonstrate the use of olive-derived polyphenols, such as oleuropein, as potentially effective therapeutic agents to improve chondrogenesis of hMSCs, to induce chondrocyte re-differentiation in OACs and clearing out senescent cells in joint tissues in order to prevent or stop the progression of the disease.
Collapse
Affiliation(s)
- Marta Varela-Eirín
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, A Coruña, Spain
| | - Paula Carpintero-Fernández
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, A Coruña, Spain
| | - Agustín Sánchez-Temprano
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, A Coruña, Spain
| | - Adrián Varela-Vázquez
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, A Coruña, Spain
| | - Carlos Luis Paíno
- Neurobiology-Research Service, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Antonio Casado-Díaz
- UGC Endocrinology and Nutrition, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Hospital Universitario Reina Sofía - CIBERFES, Universidad de Córdoba, Córdoba, Spain
| | - Alfonso Calañas Continente
- UGC Endocrinology and Nutrition, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Hospital Universitario Reina Sofía - CIBERFES, Universidad de Córdoba, Córdoba, Spain
| | - Virginia Mato
- Centre for Medical Informatics and Radiological Diagnosis, Universidade da Coruña, A Coruña, Spain
| | - Eduardo Fonseca
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, A Coruña, Spain
| | - Mustapha Kandouz
- Department of Pathology, School of Medicine, Wayne State University, Detroit, MI 48202, USA
| | - Alfonso Blanco
- Flow Cytometry Core Technologies, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - José Ramón Caeiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Choupana s/n, Santiago de Compostela, Spain
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, A Coruña, Spain
| |
Collapse
|
5
|
Connexin 43 Deficiency Is Associated with Reduced Myocardial Scar Size and Attenuated TGFβ1 Signaling after Transient Coronary Occlusion in Conditional Knock-Out Mice. Biomolecules 2020; 10:biom10040651. [PMID: 32340244 PMCID: PMC7226061 DOI: 10.3390/biom10040651] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022] Open
Abstract
Previous studies demonstrated a reduction in myocardial scar size in heterozygous Cx43+/- mice subjected to permanent coronary occlusion. However, patients presenting with ST segment elevation myocardial infarction often undergo rapid coronary revascularization leading to prompt restoration of coronary flow. Therefore, we aimed to assess changes in scar size and left ventricular remodeling following transient myocardial ischemia (45 min) followed by 14 days of reperfusion using Cx43fl/fl (controls) and Cx43Cre-ER(T)/fl inducible knock-out (Cx43 content: 50%) mice treated with vehicle or 4-hydroxytamoxifen (4-OHT) to induce a Cre-ER(T)-mediated global deletion of the Cx43 floxed allele. The scar area (picrosirius red), measured 14 days after transient coronary occlusion, was similarly reduced in both vehicle and 4-OHT-treated Cx43Cre-ER(T)/fl mice, compared to Cx43fl/fl animals, having normal Cx43 levels (15.78% ± 3.42% and 16.54% ± 2.31% vs. 25.40% ± 3.14% and 22.43% ± 3.88% in vehicle and 4-OHT-treated mice, respectively, p = 0.027). Left ventricular dilatation was significantly attenuated in both Cx43-deficient groups (p = 0.037 for left ventricular end-diastolic diameter). These protective effects were correlated with an attenuated enhancement in pro-transforming growth factor beta 1 (TGFβ1) expression after reperfusion. In conclusion, our data demonstrate that Cx43 deficiency induces a protective effect on scar formation after transient coronary occlusion in mice, an effect associated with reduced left ventricular remodeling and attenuated enhancement in pro-TGFβ1 expression.
Collapse
|
6
|
Opposite Effects of Moderate and Extreme Cx43 Deficiency in Conditional Cx43-Deficient Mice on Angiotensin II-Induced Cardiac Fibrosis. Cells 2019; 8:cells8101299. [PMID: 31652649 PMCID: PMC6830333 DOI: 10.3390/cells8101299] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 01/27/2023] Open
Abstract
Connexin 43 (Cx43) is essential for cardiac electrical coupling, but its effects on myocardial fibrosis is controversial. Here, we analyzed the role of Cx43 in myocardial fibrosis caused by angiotensin II (AngII) using Cx43fl/fl and Cx43Cre-ER(T)/fl inducible knock-out (Cx43 content: 50%) mice treated with vehicle or 4-hydroxytamoxifen (4-OHT) to induce a Cre-ER(T)-mediated global deletion of the Cx43 floxed allele. Myocardial collagen content was enhanced by AngII in all groups (n = 8–10/group, p < 0.05). However, animals with partial Cx43 deficiency (vehicle-treated Cx43Cre-ER(T)/fl) had a significantly higher AngII-induced collagen accumulation that reverted when treated with 4-OHT, which abolished Cx43 expression. The exaggerated fibrotic response to AngII in partially deficient Cx43Cre-ER(T)/fl mice was associated with enhanced p38 MAPK activation and was not evident in Cx43 heterozygous (Cx43+/-) mice. In contrast, normalization of interstitial collagen in 4-OHT-treated Cx43Cre-ER(T)/fl animals correlated with enhanced MMP-9 activity, IL-6 and NOX2 mRNA expression, and macrophage content, and with reduced α-SMA and SM22α in isolated fibroblasts. In conclusion, our data demonstrates an exaggerated, p38 MAPK-dependent, fibrotic response to AngII in partially deficient Cx43Cre-ER(T)/fl mice, and a paradoxical normalization of collagen deposition in animals with an almost complete Cx43 ablation, an effect associated with increased MMP-9 activity and inflammatory response and reduced fibroblasts differentiation.
Collapse
|
7
|
Jiang J, Hoagland D, Palatinus JA, He H, Iyyathurai J, Jourdan LJ, Bultynck G, Wang Z, Zhang Z, Schey K, Poelzing S, McGowan FX, Gourdie RG. Interaction of α Carboxyl Terminus 1 Peptide With the Connexin 43 Carboxyl Terminus Preserves Left Ventricular Function After Ischemia-Reperfusion Injury. J Am Heart Assoc 2019; 8:e012385. [PMID: 31422747 PMCID: PMC6759879 DOI: 10.1161/jaha.119.012385] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background α Carboxyl terminus 1 (αCT1) is a 25–amino acid therapeutic peptide incorporating the zonula occludens‐1 (ZO‐1)–binding domain of connexin 43 (Cx43) that is currently in phase 3 clinical testing on chronic wounds. In mice, we reported that αCT1 reduced arrhythmias after cardiac injury, accompanied by increases in protein kinase Cε phosphorylation of Cx43 at serine 368. Herein, we characterize detailed molecular mode of action of αCT1 in mitigating cardiac ischemia‐reperfusion injury. Methods and Results To study αCT1‐mediated increases in phosphorylation of Cx43 at serine 368, we undertook mass spectrometry of protein kinase Cε phosphorylation assay reactants. This indicated potential interaction between negatively charged residues in the αCT1 Asp‐Asp‐Leu‐Glu‐Iso sequence and lysines (Lys345, Lys346) in an α‐helical sequence (helix 2) within the Cx43‐CT. In silico modeling provided further support for this interaction, indicating that αCT1 may interact with both Cx43 and ZO‐1. Using surface plasmon resonance, thermal shift, and phosphorylation assays, we characterized a series of αCT1 variants, identifying peptides that interacted with either ZO‐1–postsynaptic density‐95/disks large/zonula occludens‐1 2 or Cx43‐CT, but with limited or no ability to bind both molecules. Only peptides competent to interact with Cx43‐CT, but not ZO‐1–postsynaptic density‐95/disks large/zonula occludens‐1 2 alone, prompted increased pS368 phosphorylation. Moreover, in an ex vivo mouse model of ischemia‐reperfusion injury, preischemic infusion only with those peptides competent to bind Cx43 preserved ventricular function after ischemia‐reperfusion. Interestingly, a short 9–amino acid variant of αCT1 (αCT11) demonstrated potent cardioprotective effects when infused either before or after ischemic injury. Conclusions Interaction of αCT1 with the Cx43, but not ZO‐1, is correlated with cardioprotection. Pharmacophores targeting Cx43‐CT could provide a translational approach to preserving heart function after ischemic injury.
Collapse
Affiliation(s)
- Jingbo Jiang
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA.,Shenzhen Children's Hospital Shenzhen China.,Department of Pediatric Cardiology Guangdong Cardiovascular Institute Guangdong General Hospital Guangdong Academy of Medical Sciences Guangzhou China
| | - Daniel Hoagland
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA
| | - Joseph A Palatinus
- Cedars-Sinai Heart Smidt Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Huamei He
- Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia and University of Pennsylvania Philadelphia PA
| | - Jegan Iyyathurai
- Department Cellular and Molecular Medicine KU Leuven Laboratory of Molecular and Cellular Signaling Leuven Belgium
| | - L Jane Jourdan
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA
| | - Geert Bultynck
- Department Cellular and Molecular Medicine KU Leuven Laboratory of Molecular and Cellular Signaling Leuven Belgium
| | - Zhen Wang
- Department of Biochemistry Vanderbilt University School of Medicine Nashville TN
| | - Zhiwei Zhang
- Department of Pediatric Cardiology Guangdong Cardiovascular Institute Guangdong General Hospital Guangdong Academy of Medical Sciences Guangzhou China
| | - Kevin Schey
- Department of Biochemistry Vanderbilt University School of Medicine Nashville TN
| | - Steven Poelzing
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA.,Department of Biomedical Engineering and Mechanics Virginia Tech Blacksburg VA
| | - Francis X McGowan
- Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia and University of Pennsylvania Philadelphia PA
| | - Robert G Gourdie
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research Virginia Tech Blacksburg VA.,Department of Biomedical Engineering and Mechanics Virginia Tech Blacksburg VA
| |
Collapse
|
8
|
The Role of Connexin-43 in the Inflammatory Process: A New Potential Therapy to Influence Keratitis. J Ophthalmol 2019; 2019:9312827. [PMID: 30805212 PMCID: PMC6360563 DOI: 10.1155/2019/9312827] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/12/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022] Open
Abstract
The studies outlined in this review highlight the relationship between inflammatory signaling molecules and connexin-43 (Cx43). Gap junction (GJ) channels and hemichannels (HCs) participate in the metabolic activity between intra- and extracellular space. Some ions and small molecules are exchanged from cell to cell or cell to extracellular space to affect the process of inflammation via GJ. We analyzed the effects of signaling molecules, such as innate immunity messengers, transcription factors, LPS, cytokine, inflammatory chemokines, and MMPs, on Cx43 expression during the inflammatory process. At the same time, we found that these signaling molecules play a critical role in the pathogenesis of keratitis. Thus, we assessed the function of Cx43 during inflammatory corneal disease. Corneal healing plays an essential role in the late stage of keratitis. We found that Cx43 is involved in wound healing. Studies have shown that the decrease of Cx43 can decrease the time of healing. We also report several Cx43 mimic peptides which can inhibit the activity of Cx43 Hc to mediate the releasing of adenosine triphosphate (ATP), which may in turn influence the inflammatory process.
Collapse
|
9
|
Trincot CE, Xu W, Zhang H, Kulikauskas MR, Caranasos TG, Jensen BC, Sabine A, Petrova TV, Caron KM. Adrenomedullin Induces Cardiac Lymphangiogenesis After Myocardial Infarction and Regulates Cardiac Edema Via Connexin 43. Circ Res 2019; 124:101-113. [PMID: 30582443 PMCID: PMC6318063 DOI: 10.1161/circresaha.118.313835] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RATIONALE Cardiac lymphangiogenesis contributes to the reparative process post-myocardial infarction, but the factors and mechanisms regulating it are not well understood. OBJECTIVE To determine if epicardial-secreted factor AM (adrenomedullin; Adm=gene) improves cardiac lymphangiogenesis post-myocardial infarction via lateralization of Cx43 (connexin 43) in cardiac lymphatic vasculature. METHODS AND RESULTS Firstly, we identified sex-dependent differences in cardiac lymphatic numbers in uninjured mice using light-sheet microscopy. Using a mouse model of Adm hi/hi ( Adm overexpression) and permanent left anterior descending ligation to induce myocardial infarction, we investigated cardiac lymphatic structure, growth, and function in injured murine hearts. Overexpression of Adm increased lymphangiogenesis and cardiac function post-myocardial infarction while suppressing cardiac edema and correlated with changes in Cx43 localization. Lymphatic function in response to AM treatment was attenuated in mice with a lymphatic-specific Cx43 deletion. In vitro experiments in cultured human lymphatic endothelial cells identified a novel mechanism to improve gap junction coupling by pharmaceutically targeting Cx43 with verapamil. Finally, we show that connexin protein expression in cardiac lymphatics is conserved between mouse and human. CONCLUSIONS AM is an endogenous, epicardial-derived factor that drives reparative cardiac lymphangiogenesis and function via Cx43, and this represents a new therapeutic pathway for improving myocardial edema after injury.
Collapse
Affiliation(s)
- Claire E. Trincot
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill
| | - Wenjing Xu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Molly R. Kulikauskas
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Thomas G. Caranasos
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill
| | - Brian C. Jensen
- Division of Cardiology, University of North Carolina at Chapel Hill
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill
- McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Amelie Sabine
- Department of Oncology, University of Lausanne and Lausanne University Hospital and Ludwig Institute for Cancer Research Lausanne, Chemin de Boveresses 155, CH-1066, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne and Lausanne University Hospital and Ludwig Institute for Cancer Research Lausanne, Chemin de Boveresses 155, CH-1066, Switzerland
- Division of Experimental Pathlogy, Lausanne University Hospital
| | - Kathleen M. Caron
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill , 111 Mason Farm Rd, MBRB 6312B, CB 7545, Chapel Hill, NC 27599
| |
Collapse
|
10
|
Targeting of chondrocyte plasticity via connexin43 modulation attenuates cellular senescence and fosters a pro-regenerative environment in osteoarthritis. Cell Death Dis 2018; 9:1166. [PMID: 30518918 PMCID: PMC6281585 DOI: 10.1038/s41419-018-1225-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/26/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA), a chronic disease characterized by articular cartilage degeneration, is a leading cause of disability and pain worldwide. In OA, chondrocytes in cartilage undergo phenotypic changes and senescence, restricting cartilage regeneration and favouring disease progression. Similar to other wound-healing disorders, chondrocytes from OA patients show a chronic increase in the gap junction channel protein connexin43 (Cx43), which regulates signal transduction through the exchange of elements or recruitment/release of signalling factors. Although immature or stem-like cells are present in cartilage from OA patients, their origin and role in disease progression are unknown. In this study, we found that Cx43 acts as a positive regulator of chondrocyte-mesenchymal transition. Overactive Cx43 largely maintains the immature phenotype by increasing nuclear translocation of Twist-1 and tissue remodelling and proinflammatory agents, such as MMPs and IL-1β, which in turn cause cellular senescence through upregulation of p53, p16INK4a and NF-κB, contributing to the senescence-associated secretory phenotype (SASP). Downregulation of either Cx43 by CRISPR/Cas9 or Cx43-mediated gap junctional intercellular communication (GJIC) by carbenoxolone treatment triggered rediferentiation of osteoarthritic chondrocytes into a more differentiated state, associated with decreased synthesis of MMPs and proinflammatory factors, and reduced senescence. We have identified causal Cx43-sensitive circuit in chondrocytes that regulates dedifferentiation, redifferentiation and senescence. We propose that chondrocytes undergo chondrocyte-mesenchymal transition where increased Cx43-mediated GJIC during OA facilitates Twist-1 nuclear translocation as a novel mechanism involved in OA progression. These findings support the use of Cx43 as an appropriate therapeutic target to halt OA progression and to promote cartilage regeneration.
Collapse
|
11
|
Radwański PB, Johnson CN, Györke S, Veeraraghavan R. Cardiac Arrhythmias as Manifestations of Nanopathies: An Emerging View. Front Physiol 2018; 9:1228. [PMID: 30233404 PMCID: PMC6131669 DOI: 10.3389/fphys.2018.01228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022] Open
Abstract
A nanodomain is a collection of proteins localized within a specialized, nanoscale structural environment, which can serve as the functional unit of macroscopic physiologic processes. We are beginning to recognize the key roles of cardiomyocyte nanodomains in essential processes of cardiac physiology such as electrical impulse propagation and excitation–contraction coupling (ECC). There is growing appreciation of nanodomain dysfunction, i.e., nanopathy, as a mechanistic driver of life-threatening arrhythmias in a variety of pathologies. Here, we offer an overview of current research on the role of nanodomains in cardiac physiology with particular emphasis on: (1) sodium channel-rich nanodomains within the intercalated disk that participate in cell-to-cell electrical coupling and (2) dyadic nanodomains located along transverse tubules that participate in ECC. The beat to beat function of cardiomyocytes involves three phases: the action potential, the calcium transient, and mechanical contraction/relaxation. In all these phases, cell-wide function results from the aggregation of the stochastic function of individual proteins. While it has long been known that proteins that exist in close proximity influence each other’s function, it is increasingly appreciated that there exist nanoscale structures that act as functional units of cardiac biophysical phenomena. Termed nanodomains, these structures are collections of proteins, localized within specialized nanoscale structural environments. The nano-environments enable the generation of localized electrical and/or chemical gradients, thereby conferring unique functional properties to these units. Thus, the function of a nanodomain is determined by its protein constituents as well as their local structural environment, adding an additional layer of complexity to cardiac biology and biophysics. However, with the emergence of experimental techniques that allow direct investigation of structure and function at the nanoscale, our understanding of cardiac physiology and pathophysiology at these scales is rapidly advancing. Here, we will discuss the structure and functions of multiple cardiomyocyte nanodomains, and novel strategies that target them for the treatment of cardiac arrhythmias.
Collapse
Affiliation(s)
- Przemysław B Radwański
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States.,Division of Pharmacy Practice and Science, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Christopher N Johnson
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Vanderbilt Center for Arrhythmia Research and Therapeutics, Nashville, TN, United States
| | - Sándor Györke
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Rengasayee Veeraraghavan
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States.,Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
12
|
Regulation of connexin 43 expression in human gingival fibroblasts. Exp Cell Res 2018; 371:238-249. [PMID: 30118696 DOI: 10.1016/j.yexcr.2018.08.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/31/2018] [Accepted: 08/13/2018] [Indexed: 12/22/2022]
Abstract
AIMS Abundance of connexin 43 (Cx43), a transmembrane protein that forms hemichannels (HCs) and gap junctions (GJs), is dynamically regulated in human gingival fibroblasts (GFBLs) during wound healing. This may be important for fast and scarless gingival wound healing as Cx43 is involved in key cell functions important during this process. Our aim was to uncover the factors that regulate Cx43 expression and abundance in GFBLs. We hypothesized that cytokines and growth factors released during wound healing coordinately regulate Cx43 abundance in GFBLs. RESULTS TGF-β1, -β2, -β3, PGE2 and IL-1β significantly upregulated, while TNF-α and IFN-γ downregulated Cx43 in cultured GFBLs. TGF-β1, -β2, -β3, IL-1β and IFN-γ modulated Cx43 abundance at both mRNA and protein levels, while TNF-α and PGE2 regulated only Cx43 protein abundance, suggesting involvement of distinct transcriptional/post-transcriptional and translational/post-translational mechanisms, respectively. TGF-β1-induced upregulation of Cx43 was mediated by TGFβRI (ALK5) and SMAD2/3 signaling, and this was potently suppressed by PGE2, IL-1β, TNF-α and IFN-γ that inhibited SMAD2/3 phosphorylation. CONCLUSION Regulation of Cx43 abundance in GFBLs involves transcriptional/post-transcriptional and translational/post-translational mechanisms that are distinctly modulated by an interplay between TGF-β isoforms and PGE2, IL-1β, TNF-α and IFN-γ.
Collapse
|
13
|
Montgomery J, Ghatnekar GS, Grek CL, Moyer KE, Gourdie RG. Connexin 43-Based Therapeutics for Dermal Wound Healing. Int J Mol Sci 2018; 19:ijms19061778. [PMID: 29914066 PMCID: PMC6032231 DOI: 10.3390/ijms19061778] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/08/2018] [Accepted: 06/12/2018] [Indexed: 12/22/2022] Open
Abstract
The most ubiquitous gap junction protein within the body, connexin 43 (Cx43), is a target of interest for modulating the dermal wound healing response. Observational studies found associations between Cx43 at the wound edge and poor healing response, and subsequent studies utilizing local knockdown of Cx43 found improvements in wound closure rate and final scar appearance. Further preclinical work conducted using Cx43-based peptide therapeutics, including alpha connexin carboxyl terminus 1 (αCT1), a peptide mimetic of the Cx43 carboxyl terminus, reported similar improvements in wound healing and scar formation. Clinical trials and further study into the mode of action have since been conducted on αCT1, and Phase III testing for treatment of diabetic foot ulcers is currently underway. Therapeutics targeting connexin activity show promise in beneficially modulating the human body’s natural healing response for improved patient outcomes across a variety of injuries.
Collapse
Affiliation(s)
- Jade Montgomery
- Virginia Tech Carilion Research Institute, Roanoke, VA 24016, USA.
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA 24061, USA.
| | | | | | - Kurtis E Moyer
- Department of Surgery, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA.
- Department of Surgery, Carilion Clinic, Roanoke, VA 24016, USA.
| | - Robert G Gourdie
- Virginia Tech Carilion Research Institute, Roanoke, VA 24016, USA.
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA 24061, USA.
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA.
| |
Collapse
|
14
|
Chanson M, Watanabe M, O'Shaughnessy EM, Zoso A, Martin PE. Connexin Communication Compartments and Wound Repair in Epithelial Tissue. Int J Mol Sci 2018; 19:ijms19051354. [PMID: 29751558 PMCID: PMC5983803 DOI: 10.3390/ijms19051354] [Citation(s) in RCA: 21] [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: 03/27/2018] [Revised: 04/26/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022] Open
Abstract
Epithelial tissues line the lumen of tracts and ducts connecting to the external environment. They are critical in forming an interface between the internal and external environment and, following assault from environmental factors and pathogens, they must rapidly repair to maintain cellular homeostasis. These tissue networks, that range from a single cell layer, such as in airway epithelium, to highly stratified and differentiated epithelial surfaces, such as the epidermis, are held together by a junctional nexus of proteins including adherens, tight and gap junctions, often forming unique and localised communication compartments activated for localised tissue repair. This review focuses on the dynamic changes that occur in connexins, the constituent proteins of the intercellular gap junction channel, during wound-healing processes and in localised inflammation, with an emphasis on the lung and skin. Current developments in targeting connexins as corrective therapies to improve wound closure and resolve localised inflammation are also discussed. Finally, we consider the emergence of the zebrafish as a concerted whole-animal model to study, visualise and track the events of wound repair and regeneration in real-time living model systems.
Collapse
Affiliation(s)
- Marc Chanson
- Department of Pediatrics and Cell Physiology & Metabolism, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland.
| | - Masakatsu Watanabe
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan.
| | - Erin M O'Shaughnessy
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| | - Alice Zoso
- Department of Pediatrics and Cell Physiology & Metabolism, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland.
| | - Patricia E Martin
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| |
Collapse
|
15
|
Connexin 43 regulates the expression of wound healing-related genes in human gingival and skin fibroblasts. Exp Cell Res 2018; 367:150-161. [PMID: 29596891 DOI: 10.1016/j.yexcr.2018.03.031] [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: 01/09/2018] [Revised: 03/23/2018] [Accepted: 03/24/2018] [Indexed: 12/21/2022]
Abstract
Fibroblasts are the most abundant connective tissue cells and play an important role in wound healing. It is possible that faster and scarless wound healing in oral mucosal gingiva relative to skin may relate to the distinct phenotype of the fibroblasts residing in these tissues. Connexin 43 (Cx43) is the most ubiquitous Cx in skin (SFBLs) and gingival fibroblasts (GFBLs), and assembles into hemichannels (HCs) and gap junctions (GJs) on the cell membrane. We hypothesized that SFBLs and GFBLs display distinct expression or function of Cx43, and that this may partly underlie the different wound healing outcomes in skin and gingiva. Here we show that Cx43 distinctly formed Cx43 GJs and HCs in human skin and gingiva in vivo. However, in SFBLs, in contrast to GFBLs, only a small proportion of total Cx43 assembled into HC plaques. Using an in vivo-like 3D culture model, we further show that the GJ, HC, and channel-independent functions of Cx43 distinctly regulated wound healing-related gene expression in GFBLs and SFBLs. Therefore, the distinct wound healing outcomes in skin and gingiva may partly relate to the inherently different assembly and function of Cx43 in the resident fibroblasts.
Collapse
|
16
|
Lindsey ML, Bolli R, Canty JM, Du XJ, Frangogiannis NG, Frantz S, Gourdie RG, Holmes JW, Jones SP, Kloner RA, Lefer DJ, Liao R, Murphy E, Ping P, Przyklenk K, Recchia FA, Schwartz Longacre L, Ripplinger CM, Van Eyk JE, Heusch G. Guidelines for experimental models of myocardial ischemia and infarction. Am J Physiol Heart Circ Physiol 2018; 314:H812-H838. [PMID: 29351451 PMCID: PMC5966768 DOI: 10.1152/ajpheart.00335.2017] [Citation(s) in RCA: 369] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial infarction is a prevalent major cardiovascular event that arises from myocardial ischemia with or without reperfusion, and basic and translational research is needed to better understand its underlying mechanisms and consequences for cardiac structure and function. Ischemia underlies a broad range of clinical scenarios ranging from angina to hibernation to permanent occlusion, and while reperfusion is mandatory for salvage from ischemic injury, reperfusion also inflicts injury on its own. In this consensus statement, we present recommendations for animal models of myocardial ischemia and infarction. With increasing awareness of the need for rigor and reproducibility in designing and performing scientific research to ensure validation of results, the goal of this review is to provide best practice information regarding myocardial ischemia-reperfusion and infarction models. Listen to this article’s corresponding podcast at ajpheart.podbean.com/e/guidelines-for-experimental-models-of-myocardial-ischemia-and-infarction/.
Collapse
Affiliation(s)
- Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Research Service, G. V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Roberto Bolli
- Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
| | - John M Canty
- Division of Cardiovascular Medicine, Departments of Biomedical Engineering and Physiology and Biophysics, The Veterans Affairs Western New York Health Care System and Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, New York
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital , Würzburg , Germany
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia Health System , Charlottesville, Virginia
| | - Steven P Jones
- Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Robert A Kloner
- HMRI Cardiovascular Research Institute, Huntington Medical Research Institutes , Pasadena, California.,Division of Cardiovascular Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana
| | - Ronglih Liao
- Harvard Medical School , Boston, Massachusetts.,Division of Genetics and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital , Boston, Massachusetts
| | - Elizabeth Murphy
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Peipei Ping
- National Institutes of Health BD2KBig Data to Knowledge (BD2K) Center of Excellence and Department of Physiology, Medicine and Bioinformatics, University of California , Los Angeles, California
| | - Karin Przyklenk
- Cardiovascular Research Institute and Departments of Physiology and Emergency Medicine, Wayne State University School of Medicine , Detroit, Michigan
| | - Fabio A Recchia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Fondazione G. Monasterio, Pisa , Italy.,Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University , Philadelphia, Pennsylvania
| | - Lisa Schwartz Longacre
- Heart Failure and Arrhythmias Branch, Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Crystal M Ripplinger
- Department of Pharmacology, School of Medicine, University of California , Davis, California
| | - Jennifer E Van Eyk
- The Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center , Los Angeles, California
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School , Essen , Germany
| |
Collapse
|
17
|
Liu S, Yeo DC, Wiraja C, Tey HL, Mrksich M, Xu C. Peptide delivery with poly(ethylene glycol) diacrylate microneedles through swelling effect. Bioeng Transl Med 2017; 2:258-267. [PMID: 29313035 PMCID: PMC5689501 DOI: 10.1002/btm2.10070] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022] Open
Abstract
Transdermal delivery of therapeutic biomolecules (including peptides) can avoid enzymatic digestion that occurs in the oral route. (Polyethylene glycol) diacrylate (PEGDA)-based microneedles, with good biocompatibility, are easily fabricated through photo-polymerization with a precisely controlled structure. It has successfully been used for the transdermal delivery of small molecule drugs such as 5-fluorouracil. However, the delivery of peptide-based therapeutics using this platform is seldom reported. This is because of the potential damage to the peptide during the photo-polymerization process of PEGDA. Herein, we introduce a method to load PEGDA microneedles with peptides without compromising peptide potency. Using gap junction inhibitor (Gap 26) as an example, the peptide was loaded into PEGDA microneedles through the swelling effect of PEGDA in the aqueous solution. The peptide-loaded microneedles were applied to a keloid scar model and exhibited inhibition expression of collagen I, a predominant marker of keloid scar, demonstrating its potential therapeutic effects.
Collapse
Affiliation(s)
- Shiying Liu
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive, 637457, Singapore
| | - David C Yeo
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive, 637457, Singapore
| | - Christian Wiraja
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive, 637457, Singapore
| | - Hong Liang Tey
- National Skin Centre 1 Mandalay Road, 308205, Singapore.,Lee Kong Chian School of Medicine Nanyang Technological University 50 Nanyang Avenue, 639798, Singapore
| | - Milan Mrksich
- Dept. of Chemistry Northwestern University 2145 Sheridan Road, Evanston, 60208 IL.,NTU-Northwestern Institute for Nanomedicine Nanyang Technological University 50 Nanyang Avenue, 639798, Singapore
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive, 637457, Singapore.,NTU-Northwestern Institute for Nanomedicine Nanyang Technological University 50 Nanyang Avenue, 639798, Singapore
| |
Collapse
|
18
|
Zhang XF, Cui X. Connexin 43: Key roles in the skin. Biomed Rep 2017; 6:605-611. [PMID: 28584630 DOI: 10.3892/br.2017.903] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/17/2017] [Indexed: 12/26/2022] Open
Abstract
Gap junctions are tightly packed intercellular channels that serve a common purpose of allowing the intercellular exchange of small metabolites, second messengers and electrical signals. Connexins (Cxs) are gap junction proteins. Currently, 20 and 21 members of Cxs have been characterized in mice and humans, respectively. Connexin 43 (Cx43) is the most ubiquitously expressed type of Cx in the skin. It is produced by various different types of skin cell, such as keratinocytes, fibroblasts, endothelial and basal cells, melanocytes and dermal papilla cells. At present, more evidence indicates that Cx43 has an important role in skin repair and skin tumor development, as well as in skin cell invasion and metastasis. In this review, current knowledge regarding the regulation and function of Cx43 is summarized and the therapeutic potential of regulating Cx43 activity is discussed.
Collapse
Affiliation(s)
- Xiao-Fei Zhang
- Department of Biological Sciences and Biotechnology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei 430070, P.R. China
| | - Xiaofeng Cui
- Department of Biological Sciences and Biotechnology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
19
|
Targeting the tight junction protein, zonula occludens-1, with the connexin43 mimetic peptide, αCT1, reduces VEGF-dependent RPE pathophysiology. J Mol Med (Berl) 2017; 95:535-552. [PMID: 28132078 DOI: 10.1007/s00109-017-1506-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 12/08/2016] [Accepted: 01/04/2017] [Indexed: 01/17/2023]
Abstract
A critical target tissue in age-related macular degeneration (AMD) is the retinal pigment epithelium (RPE), which forms the outer blood-retina barrier (BRB). RPE-barrier dysfunction might result from attenuation/disruption of intercellular tight junctions. Zonula occludens-1 (ZO-1) is a major structural protein of intercellular junctions. A connexin43-based peptide mimetic, αCT1, was developed to competitively block interactions at the PDZ2 domain of ZO-1, thereby inhibiting ligands that selectively bind to this domain. We hypothesized that targeting ZO-1 signaling using αCT1 would maintain BRB integrity and reduce RPE pathophysiology by stabilizing gap- and/or tight-junctions. RPE-cell barrier dysfunction was generated in mice using laser photocoagulation triggering choroidal neovascularization (CNV) or bright light exposure leading to morphological damage. αCT1 was delivered via eye drops. αCT1 treatment reduced CNV development and fluid leakage as determined by optical coherence tomography, and damage was correlated with disruption in cellular integrity of surrounding RPE cells. Light damage significantly disrupted RPE cell morphology as determined by ZO-1 and occludin staining and tiling pattern analysis, which was prevented by αCT1 pre-treatment. In vitro experiments using RPE and MDCK monolayers indicated that αCT1 stabilizes tight junctions, independent of its effects on Cx43. Taken together, stabilization of intercellular junctions by αCT1 was effective in ameliorating RPE dysfunction in models of AMD-like pathology. KEY MESSAGE The connexin43 mimetic αCT1 accumulates in the mouse retinal pigment epithelium following topical delivery via eye drops. αCT1 eye drops prevented RPE-cell barrier dysfunction in two mouse models. αCT1 stabilizes intercellular tight junctions. Stabilization of cellular junctions via αCT1 may serve as a novel therapeutic approach for both wet and dry age-related macular degeneration.
Collapse
|
20
|
Wong P, Tan T, Chan C, Laxton V, Chan YWF, Liu T, Wong WT, Tse G. The Role of Connexins in Wound Healing and Repair: Novel Therapeutic Approaches. Front Physiol 2016; 7:596. [PMID: 27999549 PMCID: PMC5138227 DOI: 10.3389/fphys.2016.00596] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/16/2016] [Indexed: 12/26/2022] Open
Abstract
Gap junctions are intercellular proteins responsible for mediating both electrical and biochemical coupling through the exchange of ions, second messengers and small metabolites. They consist of two connexons, with (one) connexon supplied by each cell. A connexon is a hexamer of connexins and currently more than 20 connexin isoforms have been described in the literature thus far. Connexins have a short half-life, and therefore gap junction remodeling constantly occurs with a high turnover rate. Post-translational modification, such as phosphorylation, can modify their channel activities. In this article, the roles of connexins in wound healing and repair are reviewed. Novel strategies for modulating the function or expression of connexins, such as the use of antisense technology, synthetic mimetic peptides and bioactive materials for the treatment of skin wounds, diabetic and pressure ulcers as well as cornea wounds, are considered.
Collapse
Affiliation(s)
- Pui Wong
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong KongHong Kong, Hong Kong
| | - Teresa Tan
- Department of Surgery, Faculty of Medicine, Chinese University of Hong KongHong Kong, Hong Kong
| | - Catherine Chan
- Department of Surgery, Faculty of Medicine, Chinese University of Hong KongHong Kong, Hong Kong
| | - Victoria Laxton
- Intensive Care Department, Royal Brompton and Harefield NHS Foundation TrustLondon, UK
| | - Yin Wah Fiona Chan
- Department of Psychology, School of Biological Sciences, University of CambridgeCambridge, UK
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjin, China
| | - Wing Tak Wong
- School of Life Sciences, Chinese University of Hong KongHong Kong, Hong Kong
| | - Gary Tse
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong KongHong Kong, Hong Kong
- Faculty of Medicine, Li Ka Shing Institute of Health Sciences, Chinese University of Hong KongHong Kong, Hong Kong
| |
Collapse
|
21
|
Willebrords J, Crespo Yanguas S, Maes M, Decrock E, Wang N, Leybaert L, Kwak BR, Green CR, Cogliati B, Vinken M. Connexins and their channels in inflammation. Crit Rev Biochem Mol Biol 2016; 51:413-439. [PMID: 27387655 PMCID: PMC5584657 DOI: 10.1080/10409238.2016.1204980] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Inflammation may be caused by a variety of factors and is a hallmark of a plethora of acute and chronic diseases. The purpose of inflammation is to eliminate the initial cell injury trigger, to clear out dead cells from damaged tissue and to initiate tissue regeneration. Despite the wealth of knowledge regarding the involvement of cellular communication in inflammation, studies on the role of connexin-based channels in this process have only begun to emerge in the last few years. In this paper, a state-of-the-art overview of the effects of inflammation on connexin signaling is provided. Vice versa, the involvement of connexins and their channels in inflammation will be discussed by relying on studies that use a variety of experimental tools, such as genetically modified animals, small interfering RNA and connexin-based channel blockers. A better understanding of the importance of connexin signaling in inflammation may open up towards clinical perspectives.
Collapse
Affiliation(s)
- Joost Willebrords
- Department of In Vitro Toxicology and
Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels,
Belgium; Joost Willebrords: + Tel: 32 2 477 45 87, Michaël Maes: Tel: +32 2
477 45 87, Sara Crespo Yanguas: Tel: +32 2 477 45 87
| | - Sara Crespo Yanguas
- Department of In Vitro Toxicology and
Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels,
Belgium; Joost Willebrords: + Tel: 32 2 477 45 87, Michaël Maes: Tel: +32 2
477 45 87, Sara Crespo Yanguas: Tel: +32 2 477 45 87
| | - Michaël Maes
- Department of In Vitro Toxicology and
Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels,
Belgium; Joost Willebrords: + Tel: 32 2 477 45 87, Michaël Maes: Tel: +32 2
477 45 87, Sara Crespo Yanguas: Tel: +32 2 477 45 87
| | - Elke Decrock
- Department of Basic Medical Sciences, Physiology Group, Ghent
University, De Pintelaan 185, 9000 Ghent, Belgium; Elke Decrock: Tel: +32 9 332 39
73, Nan Wang: Tel: +32 9 332 39 38, Luc Leybaert: Tel: +32 9 332 33 66
| | - Nan Wang
- Department of Basic Medical Sciences, Physiology Group, Ghent
University, De Pintelaan 185, 9000 Ghent, Belgium; Elke Decrock: Tel: +32 9 332 39
73, Nan Wang: Tel: +32 9 332 39 38, Luc Leybaert: Tel: +32 9 332 33 66
| | - Luc Leybaert
- Department of Basic Medical Sciences, Physiology Group, Ghent
University, De Pintelaan 185, 9000 Ghent, Belgium; Elke Decrock: Tel: +32 9 332 39
73, Nan Wang: Tel: +32 9 332 39 38, Luc Leybaert: Tel: +32 9 332 33 66
| | - Brenda R. Kwak
- Department of Pathology and Immunology and Division of Cardiology,
University of Geneva, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland; Brenda R.
Kwak: Tel: +41 22 379 57 37
| | - Colin R. Green
- Department of Ophthalmology and New Zealand National Eye Centre,
University of Auckland, New Zealand; Colin R. Green: Tel: +64 9 923 61 35
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal
Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva 87,
05508-270 São Paulo, Brazil; Bruno Cogliati: Tel: +55 11 30 91 12 00
| | - Mathieu Vinken
- Department of In Vitro Toxicology and
Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels,
Belgium; Joost Willebrords: + Tel: 32 2 477 45 87, Michaël Maes: Tel: +32 2
477 45 87, Sara Crespo Yanguas: Tel: +32 2 477 45 87
| |
Collapse
|
22
|
Cell communication across gap junctions: a historical perspective and current developments. Biochem Soc Trans 2016; 43:450-9. [PMID: 26009190 DOI: 10.1042/bst20150056] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Collaborative communication lies at the centre of multicellular life. Gap junctions (GJs) are surface membrane structures that allow direct communication between cells. They were discovered in the 1960s following the convergence of the detection of low-resistance electrical interactions between cells and anatomical studies of intercellular contact points. GJs purified from liver plasma membranes contained a 27 kDa protein constituent; it was later named Cx32 (connexin 32) after its full sequence was determined by recombinant technology. Identification of Cx43 in heart and later by a further GJ protein, Cx26 followed. Cxs have a tetraspan organization in the membrane and oligomerize during intracellular transit to the plasma membrane; these were shown to be hexameric hemichannels (connexons) that could interact end-to-end to generate GJs at areas of cell-to-cell contact. The structure of the GJ was confirmed and refined by a combination of biochemical and structural approaches. Progress continues towards obtaining higher atomic 3D resolution of the GJ channel. Today, there are 20 and 21 highly conserved members of the Cx family in the human and mouse genomes respectively. Model organisms such as Xenopus oocytes and zebra fish are increasingly used to relate structure to function. Proteins that form similar large pore membrane channels in cells called pannexins have also been identified in chordates. Innexins form GJs in prechordates; these two other proteins, although functionally similar, are very different in amino acid sequence to the Cxs. A time line tracing the historical progression of wide ranging research in GJ biology over 60 years is mapped out. The molecular basis of channel dysfunctions in disease is becoming evident and progress towards addressing Cx channel-dependent pathologies, especially in ischaemia and tissue repair, continues.
Collapse
|
23
|
Ongstad E, Kohl P. Fibroblast-myocyte coupling in the heart: Potential relevance for therapeutic interventions. J Mol Cell Cardiol 2016; 91:238-46. [PMID: 26774702 DOI: 10.1016/j.yjmcc.2016.01.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/09/2016] [Accepted: 01/11/2016] [Indexed: 01/03/2023]
Abstract
Cardiac myocyte-fibroblast electrotonic coupling is a well-established fact in vitro. Indirect evidence of its presence in vivo exists, but few functional studies have been published. This review describes the current knowledge of fibroblast-myocyte electrical signaling in the heart. Further research is needed to understand the frequency and extent of heterocellular interactions in vivo in order to gain a better understanding of their relevance in healthy and diseased myocardium. It is hoped that associated insight into myocyte-fibroblast coupling in the heart may lead to the discovery of novel therapeutic targets and the development of agents for improving outcomes of myocardial scarring and fibrosis.
Collapse
Affiliation(s)
- Emily Ongstad
- Clemson University, Department of Bioengineering, Clemson, SC, USA; Virginia Tech Carilion Research Institute, Roanoke, VA, USA.
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg - Bad Krozingen, Faculty of Medicine, University Freiburg, Germany; Cardiac Biophysics and Systems Biology, National Heart and Lung Institute, Imperial College London, UK
| |
Collapse
|
24
|
Glass BJ, Hu RG, Phillips ARJ, Becker DL. The action of mimetic peptides on connexins protects fibroblasts from the negative effects of ischemia reperfusion. Biol Open 2015; 4:1473-80. [PMID: 26471768 PMCID: PMC4728352 DOI: 10.1242/bio.013573] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Connexins have been proposed as a target for therapeutic treatment of a variety of conditions. The main approaches have been by antisense or small peptides specific against connexins. Some of these peptides enhance communication while others interfere with connexin binding partners or bind to the intracellular and extracellular loops of connexins. Here, we explored the mechanism of action of a connexin mimetic peptide by evaluating its effect on gap junction channels, connexin protein levels and hemichannel activity in fibroblast cells under normal conditions and following ischemia reperfusion injury which elevates Cx43 levels, increases hemichannel activity and causes cell death. Our results showed that the effects of the mimetic peptide were concentration-dependent. High concentrations (100-300 μM) significantly reduced Cx43 protein levels and GJIC within 2 h, while these effects did not appear until 6 h when using lower concentrations (10-30 μM). Cell death can be reduced when hemichannel opening and GJIC were minimised. Summary: Connexin mimetic peptides can reduce the levels of connexin proteins in cells and can prevent the spread of cell death that occurs following ischemia reperfusion injury, which has therapeutic potential.
Collapse
Affiliation(s)
- Beverley J Glass
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rebecca G Hu
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232
| | - Anthony R J Phillips
- CoDa Therapeutics, Inc., 10 College Hill Road, Herne Bay, Auckland 1011, New Zealand School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232
| |
Collapse
|
25
|
Sutcliffe J, Chin K, Thrasivoulou C, Serena T, O'Neil S, Hu R, White A, Madden L, Richards T, Phillips A, Becker D. Abnormal connexin expression in human chronic wounds. Br J Dermatol 2015; 173:1205-15. [DOI: 10.1111/bjd.14064] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2015] [Indexed: 12/21/2022]
Affiliation(s)
- J.E.S. Sutcliffe
- Department of Cell and Developmental Biology; University College London; Gower Street London WC1E 6BT U.K
| | - K.Y. Chin
- Department of Cell and Developmental Biology; University College London; Gower Street London WC1E 6BT U.K
| | - C. Thrasivoulou
- Department of Cell and Developmental Biology; University College London; Gower Street London WC1E 6BT U.K
| | - T.E. Serena
- Newbridge Medical Research Corp.; Warren PA 16365 U.S.A
| | - S. O'Neil
- CoDa Therapeutics; 10 College Hill Auckland 1011 New Zealand
| | - R. Hu
- Lee Kong Chian School of Medicine; Nanyang Technological University; 11 Mandalay Road Singapore 308232 Singapore
| | - A.M. White
- CoDa Therapeutics; 10 College Hill Auckland 1011 New Zealand
| | - L. Madden
- Lee Kong Chian School of Medicine; Nanyang Technological University; 11 Mandalay Road Singapore 308232 Singapore
| | - T. Richards
- Department of Cell and Developmental Biology; University College London; Gower Street London WC1E 6BT U.K
| | - A.R.J. Phillips
- CoDa Therapeutics; 10 College Hill Auckland 1011 New Zealand
| | - D.L. Becker
- Lee Kong Chian School of Medicine; Nanyang Technological University; 11 Mandalay Road Singapore 308232 Singapore
- Institute of Medical Biology; A*STAR; 138648 Singapore
| |
Collapse
|
26
|
Abstract
INTRODUCTION About 2% of the Western world population suffer from chronic wounds, resulting from underlying disorders (e.g., diabetes, excessive pressure, vascular insufficiencies and vasculitis), with a significant adverse effect on Quality of Life. Despite high incidence and economic burden, management of chronic wounds is still far from effective and novel therapies are in urgent need. Wound healing is a dynamic process of transient expression, function and clearance of mediators, enzymes and cell types. Failure to initiate, terminate or regulate leads to pathologic wound healing. AREAS COVERED The present review discusses patents of the seven most promising classes of biological agents, mostly published in 2009 - 2014 (CYP11B1 inhibitors, peptide growth factors, prolyl-4-hydroxylase and matrix metalloproteinase inhibitors, bone marrow-derived mesenchymal stem cells, elastase and connexin43 inhibitors). Relevant information from peer-reviewed journals is also presented. EXPERT OPINION The aforementioned biological agents have different mechanisms of action, and considering the multifactorial pathogenesis of chronic wounds, they hold promise in treating chronic wounds. However, as administration of a certain biological agent may be beneficial in an early phase, it may slow down wound healing in a later phase. Basic and clinical research on chronic wound healing should therefore investigate the efficacy of these agents, alone and in concert, during the consecutive phases of wound healing.
Collapse
Affiliation(s)
- Chris J van Koppen
- ElexoPharm GmbH , Im Stadtwald, Building A1.2, 66123 Saarbrücken , Germany +49 681 30268320 ; +49 681 9102894 ;
| | | |
Collapse
|
27
|
Tarzemany R, Jiang G, Larjava H, Häkkinen L. Expression and function of connexin 43 in human gingival wound healing and fibroblasts. PLoS One 2015; 10:e0115524. [PMID: 25584940 PMCID: PMC4293150 DOI: 10.1371/journal.pone.0115524] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/25/2014] [Indexed: 11/18/2022] Open
Abstract
Connexins (C×s) are a family of transmembrane proteins that form hemichannels and gap junctions (GJs) on the cell membranes, and transfer small signaling molecules between the cytoplasm and extracellular space and between connecting cells, respectively. Among C×s, suppressing C×43 expression or function promotes skin wound closure and granulation tissue formation, and may alleviate scarring, but the mechanisms are not well understood. Oral mucosal gingiva is characterized by faster wound closure and scarless wound healing outcome as compared to skin wounds. Therefore, we hypothesized that C×43 function is down regulated during human gingival wound healing, which in fibroblasts promotes expression of genes conducive for fast and scarless wound healing. Cultured gingival fibroblasts expressed C×43 as their major connexin. Immunostaining of unwounded human gingiva showed that C×43 was abundantly present in the epithelium, and in connective tissue formed large C×43 plaques in fibroblasts. At the early stages of wound healing, C×43 was strongly down regulated in wound epithelial cells and fibroblasts, returning to the level of normal tissue by day 60 post-wounding. Blocking of C×43 function by C×43 mimetic peptide Gap27 suppressed GJ-mediated dye transfer, promoted migration, and caused significant changes in the expression of wound healing-associated genes in gingival fibroblasts. In particular, out of 54 genes analyzed, several MMPs and TGF-β1, involved in regulation of inflammation and extracellular matrix (ECM) turnover, and VEGF-A, involved in angiogenesis, were significantly upregulated while pro-fibrotic ECM molecules, including Collagen type I, and cell contractility-related molecules were significantly down regulated. These responses involved MAPK, GSK3α/β and TGF-β signaling pathways, and AP1 and SP1 transcription factors. Thus, suppressed function of C×43 in fibroblasts promotes their migration, and regulates expression of wound healing-associated genes via AP1, SP1, MAPK, GSK3α/β and TGF-β signaling pathways, and may promote fast and scarless wound healing in human gingiva.
Collapse
Affiliation(s)
- Rana Tarzemany
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Guoqiao Jiang
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Hannu Larjava
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Lari Häkkinen
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
28
|
Kelly JJ, Simek J, Laird DW. Mechanisms linking connexin mutations to human diseases. Cell Tissue Res 2014; 360:701-21. [DOI: 10.1007/s00441-014-2024-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 09/26/2014] [Indexed: 11/30/2022]
|
29
|
Li YH, Zhang CL, Zhang XY, Zhou HX, Meng LL. Effects of mild induced hypothermia on hippocampal connexin 43 and glutamate transporter 1 expression following traumatic brain injury in rats. Mol Med Rep 2014; 11:1991-6. [PMID: 25394735 DOI: 10.3892/mmr.2014.2928] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 06/17/2014] [Indexed: 11/06/2022] Open
Abstract
Traumatic brain injury (TBI) is a common cause of worldwide disability and mortality. Currently, the incidence and prevalence of TBI is markedly increasing and an effective therapy is lacking. Therapeutic hypothermia (32‑35˚C) has been reported to reduce intracranial pressure and induce putative neuroprotective effects. However, the underlying molecular mechanisms remain to be elucidated. The aim of the present study was to investigate the effects of mild induced hypothermia (MIH) on the expression of connexin 43 (Cx43) and glutamate transporter 1 (GLT‑1) in the hippocampus following TBI in rats. A rat model of TBI was created using a modified weight‑drop device, followed by 4 h of hypothermia (33˚C) or normothermia (37˚C). A wet‑dry weight method was used to assess brain edema and spatial learning ability was evaluated using a Morris water maze. The levels of Cx43 and GLT‑1 were detected by immunohistochemical and western blot analysis, respectively. The results demonstrated that MIH treatment improved TBI‑induced brain edema and neurological function deficits. In addition, therapeutic MIH significantly downregulated Cx43 expression and upregulated the levels of GLT‑1 in the hippocampus post‑TBI. These findings suggested that treatment with MIH may provide a novel neuroprotective therapeutic strategy for TBI through reversing the increase in Cx43 protein and the decrease in GLT‑1.
Collapse
Affiliation(s)
- Yue-Hong Li
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Chun-Lai Zhang
- Department of Cardiology, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Xiao-Yan Zhang
- School of Basic Medical Sciences, Hebei United University, Tangshan, Hebei 063000, P.R. China
| | - Hong-Xia Zhou
- School of Basic Medical Sciences, Hebei United University, Tangshan, Hebei 063000, P.R. China
| | - Ling-Li Meng
- School of Basic Medical Sciences, Hebei United University, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
30
|
Strungs EG, Ongstad EL, O'Quinn MP, Palatinus JA, Jourdan LJ, Gourdie RG. Cryoinjury models of the adult and neonatal mouse heart for studies of scarring and regeneration. Methods Mol Biol 2013; 1037:343-53. [PMID: 24029946 DOI: 10.1007/978-1-62703-505-7_20] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
A major limitation in studies of the injured heart is animal-to-animal variability in wound size resulting from commonly used techniques such as left anterior descending coronary artery ligation. This variability can make standard errors sufficiently large that mean separation between treatment and control groups can be difficult without replicating numbers (n) of animals in groups by excessive amounts. Here, we describe the materials and protocol necessary for delivering a standardized non-transmural cryoinjury to the left ventricle of an adult mouse heart that may in part obviate the issue of injury variance between animals. As reported previously, this cryoinjury model generates a necrotic wound to the ventricle of consistent size and shape that resolves into a scar of uniform size, shape, and organization. The cryo-model also provides an extended injury border zone that exhibits classic markers of remodeling found in surviving cardiac tissue at the edge of a myocardial infarction, including connexin43 (Cx43) lateralization. In a further extension of the method, we describe how we have adapted the model to deliver a cryoinjury to the apex of the heart of neonatal mice-a modification that may be useful for studies of myocardial regeneration in mammals.
Collapse
Affiliation(s)
- Erik G Strungs
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | | | | |
Collapse
|