1
|
Spagnol G, Trease A, Zheng L, Sobota S, Schmidt M, Cheku S, Sorgen PL. Cx45 regulation by kinases and impact of expression in heart failure. J Mol Cell Cardiol 2025; 203:91-105. [PMID: 40280467 DOI: 10.1016/j.yjmcc.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Phosphorylation plays a crucial role in connexin regulation by modulating gap junction intercellular communication (GJIC), localization, stability, and interactions with signaling proteins. Few kinases are known to phosphorylate Cx45, and their target residues remain unknown. A phosphorylation screen identified several Cx45-targeting kinases activated in heart disease, among which c-Src was found by mass spectroscopy to phosphorylate residues Y324 and Y356. Unlike Cx43, c-Src phosphorylation of Cx45 did not impair GJIC, alter junctional localization, or affect interactions with cytoskeletal proteins β-tubulin, Drebrin, and ZO-1. In LA-25 cells where Cx43 is internalized after temperature sensitive activation of v-Src, expression of Cx45 unexpectedly maintained Cx43 at the plasma membrane. Phospho-specific antibodies helped identify that while Cx43 had a tyrosine phosphorylation pattern favoring turnover, the serine phosphorylation pattern was conducive for GJIC. Furthermore, in a rat model of heart failure, Cx45 was expressed in the ventricle and co-localized with Cx43, leading to altered dye coupling indicative of a shift toward Cx45-like channel permeability. Altogether, our data suggests that in heart failure, c-Src activation on its own would not have an adverse effect on Cx45 function and that aberrant Cx45 expression helps Cx43 transport to and maintain at the intercalated disc. Yet the dominant effect of Cx45 in heteromeric channels could ultimately make Cx45 a key driver of cardiac dysfunction. Finally, the observation that Cx45-mediated coupling remains functional even in the same pathological environment where Cx43-mediated communication is inhibited suggests that kinase regulation of connexins is isoform-specific and not universally predictable.
Collapse
Affiliation(s)
- Gaelle Spagnol
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Andrew Trease
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Li Zheng
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Stephen Sobota
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Marissa Schmidt
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sunayn Cheku
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Paul L Sorgen
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
2
|
Meinung CP, Boi L, Pandamooz S, Mazaud D, Ghézali G, Rouach N, Neumann ID. OXTR-mediated signaling in astrocytes contributes to anxiolysis. Mol Psychiatry 2025; 30:2620-2634. [PMID: 39702695 DOI: 10.1038/s41380-024-02870-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/13/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Astrocytes are an indispensable part of signal processing within the mammalian brain. Thus, the mode of action of a neuropeptide such as oxytocin (OXT) can only be fully understood considering this integral part of the CNS. Here, we show that OXT regulates astrocytic gene expression, intracellular signaling and specific proteins both in vitro and in vivo. This translates into rapid regulation of astroglial structural and functional properties including cytoskeletal plasticity, coverage of synapses and gap-junction coupling. At the molecular level, we identify the previously undescribed Sp1-Gem signaling cascade as the key driver for these cell type-specific OXT effects. Finally at the behavioral level, we found in vivo that OXT requires astrocytes to exert its well described anxiolytic properties within the hypothalamic paraventricular nucleus. Thus, our study points to OXT receptor-expressing astrocytes as a critical component of the brain OXT system.
Collapse
Affiliation(s)
- Carl-Philipp Meinung
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Laura Boi
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Sareh Pandamooz
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - David Mazaud
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Grégory Ghézali
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
3
|
Wen C, Yang R, Yi J, Cao Y, Song Y, An L, Wang Z, Gao H. Downregulation of EB1 impedes Cx43 localization and cardiac conduction after hypothermic ischemia-reperfusion in rats. PeerJ 2025; 13:e19276. [PMID: 40247841 PMCID: PMC12005192 DOI: 10.7717/peerj.19276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Background Hypothermic ischemia-reperfusion arrhythmia is a common complication after cardiopulmonary bypass heart surgery, which can lead to hemodynamic disorders and even sudden cardiac death and is still not effectively prevented. This study aims to investigate the role and mechanisms of EB1 in hypothermic ischemia-reperfusion arrhythmia. Methods 4-6 week old male Sprague-Dawley (SD) rats were randomly assigned to four groups with a control group receiving no treatment. In the treatment groups, the rats received an injection of a negative control adenovirus (AAV9-CON) or an adenoviral vector containing Mapre1 gene (AAV9-EB1) or an equal volume of saline via the tail vein. After 4 weeks, untreated rat hearts underwent continuous isolated heart perfusion for 5 min, while the treatment groups were subjected to Langendorff isolated heart ischemia-reperfusion. The multi-electrode array (MEA) technique was used to measure the conduction heterogeneity of rat heart, evaluating the protective effects of EB1 overexpression against reperfusion arrhythmias. Additionally, histological staining and western blotting were used to explore the potential pathways by which EB1 exerts its anti-arrhythmic effects, potentially through promoting the localization of connexin 43 (Cx43) to the intercalated discs (IDs). Furthermore, western blot analysis was conducted to assess microtubule stability and evaluate the possible mechanism by which EB1 facilitates the localization of Cx43 to the IDs. Results Following ischemia-reperfusion, EB1 expression was downregulated, accompanied by a reduction in Cx43. Overexpression of myocardial EB1 reduced the incidence of reperfusion arrhythmias and shortened their duration, which was associated with improved myocardial conduction. Male SD rats injected with AAV overexpressing EB1 had significantly higher levels of both total myocardial Cx43 and gap junction Cx43 after ischemia-reperfusion compared to the non-overexpression groups. Histological staining revealed lateralization of Cx43 in ischemia-reperfusion myocardium, which was corrected by EB1 overexpression. Additionally, EB1 overexpression increased the distribution of Cx43 at the IDs, overall reducing Cx43 remodeling. Moreover, EB1 overexpression can also alleviate microtubule damage caused by ischemia-reperfusion, which may be an important mechanism for the transport of Cx43 to the IDs. Conclusions EB1 downregulation following hypothermic ischemia-reperfusion was accompanied by a reduction in gap junction Cx43. EB1 overexpression improved cardiac conduction and reduced reperfusion arrhythmias by promoting Cx43 localization to IDs, facilitating gap junctions (GJs) formation. These findings contribute to the development of new therapeutic targets for reperfusion arrhythmias.
Collapse
Affiliation(s)
- Chunlei Wen
- Guizhou Medical University, Guiyang, Guizhou, China
- Department of Anesthesiology, Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou, China
| | | | - Jing Yi
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Cao
- Department of Anesthesiology, The Second People’s Hospital of Guiyang, Guiyang, Guizhou, China
| | - Yuting Song
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Li An
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zijun Wang
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Hong Gao
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
4
|
Boengler K, Mantuano B, Toledano S, Binah O, Schulz R. Overexpression of Cx43: Is It an Effective Approach for the Treatment of Cardiovascular Diseases? Biomolecules 2025; 15:370. [PMID: 40149906 PMCID: PMC11940156 DOI: 10.3390/biom15030370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
In the heart, Connexin 43 (Cx43) is involved in intercellular communication through gap junctions and exosomes. In addition, Cx43-formed hemichannels at the plasma membrane are important for ion homeostasis and cellular volume regulation. Through its localization within nuclei and mitochondria, Cx43 influences the function of the respective organelles. Several cardiovascular diseases such as heart failure, ischemia/reperfusion injury, hypertrophic cardiomyopathy and arrhythmias are characterized by Cx43 downregulation and a dysregulated Cx43 function. Accordingly, a putative therapeutic approach of these diseases would include the induction of Cx43 expression in the damaged heart, albeit such induction may have both beneficial and detrimental effects. In this review we discuss the consequences of increasing cardiac Cx43 expression, and discuss this manipulation as a strategy for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Kerstin Boengler
- Institute of Physiology, Justus-Liebig University, 35392 Giessen, Germany
| | - Beatrice Mantuano
- Department of Clinical and Biological Sciences, University of Torino, 10125 Torino, Italy
| | - Shira Toledano
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3190601, Israel
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3190601, Israel
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, 35392 Giessen, Germany
| |
Collapse
|
5
|
Frederico MJS, Sulis PM, Pereira LL, Rey D, Aragón M, Silva FRMB. Potential Effect of Cinnamaldehyde on Insulin Resistance Is Mediated by Glucose and Lipid Homeostasis. Nutrients 2025; 17:297. [PMID: 39861427 PMCID: PMC11767522 DOI: 10.3390/nu17020297] [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: 12/18/2024] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetes mellitus is a metabolic syndrome that has grown globally to become a significant public health challenge. Hypothesizing that the plasma membrane protein, transient receptor potential ankyrin-1, is a pivotal target in insulin resistance, we investigated the mechanism of action of cinnamaldehyde (CIN), an electrophilic TRPA1 agonist, in skeletal muscle, a primary insulin target. Specifically, we evaluated the effect of CIN on insulin resistance, hepatic glycogen accumulation and muscle and adipose tissue glucose uptake. Furthermore, the in vitro role of CIN in glucose uptake and intracellular signaling was determined in insulin-resistant rats whose calcium influx was analyzed. Moreover, the serum lipid profile was assessed following short-term CIN treatment in rats, and lipid tolerance was analyzed. The effects of CIN on insulin resistance were mediated by TRPA1, with downstream signaling involving the activation of PI3-K, MAPK, PKC, as well as extracellular calcium and calcium release from intracellular stores. Additionally, cytoskeleton integrity was required for the complete action of CIN on glucose uptake in muscle. CIN also ameliorated the serum lipid profile and improved triglyceride tolerance following acute vivo exposure.
Collapse
Affiliation(s)
- Marisa Jadna Silva Frederico
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Rua João Pio Duarte Silva, 241, Sala G 301, Florianópolis 88038-000, SC, Brazil; (M.J.S.F.); (P.M.S.); (L.L.P.); (D.R.)
- Laboratório de Bioquímica e Farmacologia, Departamento de Farmacologia e Fisiologia, Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Escola de Medicina, Universidade Federal do Ceará, Rua Coronel Nunes de Melo, Fortaleza 60430-275, CE, Brazil
| | - Paola Miranda Sulis
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Rua João Pio Duarte Silva, 241, Sala G 301, Florianópolis 88038-000, SC, Brazil; (M.J.S.F.); (P.M.S.); (L.L.P.); (D.R.)
| | - Landerson Lopes Pereira
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Rua João Pio Duarte Silva, 241, Sala G 301, Florianópolis 88038-000, SC, Brazil; (M.J.S.F.); (P.M.S.); (L.L.P.); (D.R.)
| | - Diana Rey
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Rua João Pio Duarte Silva, 241, Sala G 301, Florianópolis 88038-000, SC, Brazil; (M.J.S.F.); (P.M.S.); (L.L.P.); (D.R.)
- Departamento de Farmacia, Universidad Nacional de Colombia, Av. Carrera 30 # 45-03 Edif. 450, Bogotá 111321, Colombia;
| | - Marcela Aragón
- Departamento de Farmacia, Universidad Nacional de Colombia, Av. Carrera 30 # 45-03 Edif. 450, Bogotá 111321, Colombia;
| | - Fátima Regina Mena Barreto Silva
- Instituto de Bioeletricidade Celular (IBIOCEL): Ciência & Saúde, Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Rua João Pio Duarte Silva, 241, Sala G 301, Florianópolis 88038-000, SC, Brazil; (M.J.S.F.); (P.M.S.); (L.L.P.); (D.R.)
| |
Collapse
|
6
|
Marsh SR, Amin MR, Toldo S, Beard C, Dogan AB, Mezzaroma E, Andres E, Stout RF, Bannon MS, Payne LB, Abbate A, Sassi Y, Letteri RA, Gourdie RG. Orally Delivered Milk-Derived Nanovesicles Loaded with Connexin 43 Peptides for Targeted Cardiac Ischemia-Reperfusion Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.01.630994. [PMID: 40093162 PMCID: PMC11908194 DOI: 10.1101/2025.01.01.630994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Extracellular vesicles have emerged as promising nanocarriers for targeted drug delivery, but their therapeutic potential is limited by challenges related to administration route, loading, targeted delivery and production at scale. Here, we report an innovative approach for targeted delivery of therapeutic peptides to injured tissues using milk-derived small extracellular vesicles (mEVs) as an abundant, safe, orally administrable nanoplatform. We demonstrate that a sub-population of mEVs naturally contain Connexin 43 (Cx43) and its Carboxyl-Terminal (CT) polypeptides, which have been shown to play crucial roles in wound healing and tissue repair. Leveraging this intrinsic property, we developed an esterification method to efficiently and uniformly load mEVs with enhanced levels of an exogenous Cx43 CT peptide (αCT11 - RPRPDDLEI), as assessed by flow cytometry-based vesicle quantification and mass spectrometry. These engineered mEVs exhibited remarkable injury targeting capabilities, with > 30-fold increases in uptake by injured cells compared to non-wounded cells in vitro and preferential accumulation in wounded tissues in vivo. Notably, αCT11-loaded mEVs orally administered after myocardial infarction reduced infarct size by >60% and preserved heart function in a mouse model of ischemia-reperfusion injury. This study represents a significant advance in nanomedicine, demonstrating the utilization of naturally occurring milk-derived extracellular vesicles as an oral delivery system for therapeutic peptides, achieving unprecedented targeting efficiency and efficacy in the treatment of myocardial ischemia-reperfusion injury.
Collapse
|
7
|
Huang X, Bai X, Yi J, Hu T, An L, Gao H. The activation of P38MAPK Signaling Pathway Impedes the Delivery of the Cx43 to the Intercalated Discs During Cardiac Ischemia-Reperfusion Injury. J Cardiovasc Transl Res 2024; 17:1140-1154. [PMID: 38696081 DOI: 10.1007/s12265-024-10515-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/11/2024] [Indexed: 10/29/2024]
Abstract
Ischemic heart disease is caused by coronary artery occlusion. Despite the increasing number and success of interventions for restoring coronary artery perfusion, myocardial ischemia-reperfusion (I/R) injury remains a significant cause of morbidity and mortality worldwide. Inspired by the impact of I/R on the Cx43 trafficking to the intercalated discs (ICDs), we aim to explore the potential mechanisms underlying the downregulation of Cx43 in ICDs after myocardial I/R. Gene set enrichment analysis (GSEA), Western blotting, and immunofluorescence experiments showed that Myocardial I/R activated the P38MAPK signaling pathway and promoted microtubule depolymerization. Inhibition of P38MAPK signaling pathway activation attenuated I/R-induced microtubule depolymerization. The ability of SB203580 to recover the distribution of Cx43 and electrophysiological parameters in I/R myocardium depended on microtubule stability. Our study suggests that microtubule depolymerization caused by the activation of the P38MAPK signaling pathway is an important mechanism underlying the downregulation of Cx43 in ICDs after myocardial I/R.
Collapse
Affiliation(s)
- Xiang Huang
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Xue Bai
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jing Yi
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Tingju Hu
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Li An
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Hong Gao
- Guizhou Hospital, Branch of the First Affiliated Hospital of Sun Yat-Sen University, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
8
|
Domingues N, Catarino S, Cristóvão B, Rodrigues L, Carvalho FA, Sarmento MJ, Zuzarte M, Almeida J, Ribeiro-Rodrigues T, Correia-Rodrigues Â, Fernandes F, Rodrigues-Santos P, Aasen T, Santos NC, Korolchuk VI, Gonçalves T, Milosevic I, Raimundo N, Girão H. Connexin43 promotes exocytosis of damaged lysosomes through actin remodelling. EMBO J 2024; 43:3627-3649. [PMID: 39044100 PMCID: PMC11377567 DOI: 10.1038/s44318-024-00177-3] [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: 12/11/2023] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
A robust and efficient cellular response to lysosomal membrane damage prevents leakage from the lysosome lumen into the cytoplasm. This response is understood to happen through either lysosomal membrane repair or lysophagy. Here we report exocytosis as a third response mechanism to lysosomal damage, which is further potentiated when membrane repair or lysosomal degradation mechanisms are impaired. We show that Connexin43 (Cx43), a protein canonically associated with gap junctions, is recruited from the plasma membrane to damaged lysosomes, promoting their secretion and accelerating cell recovery. The effects of Cx43 on lysosome exocytosis are mediated by a reorganization of the actin cytoskeleton that increases plasma membrane fluidity and decreases cell stiffness. Furthermore, we demonstrate that Cx43 interacts with the actin nucleator Arp2, the activity of which was shown to be necessary for Cx43-mediated actin rearrangement and lysosomal exocytosis following damage. These results define a novel mechanism of lysosomal quality control whereby Cx43-mediated actin remodelling potentiates the secretion of damaged lysosomes.
Collapse
Affiliation(s)
- Neuza Domingues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Steve Catarino
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Beatriz Cristóvão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria João Sarmento
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Jani Almeida
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Teresa Ribeiro-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Ânia Correia-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Fábio Fernandes
- Institute for Bioengineering and Biosciences (IBB) and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Paulo Rodrigues-Santos
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Trond Aasen
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Teresa Gonçalves
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- University of Oxford, Centre for Human Genetics, Nuffield Department of Medicine, Oxford, UK
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Henrique Girão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal.
| |
Collapse
|
9
|
Jara O, Maripillán J, Momboisse F, Cárdenas AM, García IE, Martínez AD. Differential Regulation of Hemichannels and Gap Junction Channels by RhoA GTPase and Actin Cytoskeleton: A Comparative Analysis of Cx43 and Cx26. Int J Mol Sci 2024; 25:7246. [PMID: 39000353 PMCID: PMC11242593 DOI: 10.3390/ijms25137246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Connexins (Cxs) are transmembrane proteins that assemble into gap junction channels (GJCs) and hemichannels (HCs). Previous researches support the involvement of Rho GTPases and actin microfilaments in the trafficking of Cxs, formation of GJCs plaques, and regulation of channel activity. Nonetheless, it remains uncertain whether distinct types of Cxs HCs and GJCs respond differently to Rho GTPases or changes in actin polymerization/depolymerization dynamics. Our investigation revealed that inhibiting RhoA, a small GTPase that controls actin polymerization, or disrupting actin microfilaments with cytochalasin B (Cyto-B), resulted in reduced GJCs plaque size at appositional membranes and increased transport of HCs to non-appositional plasma membrane regions. Notably, these effects were consistent across different Cx types, since Cx26 and Cx43 exhibited similar responses, despite having distinct trafficking routes to the plasma membrane. Functional assessments showed that RhoA inhibition and actin depolymerization decreased the activity of Cx43 GJCs while significantly increasing HC activity. However, the functional status of GJCs and HCs composed of Cx26 remained unaffected. These results support the hypothesis that RhoA, through its control of the actin cytoskeleton, facilitates the transport of HCs to appositional cell membranes for GJCs formation while simultaneously limiting the positioning of free HCs at non-appositional cell membranes, independently of Cx type. This dynamic regulation promotes intercellular communications and reduces non-selective plasma membrane permeability through a Cx-type dependent mechanism, whereby the activity of Cx43 HCs and GJCs are differentially affected but Cx26 channels remain unchanged.
Collapse
Affiliation(s)
- Oscar Jara
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, USA
| | - Jaime Maripillán
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
| | - Fanny Momboisse
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, 75013 Paris, France
| | - Ana María Cárdenas
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
| | - Isaac E García
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
- Laboratorio de Fisiología Molecular y Biofísica, Facultad de Odontología, Universidad de Valparaíso, Valparaíso 2360004, Chile
- Centro de Investigación en Ciencias Odontológicas y Médicas, CICOM, Universidad de Valparaíso, Valparaíso 2360004, Chile
| | - Agustín D Martínez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
| |
Collapse
|
10
|
Padget RL, Zeitz MJ, Blair GA, Wu X, North MD, Tanenbaum MT, Stanley KE, Phillips CM, King DR, Lamouille S, Gourdie RG, Hoeker GS, Swanger SA, Poelzing S, Smyth JW. Acute Adenoviral Infection Elicits an Arrhythmogenic Substrate Prior to Myocarditis. Circ Res 2024; 134:892-912. [PMID: 38415360 PMCID: PMC11003857 DOI: 10.1161/circresaha.122.322437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Viral cardiac infection represents a significant clinical challenge encompassing several etiological agents, disease stages, complex presentation, and a resulting lack of mechanistic understanding. Myocarditis is a major cause of sudden cardiac death in young adults, where current knowledge in the field is dominated by later disease phases and pathological immune responses. However, little is known regarding how infection can acutely induce an arrhythmogenic substrate before significant immune responses. Adenovirus is a leading cause of myocarditis, but due to species specificity, models of infection are lacking, and it is not understood how adenoviral infection may underlie sudden cardiac arrest. Mouse adenovirus type-3 was previously reported as cardiotropic, yet it has not been utilized to understand the mechanisms of cardiac infection and pathology. METHODS We have developed mouse adenovirus type-3 infection as a model to investigate acute cardiac infection and molecular alterations to the infected heart before an appreciable immune response or gross cardiomyopathy. RESULTS Optical mapping of infected hearts exposes decreases in conduction velocity concomitant with increased Cx43Ser368 phosphorylation, a residue known to regulate gap junction function. Hearts from animals harboring a phospho-null mutation at Cx43Ser368 are protected against mouse adenovirus type-3-induced conduction velocity slowing. Additional to gap junction alterations, patch clamping of mouse adenovirus type-3-infected adult mouse ventricular cardiomyocytes reveals prolonged action potential duration as a result of decreased IK1 and IKs current density. Turning to human systems, we find human adenovirus type-5 increases phosphorylation of Cx43Ser368 and disrupts synchrony in human induced pluripotent stem cell-derived cardiomyocytes, indicating common mechanisms with our mouse whole heart and adult cardiomyocyte data. CONCLUSIONS Together, these findings demonstrate that adenoviral infection creates an arrhythmogenic substrate through direct targeting of gap junction and ion channel function in the heart. Such alterations are known to precipitate arrhythmias and likely contribute to sudden cardiac death in acutely infected patients.
Collapse
Affiliation(s)
- Rachel L. Padget
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Michael J. Zeitz
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Grace A. Blair
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Xiaobo Wu
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Michael D. North
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | | | - Kari E. Stanley
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Chelsea M. Phillips
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - D. Ryan King
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Samy Lamouille
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Robert G. Gourdie
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Gregory S. Hoeker
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
| | - Sharon A. Swanger
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Steven Poelzing
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - James W. Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
11
|
Liu H, Wang L, Xu H, Tan B, Yi Q, Deng H, Chen Y, He B, Tian J, Zhu J. Quantitative proteomic and phosphoproteomic analysis reveal the relationship between mitochondrial dysfunction and cytoskeletal remodeling in hiPSC-CMs deficient in PINK1. J Transl Med 2023; 21:581. [PMID: 37649075 PMCID: PMC10466879 DOI: 10.1186/s12967-023-04467-y] [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: 05/17/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are seed cells that can be used for alternative treatment of myocardial damage. However, their immaturity limits their clinical application. Mitochondrial development accompanies cardiomyocyte maturation, and PINK1 plays an important role in the regulation of mitochondrial quality. However, the role and mechanism of PINK1 in cardiomyocyte development remain unclear. METHODS We used proteomic and phosphoproteomic to identify protein and phosphosite changes in hiPSC-CMs deficient in PINK1. Bioinformatics analysis was performed to identify the potential biological functions and regulatory mechanisms of these differentially expressed proteins and validate potential downstream mechanisms. RESULTS Deletion of PINK1 resulted in mitochondrial structural breakdown and dysfunction, accompanied by disordered myofibrils arrangement. hiPSC-CMs deficient in PINK1 exhibited significantly decreased expression of mitochondrial ATP synthesis proteins and inhibition of the oxidative phosphorylation pathway. In contrast, the expression of proteins related to cardiac pathology was increased, and the phosphoproteins involved in cytoskeleton construction were significantly altered. Mechanistically, PINK1 deletion damaged the mitochondrial cristae of hiPSC-CMs and reduced the efficiency of mitochondrial respiratory chain assembly. CONCLUSION The significantly differentially expressed proteins identified in this study highlight the important role of PINK1 in regulating mitochondrial quality in hiPSC-CMs. PINK1-mediated mitochondrial respiratory chain assembly is the basis for mitochondrial function. Whereas the cytoskeleton may be adaptively altered in response to mitochondrial dysfunction caused by PINK1 deletion, inadequate energy supply hinders myocardial development. These findings facilitate the exploration of the mechanism of PINK1 in cardiomyocyte development and guide efforts to promote the maturation of hiPSC-CMs.
Collapse
Affiliation(s)
- Huiwen Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wang
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Xu
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Tan
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Yi
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongrong Deng
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yunxia Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bolin He
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Blood Transfusion, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Tian
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiovascular (Internal Medicine), Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Zhu
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Zhang J, Wu X, Liang Y, Kelly G, Burt JM, Zhang L, Wang T. Particulate matter increases connexin 43 expression and exacerbates endothelial barrier disruption. Am J Transl Res 2023; 15:5099-5109. [PMID: 37692924 PMCID: PMC10492082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/29/2023] [Indexed: 09/12/2023]
Abstract
OBJECTIVES Particulate Matter (PM) air pollution is known to exacerbate cardiopulmonary diseases. We previously demonstrated that PM mediates endothelial injury and barrier disruption by modulating the endothelial cytoskeleton and cell-cell junctions, but the effects of PM exposure on cell-cell communication and gap junction activity are still unknown. METHODS This study focused on the characterization of PM-regulated endothelial dysfunction through connexin 43 (Cx43), the most abundant gap junction protein expressed in lung endothelial cells (ECs), using cultured human lung endothelial cells and a well-characterized PM sample. RESULTS PM exposure induced a time-dependent increase of Cx43 in human lung ECs at both the mRNA and protein levels. N-acetylcysteine (NAC), a reactive oxygen species (ROS) scavenger, significantly suppressed PM-induced Cx43 expression. Cx43 proteins on the plasma membrane and ER/Golgi apparatus were elevated in response to a PM challenge. In addition, PM induced gap junction activity, which was indicated by green fluorescence dye transfer between two adjacent ECs. Moreover, GAP27, a selective Cx43 channel inhibitor, attenuated PM-induced human lung EC barrier disruption, which was reflected by rescued trans-endothelial electrical resistance (TER) with an electric cell-substrate impedance sensing system. Moreover, knocking down Cx43 alleviated PM-induced myosin light chain (MLC) phosphorylation. CONCLUSIONS These results strongly suggest that Cx43 plays a key role in PM-mediated endothelial barrier disruption and signal transduction. Cx43 may be a therapeutic target in PM-mediated cardiopulmonary disorders.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Medicine, University of ArizonaTuscon, AZ, USA
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical UniversityBeijing, China
| | - Xiaomin Wu
- Department of Medicine, University of ArizonaTuscon, AZ, USA
| | - Ying Liang
- Department of Medicine, University of ArizonaTuscon, AZ, USA
| | - Gabriel Kelly
- Department of Medicine, University of ArizonaTuscon, AZ, USA
| | - Janis M Burt
- Department of Physiology, University of ArizonaTuscon, AZ, USA
| | - Liming Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical UniversityBeijing, China
| | - Ting Wang
- Department of Medicine, University of ArizonaTuscon, AZ, USA
- Center of Translational Science, Florida International University11350 SW Village Parkway, Port St. Lucie, FL, USA
| |
Collapse
|
13
|
Segretain D, Di Marco M, Dufeu C, Carette D, Trubuil A, Pointis G. Cooperative cell-cell actin network remodeling to perform Gap junction endocytosis. Basic Clin Androl 2023; 33:20. [PMID: 37533006 PMCID: PMC10399049 DOI: 10.1186/s12610-023-00194-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/16/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND The endocytosis of Gap junction plaques (GJP) requires cytoskeletal forces to internalize such large membranous structures. Actin, which partners the connexin proteins constituting Gap junctions and is located close to Annular Gap Junctions (AGJ), could be actively involved in this physiological process. RESULTS Electron Microscopy and Light Microscopy images, associated with time-lapse analysis and 3D reconstruction, used at high resolution and enhanced using ImageJ based software analysis, revealed that: i) actin cables, originating from Donor cells, insert on the edge of GJP and contribute to their invagination, giving rise to AGJ, whereas actin cables on the Acceptor cell side of the plaque are not modified; ii) actin cables from the Donor cell are continuous with the actin network present over the entire GJP surface. These actin cables fuse at a single point distant from the plaque, which then detaches itself from the membrane, condensing to form an actin mass during the final internalization process; iii) the Acceptor cell participates in the last step of the endocytic invagination process by forming an annular actin structure known as an actin ring. CONCLUSIONS Together, these data suggest that the endocytosis of GJP is an example of a unique cooperative mechanism between the Donor (the traction of its actin cables) and the Acceptor cells (forming the actin ring).
Collapse
Affiliation(s)
- Dominique Segretain
- UMR S1147, Université Paris Descartes, 45 Rue Des Saints-Pères, 75006, Paris, France.
| | - Mathilde Di Marco
- UMR S1147, Université Paris Descartes, 45 Rue Des Saints-Pères, 75006, Paris, France
- Present Address: Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005, Paris, France
| | - Chloé Dufeu
- Faculté de Pharmacie, Université Paris-Saclay, Saclay, France
| | | | - Alain Trubuil
- MaIAGE, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, France
| | - Georges Pointis
- INSERM U 1065, Team 5 Physiopathological Control of Germ Cell Proliferation: Genomic and Non-Genomic Mechanisms, University of Nice Sophia-Antipolis, 151 Route Saint-Antoine de Ginestière BP 2 3194, 06204, Nice Cedex 3, France
| |
Collapse
|
14
|
Acharya BR, Fang JS, Jeffery ED, Chavkin NW, Genet G, Vasavada H, Nelson EA, Sheynkman GM, Humphries MJ, Hirschi KK. Connexin 37 sequestering of activated-ERK in the cytoplasm promotes p27-mediated endothelial cell cycle arrest. Life Sci Alliance 2023; 6:e202201685. [PMID: 37197981 PMCID: PMC10192821 DOI: 10.26508/lsa.202201685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023] Open
Abstract
Connexin37-mediated regulation of cell cycle modulators and, consequently, growth arrest lack mechanistic understanding. We previously showed that arterial shear stress up-regulates Cx37 in endothelial cells and activates a Notch/Cx37/p27 signaling axis to promote G1 cell cycle arrest, and this is required to enable arterial gene expression. However, how induced expression of a gap junction protein, Cx37, up-regulates cyclin-dependent kinase inhibitor p27 to enable endothelial growth suppression and arterial specification is unclear. Herein, we fill this knowledge gap by expressing wild-type and regulatory domain mutants of Cx37 in cultured endothelial cells expressing the Fucci cell cycle reporter. We determined that both the channel-forming and cytoplasmic tail domains of Cx37 are required for p27 up-regulation and late G1 arrest. Mechanistically, the cytoplasmic tail domain of Cx37 interacts with, and sequesters, activated ERK in the cytoplasm. This then stabilizes pERK nuclear target Foxo3a, which up-regulates p27 transcription. Consistent with previous studies, we found this Cx37/pERK/Foxo3a/p27 signaling axis functions downstream of arterial shear stress to promote endothelial late G1 state and enable up-regulation of arterial genes.
Collapse
Affiliation(s)
- Bipul R Acharya
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Jennifer S Fang
- Department of Molecular Biology & Biochemistry, University of California at Irvine, Irvine, CA, USA
| | - Erin D Jeffery
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Nicholas W Chavkin
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Gael Genet
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Hema Vasavada
- Departments of Medicine and Genetics, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Elizabeth A Nelson
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Gloria M Sheynkman
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Martin J Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Karen K Hirschi
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Departments of Medicine and Genetics, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
15
|
Scott H, Dong L, Stevenson A, MacDonald AI, Srinivasan S, Massimi P, Banks L, Martin PE, Johnstone SR, Graham SV. The human discs large protein 1 interacts with and maintains connexin 43 at the plasma membrane in keratinocytes. J Cell Sci 2023; 136:jcs259984. [PMID: 37288673 PMCID: PMC10309592 DOI: 10.1242/jcs.259984] [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: 03/08/2022] [Accepted: 04/21/2023] [Indexed: 05/10/2023] Open
Abstract
Gap junction channels, composed of connexins, allow direct cell-to-cell communication. Connexin 43 (Cx43; also known as GJA1) is widely expressed in tissues, including the epidermis. In a previous study of human papillomavirus-positive cervical epithelial tumour cells, we identified Cx43 as a binding partner of the human homologue of Drosophila Discs large (Dlg1; also known as SAP97). Dlg1 is a member of the membrane associated-guanylate kinase (MAGUK) scaffolding protein family, which is known to control cell shape and polarity. Here, we show that Cx43 also interacts with Dlg1 in uninfected keratinocytes in vitro and in keratinocytes, dermal cells and adipocytes in normal human epidermis in vivo. Depletion of Dlg1 in keratinocytes did not alter Cx43 transcription but was associated with a reduction in Cx43 protein levels. Reduced Dlg1 levels in keratinocytes resulted in a reduction in Cx43 at the plasma membrane with a concomitant reduction in gap junctional intercellular communication and relocation of Cx43 to the Golgi compartment. Our data suggest a key role for Dlg1 in maintaining Cx43 at the plasma membrane in keratinocytes.
Collapse
Affiliation(s)
- Harry Scott
- MRC-University of Glasgow Centre for Virus Research, School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK
| | - Li Dong
- MRC-University of Glasgow Centre for Virus Research, School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK
| | - Andrew Stevenson
- MRC-University of Glasgow Centre for Virus Research, School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK
| | - Alasdair I. MacDonald
- MRC-University of Glasgow Centre for Virus Research, School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK
| | - Sharmila Srinivasan
- Translation Research Platform for Veterinary Biologicals, Chennai, Tamil Nadu, India
| | - Paola Massimi
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Lawrence Banks
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Patricia E. Martin
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK
| | - Scott R. Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke VA 24016, USA
| | - Sheila V. Graham
- MRC-University of Glasgow Centre for Virus Research, School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK
| |
Collapse
|
16
|
Basu I, Li H, Trease AJ, Sorgen PL. Regulation of Cx43 Gap Junction Intercellular Communication by Bruton's Tyrosine Kinase and Interleukin-2-Inducible T-Cell Kinase. Biomolecules 2023; 13:biom13040660. [PMID: 37189407 DOI: 10.3390/biom13040660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
T and B cell receptor signaling involves the activation of Akt, MAPKs, and PKC as well as an increase in intracellular Ca2+ and calmodulin activation. While these coordinate the rapid turnover of gap junctions, also implicated in this process is Src, which is not activated as part of T and B cell receptor signaling. An in vitro kinase screen identified that Bruton's tyrosine kinase (BTK) and interleukin-2-inducible T-cell kinase (ITK) phosphorylate Cx43. Mass spectroscopy revealed that BTK and ITK phosphorylate Cx43 residues Y247, Y265, and Y313, which are identical to the residues phosphorylated by Src. Overexpression of BTK or ITK in the HEK-293T cells led to increased Cx43 tyrosine phosphorylation as well as decreased gap junction intercellular communication (GJIC) and Cx43 membrane localization. In the lymphocytes, activation of the B cell receptor (Daudi cells) or T cell receptor (Jurkat cells) increased the BTK and ITK activity, respectively. While this led to increased tyrosine phosphorylation of Cx43 and decreased GJIC, the cellular localization of Cx43 changed little. We have previously identified that Pyk2 and Tyk2 also phosphorylate Cx43 at residues Y247, Y265, and Y313 with a similar cellular fate to that of Src. With phosphorylation critical to Cx43 assembly and turnover, and kinase expression varying between different cell types, there would be a need for different kinases to achieve the same regulation of Cx43. The work presented herein suggests that in the immune system, ITK and BTK have the capacity for the tyrosine phosphorylation of Cx43 to alter the gap junction function in a similar manner as Pyk2, Tyk2, and Src.
Collapse
Affiliation(s)
- Ishika Basu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Hanjun Li
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Andrew J Trease
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Paul L Sorgen
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
17
|
Correa F, Enríquez-Cortina C, Silva-Palacios A, Román-Anguiano N, Gil-Hernández A, Ostolga-Chavarría M, Soria-Castro E, Hernández-Rizo S, Heros PDL, Chávez-Canales M, Zazueta C. Actin-Cytoskeleton Drives Caveolae Signaling to Mitochondria during Postconditioning. Cells 2023; 12:492. [PMID: 36766835 PMCID: PMC9914812 DOI: 10.3390/cells12030492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/09/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Caveolae-associated signaling toward mitochondria contributes to the cardioprotective mechanisms against ischemia-reperfusion (I/R) injury induced by ischemic postconditioning. In this work, we evaluated the role that the actin-cytoskeleton network exerts on caveolae-mitochondria communication during postconditioning. Isolated rat hearts subjected to I/R and to postconditioning were treated with latrunculin A, a cytoskeleton disruptor. Cardiac function was compared between these hearts and those exposed only to I/R and to the cardioprotective maneuver. Caveolae and mitochondria structures were determined by electron microscopy and maintenance of the actin-cytoskeleton was evaluated by phalloidin staining. Caveolin-3 and other putative caveolae-conforming proteins were detected by immunoblot analysis. Co-expression of caveolin-3 and actin was evaluated both in lipid raft fractions and in heart tissue from the different groups. Mitochondrial function was assessed by respirometry and correlated with cholesterol levels. Treatment with latrunculin A abolishes the cardioprotective postconditioning effect, inducing morphological and structural changes in cardiac tissue, reducing F-actin staining and diminishing caveolae formation. Latrunculin A administration to post-conditioned hearts decreases the interaction between caveolae-forming proteins, the co-localization of caveolin with actin and inhibits oxygen consumption rates in both subsarcolemmal and interfibrillar mitochondria. We conclude that actin-cytoskeleton drives caveolae signaling to mitochondria during postconditioning, supporting their functional integrity and contributing to cardiac adaption against reperfusion injury.
Collapse
Affiliation(s)
- Francisco Correa
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Colonia Sección XVI, Mexico City 14080, Mexico
| | - Cristina Enríquez-Cortina
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Colonia Sección XVI, Mexico City 14080, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Colonia Sección XVI, Mexico City 14080, Mexico
| | - Nadia Román-Anguiano
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Colonia Sección XVI, Mexico City 14080, Mexico
| | - Aurora Gil-Hernández
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Colonia Sección XVI, Mexico City 14080, Mexico
| | - Marcos Ostolga-Chavarría
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Colonia Sección XVI, Mexico City 14080, Mexico
| | - Elizabeth Soria-Castro
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Colonia Sección XVI, Mexico City 14080, Mexico
| | - Sharik Hernández-Rizo
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 14080, Mexico
| | - Paola de los Heros
- Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico
| | - María Chávez-Canales
- Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Colonia Sección XVI, Mexico City 14080, Mexico
| |
Collapse
|
18
|
Kwek XY, Hall AR, Lim WW, Katwadi K, Soong PL, Grishina E, Lin KH, Crespo-Avilan G, Yap EP, Ismail NI, Chinda K, Chung YY, Wei H, Shim W, Montaigne D, Tinker A, Ong SB, Hausenloy DJ. Role of cardiac mitofusins in cardiac conduction following simulated ischemia-reperfusion. Sci Rep 2022; 12:21049. [PMID: 36473917 PMCID: PMC9727036 DOI: 10.1038/s41598-022-25625-0] [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: 08/09/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction induced by acute cardiac ischemia-reperfusion (IR), may increase susceptibility to arrhythmias by perturbing energetics, oxidative stress production and calcium homeostasis. Although changes in mitochondrial morphology are known to impact on mitochondrial function, their role in cardiac arrhythmogenesis is not known. To assess action potential duration (APD) in cardiomyocytes from the Mitofusins-1/2 (Mfn1/Mfn2)-double-knockout (Mfn-DKO) compared to wild-type (WT) mice, optical-electrophysiology was conducted. To measure conduction velocity (CV) in atrial and ventricular tissue from the Mfn-DKO and WT mice, at both baseline and following simulated acute IR, multi-electrode array (MEA) was employed. Intracellular localization of connexin-43 (Cx43) at baseline was evaluated by immunohistochemistry, while Cx-43 phosphorylation was assessed by Western-blotting. Mfn-DKO cardiomyocytes demonstrated an increased APD. At baseline, CV was significantly lower in the left ventricle of the Mfn-DKO mice. CV decreased with simulated-ischemia and returned to baseline levels during simulated-reperfusion in WT but not in atria of Mfn-DKO mice. Mfn-DKO hearts displayed increased Cx43 lateralization, although phosphorylation of Cx43 at Ser-368 did not differ. In summary, Mfn-DKO mice have increased APD and reduced CV at baseline and impaired alterations in CV following cardiac IR. These findings were associated with increased Cx43 lateralization, suggesting that the mitofusins may impact on post-MI cardiac-arrhythmogenesis.
Collapse
Affiliation(s)
- Xiu-Yi Kwek
- grid.419385.20000 0004 0620 9905National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore
| | - Andrew R. Hall
- grid.83440.3b0000000121901201The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Wei-Wen Lim
- grid.419385.20000 0004 0620 9905National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore ,grid.428397.30000 0004 0385 0924Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Khairunnisa Katwadi
- grid.428397.30000 0004 0385 0924Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Poh Loong Soong
- grid.4280.e0000 0001 2180 6431Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Cardiovascular Translational Program, Cardiovascular Research Institute (CVRI), National University of Singapore, Singapore, Singapore ,grid.412106.00000 0004 0621 9599Department of Medicine, National University Hospital of Singapore (NUHS), Singapore, Singapore ,Ternion Biosciences, Singapore, Singapore
| | | | | | - Gustavo Crespo-Avilan
- grid.419385.20000 0004 0620 9905National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore ,grid.428397.30000 0004 0385 0924Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore ,grid.8664.c0000 0001 2165 8627Department of Biochemistry, Medical Faculty, Justus Liebig-University, Giessen, Germany
| | - En Ping Yap
- grid.419385.20000 0004 0620 9905National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore
| | - Nur Izzah Ismail
- grid.10784.3a0000 0004 1937 0482Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, Chinese University of Hong Kong (CUHK), Hong Kong, SAR China ,grid.10784.3a0000 0004 1937 0482Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), Hong Kong, SAR China ,Hong Kong Hub of Paediatric Excellence (HK HOPE), Hong Kong Children’s Hospital (HKCH), Kowloon Bay, Hong Kong, SAR China
| | - Kroekkiat Chinda
- grid.412029.c0000 0000 9211 2704Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand ,grid.412029.c0000 0000 9211 2704Integrative Cardiovascular Research Unit, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Ying Ying Chung
- grid.428397.30000 0004 0385 0924Centre for Vision Research, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Heming Wei
- grid.414963.d0000 0000 8958 3388Research Laboratory, KK Women’s & Children’s Hospital, Singapore, Singapore
| | - Winston Shim
- grid.486188.b0000 0004 1790 4399Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore, Singapore
| | - David Montaigne
- grid.503422.20000 0001 2242 6780Inserm, CHU Lille, Institut Pasteur Lille, U1011-European Genomic Institute for Diabetes (EGID), University of Lille, 59000 Lille, France
| | - Andrew Tinker
- grid.4868.20000 0001 2171 1133Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Sang-Bing Ong
- grid.10784.3a0000 0004 1937 0482Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, Chinese University of Hong Kong (CUHK), Hong Kong, SAR China ,grid.10784.3a0000 0004 1937 0482Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), Hong Kong, SAR China ,Hong Kong Hub of Paediatric Excellence (HK HOPE), Hong Kong Children’s Hospital (HKCH), Kowloon Bay, Hong Kong, SAR China ,grid.9227.e0000000119573309Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology-The Chinese University of Hong Kong (KIZ-CUHK), Chinese Academy of Sciences, Kunming, Yunnan China ,grid.10784.3a0000 0004 1937 0482Shenzhen Research Institute (SZRI), Chinese University of Hong Kong (CUHK), Shenzhen, China
| | - Derek J. Hausenloy
- grid.419385.20000 0004 0620 9905National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore ,grid.83440.3b0000000121901201The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK ,grid.428397.30000 0004 0385 0924Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Shimura D, Shaw RM. GJA1-20k and Mitochondrial Dynamics. Front Physiol 2022; 13:867358. [PMID: 35399255 PMCID: PMC8983841 DOI: 10.3389/fphys.2022.867358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/08/2022] [Indexed: 01/07/2023] Open
Abstract
Connexin 43 (Cx43) is the primary gap junction protein of mammalian heart ventricles and is encoded by the gene Gja1 which has a single coding exon and therefore cannot be spliced. We previously identified that Gja1 mRNA undergoes endogenous internal translation initiated at one of several internal AUG (M) start codons, generating N-terminal truncated protein isoforms that retain the C-terminus distal to the start site. GJA1-20k, whose translation initiates at mRNA M213, is usually the most abundant isoform in cells and greatly increases after ischemic and metabolic stress. GJA1-20k consists of a small segment of the last transmembrane domain and the complete C-terminus tail of Cx43, with a total size of about 20 kDa. The original role identified for GJA1-20k is as an essential subunit that facilitates the trafficking of full-length Cx43 hexameric hemichannels to cell-cell contacts, generating traditional gap junctions between adjacent cells facilitating, in cardiac muscle, efficient spread of electrical excitation. GJA1-20k deficient mice (generated by a M213L substitution in Gja1) suffer poor electrical coupling between cardiomycytes and arrhythmogenic sudden death two to 4 weeks after their birth. We recently identified that exogenous GJA1-20k expression also mimics the effect of ischemic preconditioning in mouse heart. Furthermore, GJA1-20k localizes to the mitochondrial outer membrane and induces a protective and DRP1 independent form of mitochondrial fission, preserving ATP production and generating less reactive oxygen species (ROS) under metabolic stress, providing powerful protection of myocardium to ischemic insult. In this manuscript, we focus on the detailed roles of GJA1-20k in mitochondria, and its interaction with the actin cytoskeleton.
Collapse
|
20
|
Distributed synthesis of sarcolemmal and sarcoplasmic reticulum membrane proteins in cardiac myocytes. Basic Res Cardiol 2021; 116:63. [PMID: 34713358 PMCID: PMC8553722 DOI: 10.1007/s00395-021-00895-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 02/05/2023]
Abstract
It is widely assumed that synthesis of membrane proteins, particularly in the heart, follows the classical secretory pathway with mRNA translation occurring in perinuclear regions followed by protein trafficking to sites of deployment. However, this view is based on studies conducted in less-specialized cells, and has not been experimentally addressed in cardiac myocytes. Therefore, we undertook direct experimental investigation of protein synthesis in cardiac tissue and isolated myocytes using single-molecule visualization techniques and a novel proximity-ligated in situ hybridization approach for visualizing ribosome-associated mRNA molecules for a specific protein species, indicative of translation sites. We identify here, for the first time, that the molecular machinery for membrane protein synthesis occurs throughout the cardiac myocyte, and enables distributed synthesis of membrane proteins within sub-cellular niches where the synthesized protein functions using local mRNA pools trafficked, in part, by microtubules. We also observed cell-wide distribution of membrane protein mRNA in myocardial tissue from both non-failing and hypertrophied (failing) human hearts, demonstrating an evolutionarily conserved distributed mechanism from mouse to human. Our results identify previously unanticipated aspects of local control of cardiac myocyte biology and highlight local protein synthesis in cardiac myocytes as an important potential determinant of the heart’s biology in health and disease.
Collapse
|
21
|
Retamal MA, Fernandez-Olivares A, Stehberg J. Over-activated hemichannels: A possible therapeutic target for human diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166232. [PMID: 34363932 DOI: 10.1016/j.bbadis.2021.166232] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022]
Abstract
In our body, all the cells are constantly sharing chemical and electrical information with other cells. This intercellular communication allows them to respond in a concerted way to changes in the extracellular milieu. Connexins are transmembrane proteins that have the particularity of forming two types of channels; hemichannels and gap junction channels. Under normal conditions, hemichannels allow the controlled release of signaling molecules to the extracellular milieu. However, under certain pathological conditions, over-activated hemichannels can induce and/or exacerbate symptoms. In the last decade, great efforts have been put into developing new tools that can modulate these over-activated hemichannels. Small molecules, antibodies and mimetic peptides have shown a potential for the treatment of human diseases. In this review, we summarize recent findings in the field of hemichannel modulation via specific tools, and how these tools could improve patient outcome in certain pathological conditions.
Collapse
Affiliation(s)
- Mauricio A Retamal
- Universidad del Desarrollo, Programa de Comunicación Celular en Cáncer, Santiago, Chile; Universidad del Desarrollo, Centro de Fisiología Celular e Integrativa, Santiago, Chile.
| | | | - Jimmy Stehberg
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
22
|
KRAP tethers IP 3 receptors to actin and licenses them to evoke cytosolic Ca 2+ signals. Nat Commun 2021; 12:4514. [PMID: 34301929 PMCID: PMC8302619 DOI: 10.1038/s41467-021-24739-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/01/2021] [Indexed: 02/06/2023] Open
Abstract
Regulation of IP3 receptors (IP3Rs) by IP3 and Ca2+ allows regenerative Ca2+ signals, the smallest being Ca2+ puffs, which arise from coordinated openings of a few clustered IP3Rs. Cells express thousands of mostly mobile IP3Rs, yet Ca2+ puffs occur at a few immobile IP3R clusters. By imaging cells with endogenous IP3Rs tagged with EGFP, we show that KRas-induced actin-interacting protein (KRAP) tethers IP3Rs to actin beneath the plasma membrane. Loss of KRAP abolishes Ca2+ puffs and the global increases in cytosolic Ca2+ concentration evoked by more intense stimulation. Over-expressing KRAP immobilizes additional IP3R clusters and results in more Ca2+ puffs and larger global Ca2+ signals. Endogenous KRAP determines which IP3Rs will respond: it tethers IP3R clusters to actin alongside sites where store-operated Ca2+ entry occurs, licenses IP3Rs to evoke Ca2+ puffs and global cytosolic Ca2+ signals, implicates the actin cytoskeleton in IP3R regulation and may allow local activation of Ca2+ entry.
Collapse
|
23
|
Kléber AG, Jin Q. Coupling between cardiac cells-An important determinant of electrical impulse propagation and arrhythmogenesis. ACTA ACUST UNITED AC 2021; 2:031301. [PMID: 34296210 DOI: 10.1063/5.0050192] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/09/2021] [Indexed: 01/30/2023]
Abstract
Cardiac arrhythmias are an important cause of sudden cardiac death-a devastating manifestation of many underlying causes, such as heart failure and ischemic heart disease leading to ventricular tachyarrhythmias and ventricular fibrillation, and atrial fibrillation causing cerebral embolism. Cardiac electrical propagation is a main factor in the initiation and maintenance of cardiac arrhythmias. In the heart, gap junctions are the basic unit at the cellular level that host intercellular low-resistance channels for the diffusion of ions and small regulatory molecules. The dual voltage clamp technique enabled the direct measurement of electrical conductance between cells and recording of single gap junction channel openings. The rapid turnover of gap junction channels at the intercalated disk implicates a highly dynamic process of trafficking and internalization of gap junction connexons. Recently, non-canonical roles of gap junction proteins have been discovered in mitochondria function, cytoskeletal organization, trafficking, and cardiac rescue. At the tissue level, we explain the concepts of linear propagation and safety factor based on the model of linear cellular structure. Working myocardium is adequately represented as a discontinuous cellular network characterized by cellular anisotropy and connective tissue heterogeneity. Electrical propagation in discontinuous cellular networks reflects an interplay of three main factors: cell-to-cell electrical coupling, flow of electrical charge through the ion channels, and the microscopic tissue structure. This review provides a state-of-the-art update of the cardiac gap junction channels and their role in cardiac electrical impulse propagation and highlights a combined approach of genetics, cell biology, and physics in modern cardiac electrophysiology.
Collapse
Affiliation(s)
- André G Kléber
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Qianru Jin
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| |
Collapse
|
24
|
Mechanisms and Regulation of Cardiac Ca V1.2 Trafficking. Int J Mol Sci 2021; 22:ijms22115927. [PMID: 34072954 PMCID: PMC8197997 DOI: 10.3390/ijms22115927] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 01/05/2023] Open
Abstract
During cardiac excitation contraction coupling, the arrival of an action potential at the ventricular myocardium triggers voltage-dependent L-type Ca2+ (CaV1.2) channels in individual myocytes to open briefly. The level of this Ca2+ influx tunes the amplitude of Ca2+-induced Ca2+ release from ryanodine receptors (RyR2) on the junctional sarcoplasmic reticulum and thus the magnitude of the elevation in intracellular Ca2+ concentration and ultimately the downstream contraction. The number and activity of functional CaV1.2 channels at the t-tubule dyads dictates the amplitude of the Ca2+ influx. Trafficking of these channels and their auxiliary subunits to the cell surface is thus tightly controlled and regulated to ensure adequate sarcolemmal expression to sustain this critical process. To that end, recent discoveries have revealed the existence of internal reservoirs of preformed CaV1.2 channels that can be rapidly mobilized to enhance sarcolemmal expression in times of acute stress when hemodynamic and metabolic demand increases. In this review, we provide an overview of the current thinking on CaV1.2 channel trafficking dynamics in the heart. We highlight the numerous points of control including the biosynthetic pathway, the endosomal recycling pathway, ubiquitination, and lysosomal and proteasomal degradation pathways, and discuss the effects of β-adrenergic and angiotensin receptor signaling cascades on this process.
Collapse
|
25
|
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
|
26
|
Del Villar SG, Voelker TL, Westhoff M, Reddy GR, Spooner HC, Navedo MF, Dickson EJ, Dixon RE. β-Adrenergic control of sarcolemmal Ca V1.2 abundance by small GTPase Rab proteins. Proc Natl Acad Sci U S A 2021. [PMID: 33558236 DOI: 10.1073/pnas.2017937118/-/dcsupplemental] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
The number and activity of Cav1.2 channels in the cardiomyocyte sarcolemma tunes the magnitude of Ca2+-induced Ca2+ release and myocardial contraction. β-Adrenergic receptor (βAR) activation stimulates sarcolemmal insertion of CaV1.2. This supplements the preexisting sarcolemmal CaV1.2 population, forming large "superclusters" wherein neighboring channels undergo enhanced cooperative-gating behavior, amplifying Ca2+ influx and myocardial contractility. Here, we determine this stimulated insertion is fueled by an internal reserve of early and recycling endosome-localized, presynthesized CaV1.2 channels. βAR-activation decreased CaV1.2/endosome colocalization in ventricular myocytes, as it triggered "emptying" of endosomal CaV1.2 cargo into the t-tubule sarcolemma. We examined the rapid dynamics of this stimulated insertion process with live-myocyte imaging of channel trafficking, and discovered that CaV1.2 are often inserted into the sarcolemma as preformed, multichannel clusters. Similarly, entire clusters were removed from the sarcolemma during endocytosis, while in other cases, a more incremental process suggested removal of individual channels. The amplitude of the stimulated insertion response was doubled by coexpression of constitutively active Rab4a, halved by coexpression of dominant-negative Rab11a, and abolished by coexpression of dominant-negative mutant Rab4a. In ventricular myocytes, βAR-stimulated recycling of CaV1.2 was diminished by both nocodazole and latrunculin-A, suggesting an essential role of the cytoskeleton in this process. Functionally, cytoskeletal disruptors prevented βAR-activated Ca2+ current augmentation. Moreover, βAR-regulation of CaV1.2 was abolished when recycling was halted by coapplication of nocodazole and latrunculin-A. These findings reveal that βAR-stimulation triggers an on-demand boost in sarcolemmal CaV1.2 abundance via targeted Rab4a- and Rab11a-dependent insertion of channels that is essential for βAR-regulation of cardiac CaV1.2.
Collapse
Affiliation(s)
- Silvia G Del Villar
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Taylor L Voelker
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Maartje Westhoff
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Gopireddy R Reddy
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| | - Heather C Spooner
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Manuel F Navedo
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| | - Eamonn J Dickson
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616;
| |
Collapse
|
27
|
Del Villar SG, Voelker TL, Westhoff M, Reddy GR, Spooner HC, Navedo MF, Dickson EJ, Dixon RE. β-Adrenergic control of sarcolemmal Ca V1.2 abundance by small GTPase Rab proteins. Proc Natl Acad Sci U S A 2021; 118:e2017937118. [PMID: 33558236 PMCID: PMC7896340 DOI: 10.1073/pnas.2017937118] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The number and activity of Cav1.2 channels in the cardiomyocyte sarcolemma tunes the magnitude of Ca2+-induced Ca2+ release and myocardial contraction. β-Adrenergic receptor (βAR) activation stimulates sarcolemmal insertion of CaV1.2. This supplements the preexisting sarcolemmal CaV1.2 population, forming large "superclusters" wherein neighboring channels undergo enhanced cooperative-gating behavior, amplifying Ca2+ influx and myocardial contractility. Here, we determine this stimulated insertion is fueled by an internal reserve of early and recycling endosome-localized, presynthesized CaV1.2 channels. βAR-activation decreased CaV1.2/endosome colocalization in ventricular myocytes, as it triggered "emptying" of endosomal CaV1.2 cargo into the t-tubule sarcolemma. We examined the rapid dynamics of this stimulated insertion process with live-myocyte imaging of channel trafficking, and discovered that CaV1.2 are often inserted into the sarcolemma as preformed, multichannel clusters. Similarly, entire clusters were removed from the sarcolemma during endocytosis, while in other cases, a more incremental process suggested removal of individual channels. The amplitude of the stimulated insertion response was doubled by coexpression of constitutively active Rab4a, halved by coexpression of dominant-negative Rab11a, and abolished by coexpression of dominant-negative mutant Rab4a. In ventricular myocytes, βAR-stimulated recycling of CaV1.2 was diminished by both nocodazole and latrunculin-A, suggesting an essential role of the cytoskeleton in this process. Functionally, cytoskeletal disruptors prevented βAR-activated Ca2+ current augmentation. Moreover, βAR-regulation of CaV1.2 was abolished when recycling was halted by coapplication of nocodazole and latrunculin-A. These findings reveal that βAR-stimulation triggers an on-demand boost in sarcolemmal CaV1.2 abundance via targeted Rab4a- and Rab11a-dependent insertion of channels that is essential for βAR-regulation of cardiac CaV1.2.
Collapse
Affiliation(s)
- Silvia G Del Villar
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Taylor L Voelker
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Maartje Westhoff
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Gopireddy R Reddy
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| | - Heather C Spooner
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Manuel F Navedo
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| | - Eamonn J Dickson
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| | - Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616;
| |
Collapse
|
28
|
Xiao S, Shimura D, Baum R, Hernandez DM, Agvanian S, Nagaoka Y, Katsumata M, Lampe PD, Kleber AG, Hong T, Shaw RM. Auxiliary trafficking subunit GJA1-20k protects connexin-43 from degradation and limits ventricular arrhythmias. J Clin Invest 2021; 130:4858-4870. [PMID: 32525845 DOI: 10.1172/jci134682] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 06/03/2020] [Indexed: 12/31/2022] Open
Abstract
Connexin-43 (Cx43) gap junctions provide intercellular coupling, which ensures rapid action potential propagation and synchronized heart contraction. Alterations in Cx43 localization and reductions in gap junction coupling occur in failing hearts, contributing to ventricular arrhythmias and sudden cardiac death. Recent reports have found that an internally translated Cx43 isoform, GJA1-20k, is an auxiliary subunit for the trafficking of Cx43 in heterologous expression systems. Here, we have created a mouse model by using CRISPR technology to mutate a single internal translation initiation site in Cx43 (M213L mutation), which generates full-length Cx43, but not GJA1-20k. We found that GJA1M213L/M213L mice had severely abnormal electrocardiograms despite preserved contractile function, reduced total Cx43, and reduced gap junctions, and they died suddenly at 2 to 4 weeks of age. Heterozygous GJA1M213L/WT mice survived to adulthood with increased ventricular ectopy. Biochemical experiments indicated that cytoplasmic Cx43 had a half-life that was 50% shorter than membrane-associated Cx43. Without GJA1-20k, poorly trafficked Cx43 was degraded. The data support that GJA1-20k, an endogenous entity translated independently of Cx43, is critical for Cx43 gap junction trafficking, maintenance of Cx43 protein, and normal electrical function of the mammalian heart.
Collapse
Affiliation(s)
- Shaohua Xiao
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Neurology, UCLA, Los Angeles, California, USA
| | - Daisuke Shimura
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - Rachel Baum
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Diana M Hernandez
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sosse Agvanian
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yoshiko Nagaoka
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Makoto Katsumata
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Paul D Lampe
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Andre G Kleber
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - TingTing Hong
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robin M Shaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
29
|
Picoli C, Soleilhac E, Journet A, Barette C, Comte M, Giaume C, Mouthon F, Fauvarque MO, Charvériat M. High-Content Screening Identifies New Inhibitors of Connexin 43 Gap Junctions. Assay Drug Dev Technol 2020; 17:240-248. [PMID: 31314551 DOI: 10.1089/adt.2019.927] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Gap junctions (GJs) are dynamic structures composed of hexamers of connexins (Cxs), a class of transmembrane proteins enabling channel-mediated direct intercellular communication through cell-cell diffusion of ions and small metabolites. In defined conditions, Cxs also work as hemichannels allowing exchanges between the cytoplasm and the extracellular medium. The most common GJ channel is formed by connexin 43 (Cx43) and plays an important role in physiological and pathological processes in excitable tissues, such as heart and brain. Hence, Cx43 has been largely envisioned as a new therapeutic target in cancer, neurological and psychiatric indications, or cardiovascular diseases. Identifying new pharmacological inhibitors of Cx43 GJs with different mechanisms of action and from diverse chemical classes is thus highly challenging. We present here a high-content screening method, based on the evaluation of fluorescent dye transfer rates between adjacent cells to monitor the function of GJs in U251 glioblastoma cells expressing high levels of Cx43. This assay was validated using well-described pharmacological GJ inhibitors such as mefloquine. The method was adapted to screen a library of 1,280 Food and Drug Administration- and European Medicines Agency-approved drugs that led to the selection of both known and new inhibitors of GJ channel function. We further focused on a specific class of microtubule-targeting agents, confirming that a proper tubulin network is required for functional Cx43 GJ channels.
Collapse
Affiliation(s)
| | - Emmanuelle Soleilhac
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | - Agnès Journet
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | - Caroline Barette
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | - Marjorie Comte
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | | | | | - Marie-Odile Fauvarque
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | | |
Collapse
|
30
|
Dynamic UTR Usage Regulates Alternative Translation to Modulate Gap Junction Formation during Stress and Aging. Cell Rep 2020; 27:2737-2747.e5. [PMID: 31141695 PMCID: PMC6857847 DOI: 10.1016/j.celrep.2019.04.114] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/20/2019] [Accepted: 04/29/2019] [Indexed: 11/22/2022] Open
Abstract
Connexin43 (Cx43; gene name GJA1) is the most ubiquitously expressed gap junction protein, and understanding of its regulation largely falls under transcription and post-translational modification. In addition to Cx43, Gja1 mRNA encodes internally translated isoforms regulating gap junction formation, whose expression is modulated by TGF-β. Here, using RLM-RACE, we identify distinct Gja1 transcripts differing only in 5′ UTR length, of which two are upregulated during TGF-β exposure and hypoxia. Introduction of these transcripts into Gja1−/− cells phenocopies the response of Gja1 to TGF-β with reduced internal translation initiation. Inhibiting pathways downstream of TGF-β selectively regulates levels of Gja1 transcript isoforms and translation products. Reporter assays reveal enhanced translation of full-length Cx43 from shorter Gja1 5′ UTR isoforms. We also observe a correlation among UTR selection, translation, and reduced gap junction formation in aged heart tissue. These data elucidate a relationship between transcript isoform expression and translation initiation regulating intercellular communication. Connexin43 gap junctions enable direct intercellular communication facilitating action potential propagation. Internal translation of connexin43 mRNA generates the truncated isoform GJA1–20k, which promotes gap junction formation. During aging, Zeitz et al. find that activation of stress-response pathways shortens connexin43 mRNA UTRs to limit GJA1–20k translation coincident with gap junction loss.
Collapse
|
31
|
Fan J, Chen Y, Yang D, Shen J, Guo X. Multi-walled carbon nanotubes induce IL-1β secretion by activating hemichannels-mediated ATP release in THP-1 macrophages. Nanotoxicology 2020; 14:929-946. [PMID: 32538272 DOI: 10.1080/17435390.2020.1777476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Multi-walled carbon nanotubes (MWCNTs) are known to induce pulmonary inflammatory effects through stimulating pro-inflammatory cytokine secretion from alveolar macrophages. Despite extensive studies on MWCNTs' pro-inflammatory reactivity, the understanding of molecular mechanisms involved is still incomplete. In this study, we investigated hemichannel's involvement in MWCNTs-induced macrophage IL-1β release. Our results showed that the unmodified and COOH MWCNTs could induce ATP release and ATP-P2X7R axis-dependent IL-1β secretion from THP-1 macrophages. By using various inhibitors, we confirmed that the MWCNTs-induced ATP release was primarily through hemichannels. EtBr dye uptake assay detected significant hemichannels opening in MWCNTs exposed THP-1 macrophages. Inhibition of hemichannels by CBX, 43Gap27, or 10Panx1 pretreatment results in decreased ATP and IL-1β release. The addition of ATP restored the reduced IL-1β secretion level from hemichannel inhibition. We also confirmed with five other types of MWCNTs that the induction of hemichannels by MWCNTs strongly correlates with their capacity to induce IL-1β secretion. Taken together, we conclude that hemichannels-mediated ATP release and subsequent NLRP3 inflammasome activation through P2X7R may be one mechanism by which MWCNTs induce macrophage IL-1β secretion. Our findings may provide a novel molecular mechanism for MWCNTs induced IL-1β secretion.
Collapse
Affiliation(s)
- Jingpu Fan
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China
| | - Yiyong Chen
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China
| | - Di Yang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China
| | - Jie Shen
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China
| |
Collapse
|
32
|
Liu S, Hu C, Luo Y, Yao K. Genome-wide DNA methylation profiles may reveal new possible epigenetic pathogenesis of sporadic congenital cataract. Epigenomics 2020; 12:771-788. [PMID: 32516005 DOI: 10.2217/epi-2019-0254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: To investigate the possible epigenetic pathogenesis of sporadic congenital cataract. Materials & methods: We conducted whole genome bisulfite sequencing on peripheral blood from sporadic binocular or monocular congenital cataract patients and cataract-free participants. Results: We found massive differentially methylated regions within the whole genomes between any two groups. Meanwhile, we identified five genes (ACTN4, ACTG1, TUBA1A, TUBA1C, TUBB4B) for the binocular and control groups and TUBA1A for the monocular and control groups as the core differentially methylated region-related genes. The proteins encoded by these core genes are involved in building cytoskeleton and intercellular junctions. Conclusion: Changes in the methylation levels of core genes may disturb the function of cytoskeleton and intercellular junctions, eventually leading to sporadic congenital cataract.
Collapse
Affiliation(s)
- Siyu Liu
- Eye Center of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310031, PR China.,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province 310031, PR China
| | - Chenyang Hu
- Eye Center of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310031, PR China.,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province 310031, PR China
| | - Yueqiu Luo
- Eye Center of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310031, PR China.,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province 310031, PR China
| | - Ke Yao
- Eye Center of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310031, PR China.,Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang Province 310031, PR China
| |
Collapse
|
33
|
Shoaito H, Chauveau S, Gosseaume C, Bourguet W, Vigouroux C, Vatier C, Pienkowski C, Fournier T, Degrelle SA. Peroxisome proliferator-activated receptor gamma-ligand-binding domain mutations associated with familial partial lipodystrophy type 3 disrupt human trophoblast fusion and fibroblast migration. J Cell Mol Med 2020; 24:7660-7669. [PMID: 32519441 PMCID: PMC7339198 DOI: 10.1111/jcmm.15401] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
The transcription factor peroxisome proliferator‐activated receptor gamma (PPARG) is essential for placental development, and alterations in its expression and/or activity are associated with human placental pathologies such as pre‐eclampsia or IUGR. However, the molecular regulation of PPARG in cytotrophoblast differentiation and in the underlying mesenchyme remains poorly understood. Our main goal was to study the impact of mutations in the ligand‐binding domain (LBD) of the PPARG gene on cytotrophoblast fusion (PPARGE352Q) and on fibroblast cell migration (PPARGR262G/PPARGL319X). Our results showed that, compared to cells with reconstituted PPARGWT, transfection with PPARGE352Q led to significantly lower PPARG activity and lower restoration of trophoblast fusion. Likewise, compared to PPARGWT fibroblasts, PPARGR262G/PPARGL319X fibroblasts demonstrated significantly inhibited cell migration. In conclusion, we report that single missense or nonsense mutations in the LBD of PPARG significantly inhibit cell fusion and migration processes.
Collapse
Affiliation(s)
- Hussein Shoaito
- INSERM, UMR-S1139 (3PHM), Université de Paris, Paris, France
| | - Sabine Chauveau
- INSERM, UMR-S1139 (3PHM), Université de Paris, Paris, France.,Laboratoire ICARE, Biopôle Clermont-Limagne, Saint-Beauzire, France
| | - Camille Gosseaume
- Inserm UMR-S938, Department of Endocrinology, Diabetology and Reproductive Endocrinology, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition (ICAN), AP-HP, Saint-Antoine Hospital, National Reference Centre of Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Sorbonne Université, Paris, France
| | - William Bourguet
- INSERM, CNRS, Centre de Biochimie Structurale (CBS), Université de Montpellier, Montpellier, France
| | - Corinne Vigouroux
- Inserm UMR-S938, Department of Endocrinology, Diabetology and Reproductive Endocrinology, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition (ICAN), AP-HP, Saint-Antoine Hospital, National Reference Centre of Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Sorbonne Université, Paris, France.,Department of Molecular Biology and Genetics, AP-HP, Saint-Antoine Hospital, Paris, France
| | - Camille Vatier
- Inserm UMR-S938, Department of Endocrinology, Diabetology and Reproductive Endocrinology, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition (ICAN), AP-HP, Saint-Antoine Hospital, National Reference Centre of Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Sorbonne Université, Paris, France
| | - Catherine Pienkowski
- Endocrinology Unit, Reference Centre for Rare Gynecologic Diseases, Toulouse, France
| | - Thierry Fournier
- INSERM, UMR-S1139 (3PHM), Université de Paris, Paris, France.,PremUp Foundation, Paris, France
| | - Séverine A Degrelle
- INSERM, UMR-S1139 (3PHM), Université de Paris, Paris, France.,PremUp Foundation, Paris, France.,Inovarion, Paris, France
| |
Collapse
|
34
|
Melgari D, Barbier C, Dilanian G, Rücker-Martin C, Doisne N, Coulombe A, Hatem SN, Balse E. Microtubule polymerization state and clathrin-dependent internalization regulate dynamics of cardiac potassium channel: Microtubule and clathrin control of K V1.5 channel. J Mol Cell Cardiol 2020; 144:127-139. [PMID: 32445844 DOI: 10.1016/j.yjmcc.2020.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/19/2022]
Abstract
Ion channel trafficking powerfully influences cardiac electrical activity as it regulates the number of available channels at the plasma membrane. Studies have largely focused on identifying the molecular determinants of the trafficking of the atria-specific KV1.5 channel, the molecular basis of the ultra-rapid delayed rectifier current IKur. Besides, regulated KV1.5 channel recycling upon changes in homeostatic state and mechanical constraints in native cardiomyocytes has been well documented. Here, using cutting-edge imaging in live myocytes, we investigated the dynamics of this channel in the plasma membrane. We demonstrate that the clathrin pathway is a major regulator of the functional expression of KV1.5 channels in atrial myocytes, with the microtubule network as the prominent organizer of KV1.5 transport within the membrane. Both clathrin blockade and microtubule disruption result in channel clusterization with reduced membrane mobility and internalization, whereas disassembly of the actin cytoskeleton does not. Mobile KV1.5 channels are associated with the microtubule plus-end tracking protein EB1 whereas static KV1.5 clusters are associated with stable acetylated microtubules. In human biopsies from patients in atrial fibrillation associated with atrial remodeling, drastic modifications in the trafficking balance occurs together with alteration in microtubule polymerization state resulting in modest reduced endocytosis and increased recycling. Consequently, hallmark of atrial KV1.5 dynamics within the membrane is clathrin- and microtubule- dependent. During atrial remodeling, predominance of anterograde trafficking activity over retrograde trafficking could result in accumulation ok KV1.5 channels in the plasma membrane.
Collapse
Affiliation(s)
- Dario Melgari
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Camille Barbier
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Gilles Dilanian
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | | | - Nicolas Doisne
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Alain Coulombe
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Stéphane N Hatem
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France; Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Elise Balse
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France.
| |
Collapse
|
35
|
Himelman E, Lillo MA, Nouet J, Gonzalez JP, Zhao Q, Xie LH, Li H, Liu T, Wehrens XH, Lampe PD, Fishman GI, Shirokova N, Contreras JE, Fraidenraich D. Prevention of connexin-43 remodeling protects against Duchenne muscular dystrophy cardiomyopathy. J Clin Invest 2020; 130:1713-1727. [PMID: 31910160 PMCID: PMC7108916 DOI: 10.1172/jci128190] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
Aberrant expression of the cardiac gap junction protein connexin-43 (Cx43) has been suggested as playing a role in the development of cardiac disease in the mdx mouse model of Duchenne muscular dystrophy (DMD); however, a mechanistic understanding of this association is lacking. Here, we identified a reduction of phosphorylation of Cx43 serines S325/S328/S330 in human and mouse DMD hearts. We hypothesized that hypophosphorylation of Cx43 serine-triplet triggers pathological Cx43 redistribution to the lateral sides of cardiomyocytes (remodeling). Therefore, we generated knockin mdx mice in which the Cx43 serine-triplet was replaced with either phospho-mimicking glutamic acids (mdxS3E) or nonphosphorylatable alanines (mdxS3A). The mdxS3E, but not mdxS3A, mice were resistant to Cx43 remodeling, with a corresponding reduction of Cx43 hemichannel activity. MdxS3E cardiomyocytes displayed improved intracellular Ca2+ signaling and a reduction of NADPH oxidase 2 (NOX2)/ROS production. Furthermore, mdxS3E mice were protected against inducible arrhythmias, related lethality, and the development of cardiomyopathy. Inhibition of microtubule polymerization by colchicine reduced both NOX2/ROS and oxidized CaMKII, increased S325/S328/S330 phosphorylation, and prevented Cx43 remodeling in mdx hearts. Together, these results demonstrate a mechanism of dystrophic Cx43 remodeling and suggest that targeting Cx43 may be a therapeutic strategy for preventing heart dysfunction and arrhythmias in DMD patients.
Collapse
Affiliation(s)
| | | | - Julie Nouet
- Department of Cell Biology and Molecular Medicine
| | | | - Qingshi Zhao
- Department of Cell Biology and Molecular Medicine
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine
| | - Hong Li
- Center for Advanced Proteomics Research, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Xander H.T. Wehrens
- Department of Molecular Physiology and Biophysics, Medicine, Neuroscience, and Pediatrics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, USA
| | - Paul D. Lampe
- Fred Hutchinson Cancer Research Center, Translational Research Program, Public Health Sciences Division, Seattle, Washington, USA
| | - Glenn I. Fishman
- Leon H. Charney Division of Cardiology, New York University Langone Health, New York, New York, USA
| | | | | | | |
Collapse
|
36
|
Epifantseva I, Xiao S, Baum RE, Kléber AG, Hong T, Shaw RM. An Alternatively Translated Connexin 43 Isoform, GJA1-11k, Localizes to the Nucleus and Can Inhibit Cell Cycle Progression. Biomolecules 2020; 10:biom10030473. [PMID: 32244859 PMCID: PMC7175147 DOI: 10.3390/biom10030473] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/10/2020] [Accepted: 03/15/2020] [Indexed: 12/14/2022] Open
Abstract
Connexin 43 (Cx43) is a gap junction protein that assembles at the cell border to form intercellular gap junction (GJ) channels which allow for cell-cell communication by facilitating the rapid transmission of ions and other small molecules between adjacent cells. Non-canonical roles of Cx43, and specifically its C-terminal domain, have been identified in the regulation of Cx43 trafficking, mitochondrial preconditioning, cell proliferation, and tumor formation, yet the mechanisms are still being explored. It was recently identified that up to six truncated isoforms of Cx43 are endogenously produced via alternative translation from internal start codons in addition to full length Cx43, all from the same mRNA produced by the gene GJA1. GJA1-11k, the 11kDa alternatively translated isoform of Cx43, does not have a known role in the formation of gap junction channels, and little is known about its function. Here, we report that over expressed GJA1-11k, unlike the other five truncated isoforms, preferentially localizes to the nucleus in HEK293FT cells and suppresses cell growth by limiting cell cycle progression from the G0/G1 phase to the S phase. Furthermore, these functions are independent of the channel-forming full-length Cx43 isoform. Understanding the apparently unique role of GJA1-11k and its generation in cell cycle regulation may uncover a new target for affecting cell growth in multiple disease models.
Collapse
Affiliation(s)
- Irina Epifantseva
- Smidt Heart Institute, Graduate Program in Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (I.E.); (S.X.); (R.E.B.); (T.H.)
| | - Shaohua Xiao
- Smidt Heart Institute, Graduate Program in Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (I.E.); (S.X.); (R.E.B.); (T.H.)
| | - Rachel E. Baum
- Smidt Heart Institute, Graduate Program in Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (I.E.); (S.X.); (R.E.B.); (T.H.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - André G. Kléber
- Department of Pathology, Beth Israel & Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - TingTing Hong
- Smidt Heart Institute, Graduate Program in Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (I.E.); (S.X.); (R.E.B.); (T.H.)
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90048, USA
| | - Robin M. Shaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
- Correspondence: ; Tel.: +(801)-587-5845
| |
Collapse
|
37
|
Kumar R, Saha S, Sinha B. Cell spread area and traction forces determine myosin-II-based cortex thickness regulation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2019; 1866:118516. [PMID: 31348954 PMCID: PMC7617199 DOI: 10.1016/j.bbamcr.2019.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/09/2019] [Accepted: 07/18/2019] [Indexed: 12/31/2022]
Abstract
Actomyosin network under the plasma membrane of cells forms a cortical layer that regulates cellular deformations during different processes. What regulates the cortex? Characterized by its thickness, it is believed to be regulated by actin dynamics, filament-length regulators and myosin motor proteins. However, its regulation by cellular morphology (e.g. cell spread area) or mechanical microenvironment (e.g. substrate stiffness) has remained largely unexplored. In this study, super- and high-resolution imaging of actin in CHO cells demonstrates that at high spread areas (>450 μm2), the cortex is thinner, better separated as layers, and sensitive to deactivation of myosin II motors or reduction of substrate stiffness (and traction forces). In less spread cells (<400 μm2) such perturbations do not elicit a response. Myosin IIA's mechanosensing is limited here due to its lowered actin-bound fraction and higher turnover rate. Cofilin, in line with its competitive inhibitory role, is found to be overexpressed in these cells. To establish the causal relation, we initiate a spread area drop by de-adhesion and find enhanced actin dynamics and fragmentation along with oscillations and increase in thickness. This is more correlated to the reduction of traction forces than the endocytosis-based reduction in cell volume. Cortex thickness control by spread area is also found be true during differentiation of THP-1 monocytes to macrophages. Thus, we propose that spread area regulates cortex and its thickness by traction-based mechanosensing of myosin II.
Collapse
Affiliation(s)
- Rinku Kumar
- Dept. of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Sajjita Saha
- Dept. of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Bidisha Sinha
- Dept. of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India.
| |
Collapse
|
38
|
Ghazizadeh Z, Kiviniemi T, Olafsson S, Plotnick D, Beerens ME, Zhang K, Gillon L, Steinbaugh MJ, Barrera V, Sui SH, Werdich AA, Kapur S, Eranti A, Gunn J, Jalkanen J, Airaksinen J, Kleber AG, Hollmén M, MacRae CA. Metastable Atrial State Underlies the Primary Genetic Substrate for MYL4 Mutation-Associated Atrial Fibrillation. Circulation 2019; 141:301-312. [PMID: 31735076 DOI: 10.1161/circulationaha.119.044268] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common clinical arrhythmia and is associated with heart failure, stroke, and increased mortality. The myocardial substrate for AF is poorly understood because of limited access to primary human tissue and mechanistic questions around existing in vitro or in vivo models. METHODS Using an MYH6:mCherry knock-in reporter line, we developed a protocol to generate and highly purify human pluripotent stem cell-derived cardiomyocytes displaying physiological and molecular characteristics of atrial cells. We modeled human MYL4 mutants, one of the few definitive genetic causes of AF. To explore non-cell-autonomous components of AF substrate, we also created a zebrafish Myl4 knockout model, which exhibited molecular, cellular, and physiologic abnormalities that parallel those in humans bearing the cognate mutations. RESULTS There was evidence of increased retinoic acid signaling in both human embryonic stem cells and zebrafish mutant models, as well as abnormal expression and localization of cytoskeletal proteins, and loss of intracellular nicotinamide adenine dinucleotide and nicotinamide adenine dinucleotide + hydrogen. To identify potentially druggable proximate mechanisms, we performed a chemical suppressor screen integrating multiple human cellular and zebrafish in vivo endpoints. This screen identified Cx43 (connexin 43) hemichannel blockade as a robust suppressor of the abnormal phenotypes in both models of MYL4 (myosin light chain 4)-related atrial cardiomyopathy. Immunofluorescence and coimmunoprecipitation studies revealed an interaction between MYL4 and Cx43 with altered localization of Cx43 hemichannels to the lateral membrane in MYL4 mutants, as well as in atrial biopsies from unselected forms of human AF. The membrane fraction from MYL4-/- human embryonic stem cell derived atrial cells demonstrated increased phospho-Cx43, which was further accentuated by retinoic acid treatment and by the presence of risk alleles at the Pitx2 locus. PKC (protein kinase C) was induced by retinoic acid, and PKC inhibition also rescued the abnormal phenotypes in the atrial cardiomyopathy models. CONCLUSIONS These data establish a mechanistic link between the transcriptional, metabolic and electrical pathways previously implicated in AF substrate and suggest novel avenues for the prevention or therapy of this common arrhythmia.
Collapse
Affiliation(s)
- Zaniar Ghazizadeh
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Tuomas Kiviniemi
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Sigurast Olafsson
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David Plotnick
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Manu E Beerens
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kun Zhang
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Leah Gillon
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Victor Barrera
- Chan School of Public Health, Boston, MA (M.J.S., V.B., S.H.S.)
| | - Shannan Ho Sui
- Chan School of Public Health, Boston, MA (M.J.S., V.B., S.H.S.)
| | - Andreas A Werdich
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sunil Kapur
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Antti Eranti
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Jarmo Gunn
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Juho Jalkanen
- Medicity Research Laboratories (J.J., M.H.), Harvard T.H
| | - Juhani Airaksinen
- Heart Center, Turku University Hospital (T.K., A.E., J.G., J.A.), Harvard T.H
- University of Turku, Finland (T.K., A.E., J.G., J.A.). Harvard T.H
| | - Andre G Kleber
- Department of Pathology, Beth Israel Deaconess Medical Center Harvard Medical School, Boston, MA (A.G.K.)
| | - Maija Hollmén
- Medicity Research Laboratories (J.J., M.H.), Harvard T.H
| | - Calum A MacRae
- Cardiovascular Medicine Division (Z.G., T.K., S.O., D.P., M.E.B., K.Z., L.G., A.A.W., S.K., C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Genetics and Network Medicine Divisions (C.A.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Harvard Stem Cell Institute, Boston, MA (C.A.M.)
| |
Collapse
|
39
|
Schultz F, Swiatlowska P, Alvarez-Laviada A, Sanchez-Alonso JL, Song Q, de Vries AAF, Pijnappels DA, Ongstad E, Braga VMM, Entcheva E, Gourdie RG, Miragoli M, Gorelik J. Cardiomyocyte-myofibroblast contact dynamism is modulated by connexin-43. FASEB J 2019; 33:10453-10468. [PMID: 31253057 PMCID: PMC6704460 DOI: 10.1096/fj.201802740rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Healthy cardiomyocytes are electrically coupled at the intercalated discs by gap junctions. In infarcted hearts, adverse gap-junctional remodeling occurs in the border zone, where cardiomyocytes are chemically and electrically influenced by myofibroblasts. The physical movement of these contacts remains unquantified. Using scanning ion conductance microscopy, we show that intercellular contacts between cardiomyocytes and myofibroblasts are highly dynamic, mainly owing to the edge dynamics (lamellipodia) of the myofibroblasts. Decreasing the amount of functional connexin-43 (Cx43) at the membrane through Cx43 silencing, suppression of Cx43 trafficking, or hypoxia-induced Cx43 internalization attenuates heterocellular contact dynamism. However, we found decreased dynamism and stabilized membrane contacts when cellular coupling was strengthened using 4-phenylbutyrate (4PB). Fluorescent-dye transfer between cells showed that the extent of functional coupling between the 2 cell types correlated with contact dynamism. Intercellular calcein transfer from myofibroblasts to cardiomyocytes is reduced after myofibroblast-specific Cx43 down-regulation. Conversely, 4PB-treated myofibroblasts increased their functional coupling to cardiomyocytes. Consistent with lamellipodia-mediated contacts, latrunculin-B decreases dynamism, lowers physical communication between heterocellular pairs, and reduces Cx43 intensity in contact regions. Our data show that heterocellular cardiomyocyte-myofibroblast contacts exhibit high dynamism. Therefore, Cx43 is a potential target for prevention of aberrant cardiomyocyte coupling and myofibroblast proliferation in the infarct border zone.-Schultz, F., Swiatlowska, P., Alvarez-Laviada, A., Sanchez-Alonso, J. L., Song, Q., de Vries, A. A. F., Pijnappels, D. A., Ongstad, E., Braga, V. M. M., Entcheva, E., Gourdie, R. G., Miragoli, M., Gorelik, J. Cardiomyocyte-myofibroblast contact dynamism is modulated by connexin-43.
Collapse
Affiliation(s)
- Francisca Schultz
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pamela Swiatlowska
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | | | - Qianqian Song
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Daniël A. Pijnappels
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emily Ongstad
- Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, Virginia, USA
| | - Vania M. M. Braga
- Department of Respiratory Sciences, Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Robert G. Gourdie
- Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, Virginia, USA
| | - Michele Miragoli
- Humanitas Clinical and Research Center, Milan, Italy;,Department of Medicine and Surgery, University of Parma, Parma, Italy,Correspondence: Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43124 Parma, Italy. E-mail:
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, London, United Kingdom;,Correspondence: National Heart and Lung Institute, 4th Floor, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Campus, Du Cane Rd., London W12 0NN, United Kingdom. E-mail:
| |
Collapse
|
40
|
Du J, Fan Y, Guo Z, Wang Y, Zheng X, Huang C, Liang B, Gao L, Cao Y, Chen Y, Zhang X, Li L, Xu L, Wu C, Weitz DA, Feng X. Compression Generated by a 3D Supracellular Actomyosin Cortex Promotes Embryonic Stem Cell Colony Growth and Expression of Nanog and Oct4. Cell Syst 2019; 9:214-220.e5. [DOI: 10.1016/j.cels.2019.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 12/19/2018] [Accepted: 05/22/2019] [Indexed: 10/26/2022]
|
41
|
Usui Y, Aramaki T, Kondo S, Watanabe M. The minimal gap-junction network among melanophores and xanthophores required for stripe-pattern formation in zebrafish. Development 2019; 146:dev.181065. [DOI: 10.1242/dev.181065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/24/2019] [Indexed: 12/16/2022]
Abstract
Connexin39.4 (Cx39.4) and Connexin41.8 (Cx41.8), two gap-junction proteins expressed in both melanophores and xanthophores, are critical for the intercellular communication among pigment cells that is necessary for generating the stripe pigment pattern of zebrafish. We previously characterized the gap-junction properties of Cx39.4 and Cx41.8, but how these proteins contribute to stripe formation remains unclear; this is because distinct types of connexins potentially form heteromeric gap junctions, which precludes accurate elucidation of individual connexin functions in vivo. Here, by arranging Cx39.4 and Cx41.8 expression in pigment cells, we identified the simplest gap-junction network required for stripe generation: Cx39.4 expression in melanophores is required but expression in xanthophores is not necessary for stripe patterning, whereas Cx41.8 expression in xanthophores is sufficient for the patterning, and Cx41.8 expression in melanophores might stabilize the stripes. Moreover, patch-clamp recordings revealed that Cx39.4 gap junctions exhibit spermidine-dependent rectification property. Our results suggest that Cx39.4 facilitates the critical cell-cell interactions between melanophores and xanthophores that mediate a unidirectional activation-signal transfer from xanthophores to melanophores, which is essential for melanophore survival.
Collapse
Affiliation(s)
- Yuu Usui
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihiro Aramaki
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shigeru Kondo
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
- CREST, Japan Science and Technology Agency, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masakatsu Watanabe
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
42
|
Gourdie RG. The Cardiac Gap Junction has Discrete Functions in Electrotonic and Ephaptic Coupling. Anat Rec (Hoboken) 2018; 302:93-100. [PMID: 30565418 DOI: 10.1002/ar.24036] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/10/2018] [Accepted: 07/17/2018] [Indexed: 12/31/2022]
Abstract
Connexin43-formed gap junctions have long been thought to contribute to cardiac conduction in the mammalian ventricle by providing direct electrotonic connectivity between the cytoplasms of neighboring cardiomyocytes. However, accumulating data from studies of non-mammalian hearts, Connexin 43 (Cx43) knockout mice and human Cx43 mutations have raised questions as to whether gap junctions are the sole means by which electrical coupling between cardiomyocytes is accomplished. Computational and experimental work over the last decade have indicated that intercellular propagation of action potentials in the heart may involve both electrotonic and ephaptic contributions-in what has been dubbed "mixed-mode" conduction. Interestingly, the Cx43 gap junction may provide a common structural platform in mammals that facilitates the operation of these two mechanisms. In addition to gap junction channels functioning in an electrotonic role, the perinexus region at the edge of gap junctions may be provide a niche for voltage-gated sodium channels from neighboring cells to be in sufficiently close proximity to enable ephaptic transmission of action potential. A novel role has recently been identified in this potential ephaptic mechanism for inter-membrane adhesion mediated by the beta subunit (beta1/Scn1b) of the sodium channel. The new perspective of the operational redundancy that is built into cardiac electrical connectivity could provide new understanding of arrhythmia mechanisms and holds the promise for new approach to anti-arrhythmic therapy. Anat Rec, 302:93-100, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Robert G Gourdie
- Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, Virginia, 24016.,Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, Virginia, 24016.,Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, Virginia, 24061
| |
Collapse
|
43
|
Basheer WA, Fu Y, Shimura D, Xiao S, Agvanian S, Hernandez DM, Hitzeman TC, Hong T, Shaw RM. Stress response protein GJA1-20k promotes mitochondrial biogenesis, metabolic quiescence, and cardioprotection against ischemia/reperfusion injury. JCI Insight 2018; 3:121900. [PMID: 30333316 DOI: 10.1172/jci.insight.121900] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/11/2018] [Indexed: 01/23/2023] Open
Abstract
Connexin 43 (Cx43), a product of the GJA1 gene, is a gap junction protein facilitating intercellular communication between cardiomyocytes. Cx43 protects the heart from ischemic injury by mechanisms that are not well understood. GJA1 mRNA can undergo alternative translation, generating smaller isoforms in the heart, with GJA1-20k being the most abundant. Here, we report that ischemic and ischemia/reperfusion (I/R) injuries upregulate endogenous GJA1-20k protein in the heart, which targets to cardiac mitochondria and associates with the outer mitochondrial membrane. Exploring the functional consequence of increased GJA1-20k, we found that AAV9-mediated gene transfer of GJA1-20k in mouse hearts increases mitochondrial biogenesis while reducing mitochondrial membrane potential, respiration, and ROS production. By doing so, GJA1-20k promotes a protective mitochondrial phenotype, as seen with ischemic preconditioning (IPC), which also increases endogenous GJA1-20k in heart lysates and mitochondrial fractions. As a result, AAV9-GJA1-20k pretreatment reduces myocardial infarct size in mouse hearts subjected to in vivo ischemic injury or ex vivo I/R injury, similar to an IPC-induced cardioprotective effect. In conclusion, GJA1-20k is an endogenous stress response protein that induces mitochondrial biogenesis and metabolic hibernation, preconditioning the heart against I/R insults. Introduction of exogenous GJA1-20k is a putative therapeutic strategy for patients undergoing anticipated ischemic injury.
Collapse
Affiliation(s)
- Wassim A Basheer
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ying Fu
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daisuke Shimura
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shaohua Xiao
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sosse Agvanian
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Diana M Hernandez
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tara C Hitzeman
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - TingTing Hong
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, California
| | - Robin M Shaw
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, California
| |
Collapse
|
44
|
Manring HR, Dorn LE, Ex-Willey A, Accornero F, Ackermann MA. At the heart of inter- and intracellular signaling: the intercalated disc. Biophys Rev 2018; 10:961-971. [PMID: 29876873 PMCID: PMC6082301 DOI: 10.1007/s12551-018-0430-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 05/22/2018] [Indexed: 12/17/2022] Open
Abstract
Proper cardiac function requires the synchronous mechanical and electrical coupling of individual cardiomyocytes. The intercalated disc (ID) mediates coupling of neighboring myocytes through intercellular signaling. Intercellular communication is highly regulated via intracellular signaling, and signaling pathways originating from the ID control cardiomyocyte remodeling and function. Herein, we present an overview of the inter- and intracellular signaling that occurs at and originates from the intercalated disc in normal physiology and pathophysiology. This review highlights the importance of the intercalated disc as an integrator of signaling events regulating homeostasis and stress responses in the heart and the center of several pathophysiological processes mediating the development of cardiomyopathies.
Collapse
Affiliation(s)
- Heather R Manring
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Lisa E Dorn
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Aidan Ex-Willey
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| | - Maegen A Ackermann
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
45
|
Basheer WA, Shaw RM. Connexin 43 and CaV1.2 Ion Channel Trafficking in Healthy and Diseased Myocardium. Circ Arrhythm Electrophysiol 2018; 9:e001357. [PMID: 27266274 DOI: 10.1161/circep.115.001357] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 04/29/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Wassim A Basheer
- From the Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (W.A.B., R.M.S.); and Department of Medicine, University of California Los Angeles (R.M.S.)
| | - Robin M Shaw
- From the Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (W.A.B., R.M.S.); and Department of Medicine, University of California Los Angeles (R.M.S.).
| |
Collapse
|
46
|
Spagnol G, Trease AJ, Zheng L, Gutierrez M, Basu I, Sarmiento C, Moore G, Cervantes M, Sorgen PL. Connexin43 Carboxyl-Terminal Domain Directly Interacts with β-Catenin. Int J Mol Sci 2018; 19:ijms19061562. [PMID: 29882937 PMCID: PMC6032326 DOI: 10.3390/ijms19061562] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022] Open
Abstract
Activation of Wnt signaling induces Connexin43 (Cx43) expression via the transcriptional activity of β-catenin, and results in the enhanced accumulation of the Cx43 protein and the formation of gap junction channels. In response to Wnt signaling, β-catenin co-localizes with the Cx43 protein itself as part of a complex at the gap junction plaque. Work from several labs have also shown indirect evidence of this interaction via reciprocal co-immunoprecipitation. Our goal for the current study was to identify whether β-catenin directly interacts with Cx43, and if so, the location of that direct interaction. Identifying residues involved in direct protein⁻protein interaction is of importance when they are correlated to the phosphorylation of Cx43, as phosphorylation can modify the binding affinities of Cx43 regulatory protein partners. Therefore, combining the location of a protein partner interaction on Cx43 along with the phosphorylation pattern under different homeostatic and pathological conditions will be crucial information for any potential therapeutic intervention. Here, we identified that β-catenin directly interacts with the Cx43 carboxyl-terminal domain, and that this interaction would be inhibited by the Src phosphorylation of Cx43CT residues Y265 and Y313.
Collapse
Affiliation(s)
- Gaelle Spagnol
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Andrew J Trease
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Li Zheng
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Mirtha Gutierrez
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Ishika Basu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Cleofes Sarmiento
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Gabriella Moore
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Matthew Cervantes
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Paul L Sorgen
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
47
|
Aasen T, Johnstone S, Vidal-Brime L, Lynn KS, Koval M. Connexins: Synthesis, Post-Translational Modifications, and Trafficking in Health and Disease. Int J Mol Sci 2018; 19:ijms19051296. [PMID: 29701678 PMCID: PMC5983588 DOI: 10.3390/ijms19051296] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 02/06/2023] Open
Abstract
Connexins are tetraspan transmembrane proteins that form gap junctions and facilitate direct intercellular communication, a critical feature for the development, function, and homeostasis of tissues and organs. In addition, a growing number of gap junction-independent functions are being ascribed to these proteins. The connexin gene family is under extensive regulation at the transcriptional and post-transcriptional level, and undergoes numerous modifications at the protein level, including phosphorylation, which ultimately affects their trafficking, stability, and function. Here, we summarize these key regulatory events, with emphasis on how these affect connexin multifunctionality in health and disease.
Collapse
Affiliation(s)
- Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, 08035 Barcelona, Spain.
| | - Scott Johnstone
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VI 22908, USA.
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, UK.
| | - Laia Vidal-Brime
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, 08035 Barcelona, Spain.
| | - K Sabrina Lynn
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
48
|
James CC, Zeitz MJ, Calhoun PJ, Lamouille S, Smyth JW. Altered translation initiation of Gja1 limits gap junction formation during epithelial-mesenchymal transition. Mol Biol Cell 2018; 29:797-808. [PMID: 29467255 PMCID: PMC5905293 DOI: 10.1091/mbc.e17-06-0406] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is activated during development, wound healing, and pathologies including fibrosis and cancer metastasis. Hallmarks of EMT are remodeling of intercellular junctions and adhesion proteins, including gap junctions. The GJA1 mRNA transcript encoding the gap junction protein connexin43 (Cx43) has been demonstrated to undergo internal translation initiation, yielding truncated isoforms that modulate gap junctions. The PI3K/Akt/mTOR pathway is central to translation regulation and is activated during EMT, leading us to hypothesize that altered translation initiation would contribute to gap junction loss. Using TGF-β-induced EMT as a model, we find reductions in Cx43 gap junctions despite increased transcription and stabilization of Cx43 protein. Biochemical experiments reveal suppression of the internally translated Cx43 isoform, GJA1-20k in a Smad3 and ERK-dependent manner. Ectopic expression of GJA1-20k does not halt EMT, but is sufficient to rescue gap junction formation. GJA1-20k localizes to the Golgi apparatus, and using superresolution localization microscopy we find retention of GJA1-43k at the Golgi in mesenchymal cells lacking GJA1-20k. NativePAGE demonstrates that levels of GJA1-20k regulate GJA1-43k hexamer oligomerization, a limiting step in Cx43 trafficking. These findings reveal alterations in translation initiation as an unexplored mechanism by which the cell regulates Cx43 gap junction formation during EMT.
Collapse
Affiliation(s)
- Carissa C James
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| | - Michael J Zeitz
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016
| | - Patrick J Calhoun
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| | - Samy Lamouille
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016
| | - James W Smyth
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA 24016.,Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061
| |
Collapse
|
49
|
Jiao M, Wu D, Wei Q. Myosin II-interacting guanine nucleotide exchange factor promotes bleb retraction via stimulating cortex reassembly at the bleb membrane. Mol Biol Cell 2018; 29:643-656. [PMID: 29321250 PMCID: PMC6004584 DOI: 10.1091/mbc.e17-10-0579] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/11/2017] [Accepted: 01/03/2018] [Indexed: 11/11/2022] Open
Abstract
Blebs are involved in various biological processes such as cell migration, cytokinesis, and apoptosis. While the expansion of blebs is largely an intracellular pressure-driven process, the retraction of blebs is believed to be driven by RhoA activation that leads to the reassembly of the actomyosin cortex at the bleb membrane. However, it is still poorly understood how RhoA is activated at the bleb membrane. Here, we provide evidence demonstrating that myosin II-interacting guanine nucleotide exchange factor (MYOGEF) is implicated in bleb retraction via stimulating RhoA activation and the reassembly of an actomyosin network at the bleb membrane during bleb retraction. Interaction of MYOGEF with ezrin, a well-known regulator of bleb retraction, is required for MYOGEF localization to retracting blebs. Notably, knockout of MYOGEF or ezrin not only disrupts RhoA activation at the bleb membrane, but also interferes with nonmuscle myosin II localization and activation, as well as actin polymerization in retracting blebs. Importantly, MYOGEF knockout slows down bleb retraction. We propose that ezrin interacts with MYOGEF and recruits it to retracting blebs, where MYOGEF activates RhoA and promotes the reassembly of the cortical actomyosin network at the bleb membrane, thus contributing to the regulation of bleb retraction.
Collapse
Affiliation(s)
- Meng Jiao
- Department of Biological Sciences, Fordham University, Bronx, NY 10458
| | - Di Wu
- Department of Biological Sciences, Fordham University, Bronx, NY 10458
| | - Qize Wei
- Department of Biological Sciences, Fordham University, Bronx, NY 10458
| |
Collapse
|
50
|
Gava F, Rigal L, Mondesert O, Pesce E, Ducommun B, Lobjois V. Gap junctions contribute to anchorage-independent clustering of breast cancer cells. BMC Cancer 2018; 18:221. [PMID: 29482519 PMCID: PMC5828067 DOI: 10.1186/s12885-018-4148-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/19/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Cancer cell aggregation is a key process involved in the formation of clusters of circulating tumor cells. We previously reported that cell-cell adhesion proteins, such as E-cadherin, and desmosomal proteins are involved in cell aggregation to form clusters independently of cell migration or matrix adhesion. Here, we investigated the involvement of gap junction intercellular communication (GJIC) during anchorage-independent clustering of MCF7 breast adenocarcinoma cells. METHODS We used live cell image acquisition and analysis to monitor the kinetics of MCF7 cell clustering in the presence/absence of GJIC pharmacological inhibitors and to screen a LOPAC® bioactive compound library. We also used a calcein transfer assay and flow cytometry to evaluate GJIC involvement in cancer cell clustering. RESULTS We first demonstrated that functional GJIC are established in the early phase of cancer cell aggregation. We then showed that pharmacological inhibition of GJIC using tonabersat and meclofenamate delayed MCF7 cell clustering and reduced calcein transfer. We also found that brefeldin A, an inhibitor of vesicular trafficking, which we identified by screening a small compound library, and latrunculin A, an actin cytoskeleton-disrupting agent, both impaired MCF7 cell clustering and calcein transfer. CONCLUSIONS Our results demonstrate that GJIC are involved from the earliest stages of anchorage-independent cancer cell aggregation. They also give insights into the regulatory mechanisms that could modulate the formation of clusters of circulating tumor cells.
Collapse
Affiliation(s)
- Fabien Gava
- Université de Toulouse, ITAV, CNRS, Toulouse, France
| | - Lise Rigal
- Université de Toulouse, ITAV, CNRS, Toulouse, France
| | | | - Elise Pesce
- Université de Toulouse, ITAV, CNRS, Toulouse, France
| | - Bernard Ducommun
- Université de Toulouse, ITAV, CNRS, Toulouse, France. .,CHU de Toulouse, Toulouse, France. .,Centre Pierre Potier, ITAV-USR3505, 1 Place Pierre Potier, 31106, Toulouse Cedex, France.
| | | |
Collapse
|