1
|
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
|
2
|
Jagielnicki M, Kucharska I, Bennett BC, Harris AL, Yeager M. Connexin Gap Junction Channels and Hemichannels: Insights from High-Resolution Structures. BIOLOGY 2024; 13:298. [PMID: 38785780 PMCID: PMC11117596 DOI: 10.3390/biology13050298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 05/25/2024]
Abstract
Connexins (Cxs) are a family of integral membrane proteins, which function as both hexameric hemichannels (HCs) and dodecameric gap junction channels (GJCs), behaving as conduits for the electrical and molecular communication between cells and between cells and the extracellular environment, respectively. Their proper functioning is crucial for many processes, including development, physiology, and response to disease and trauma. Abnormal GJC and HC communication can lead to numerous pathological states including inflammation, skin diseases, deafness, nervous system disorders, and cardiac arrhythmias. Over the last 15 years, high-resolution X-ray and electron cryomicroscopy (cryoEM) structures for seven Cx isoforms have revealed conservation in the four-helix transmembrane (TM) bundle of each subunit; an αβ fold in the disulfide-bonded extracellular loops and inter-subunit hydrogen bonding across the extracellular gap that mediates end-to-end docking to form a tight seal between hexamers in the GJC. Tissue injury is associated with cellular Ca2+ overload. Surprisingly, the binding of 12 Ca2+ ions in the Cx26 GJC results in a novel electrostatic gating mechanism that blocks cation permeation. In contrast, acidic pH during tissue injury elicits association of the N-terminal (NT) domains that sterically blocks the pore in a "ball-and-chain" fashion. The NT domains under physiologic conditions display multiple conformational states, stabilized by protein-protein and protein-lipid interactions, which may relate to gating mechanisms. The cryoEM maps also revealed putative lipid densities within the pore, intercalated among transmembrane α-helices and between protomers, the functions of which are unknown. For the future, time-resolved cryoEM of isolated Cx channels as well as cryotomography of GJCs and HCs in cells and tissues will yield a deeper insight into the mechanisms for channel regulation. The cytoplasmic loop (CL) and C-terminal (CT) domains are divergent in sequence and length, are likely involved in channel regulation, but are not visualized in the high-resolution X-ray and cryoEM maps presumably due to conformational flexibility. We expect that the integrated use of synergistic physicochemical, spectroscopic, biophysical, and computational methods will reveal conformational dynamics relevant to functional states. We anticipate that such a wealth of results under different pathologic conditions will accelerate drug discovery related to Cx channel modulation.
Collapse
Affiliation(s)
- Maciej Jagielnicki
- The Phillip and Patricia Frost Institute for Chemistry and Molecular Science, Department of Chemistry, University of Miami, 1201 Memorial Drive, Miami, FL 33146, USA; (M.J.); (I.K.)
| | - Iga Kucharska
- The Phillip and Patricia Frost Institute for Chemistry and Molecular Science, Department of Chemistry, University of Miami, 1201 Memorial Drive, Miami, FL 33146, USA; (M.J.); (I.K.)
| | - Brad C. Bennett
- Department of Biological and Environmental Sciences, Howard College of Arts and Sciences, Samford University, Birmingham, AL 35229, USA;
| | - Andrew L. Harris
- Rutgers New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ 07103, USA;
| | - Mark Yeager
- The Phillip and Patricia Frost Institute for Chemistry and Molecular Science, Department of Chemistry, University of Miami, 1201 Memorial Drive, Miami, FL 33146, USA; (M.J.); (I.K.)
- The Phillip and Patricia Frost Institute for Chemistry and Molecular Science, Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL 33146, USA
| |
Collapse
|
3
|
Baker KM, Abt M, Doud EH, Oblak AL, Yeh ES. Mapping the Anti-Cancer Activity of α-Connexin Carboxyl-Terminal (aCT1) Peptide in Resistant HER2+ Breast Cancer. Cancers (Basel) 2024; 16:423. [PMID: 38275864 PMCID: PMC10814893 DOI: 10.3390/cancers16020423] [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: 12/20/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Connexin 43 (Cx43) is a protein encoded by the GJA1 gene and is a component of cell membrane structures called gap junctions, which facilitate intercellular communication. Prior evidence indicates that elevated GJA1 expression in the HER2-positive (HER2+) subtype of breast cancer is associated with poor prognosis. Prior evidence also suggests that HER2+ breast cancers that have become refractory to HER2-targeted agents have a loss of Cx43 gap junction intercellular communication (GJIC). In this study, a Cx43-targeted agent called alpha-connexin carboxyl-terminal peptide (aCT1) is examined to determine whether GJIC can be rescued in refractory HER2+ breast cancer cells. A proposed mechanism of action for aCT1 is binding to the tight junction protein Zonal Occludens-1 (ZO-1). However, the true scope of activity for aCT1 has not been explored. In this study, mass spectrometry proteomic analysis is used to determine the breadth of aCT1-interacting proteins. The NanoString nCounter Breast Cancer 360 panel is also used to examine the effect of aCT1 on cancer signaling in HER2+ breast cancer cells. Findings from this study show a dynamic range of binding partners for aCT1, many of which regulate gene expression and RNA biology. nCounter analysis shows that a number of pathways are significantly impacted by aCT1, including upregulation of apoptotic factors, leading to the prediction and demonstration that aCT1 can boost the cell death effects of cisplatin and lapatinib in HER2+ breast cancer cells that have become resistant to HER2-targeted agents.
Collapse
Affiliation(s)
- Kimberly M. Baker
- Department of Biology, University of Indianapolis, Indianapolis, IN 46227, USA;
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Melissa Abt
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Emma H. Doud
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Adrian L. Oblak
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Elizabeth S. Yeh
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| |
Collapse
|
4
|
Gebicke-Haerter PJ. The computational power of the human brain. Front Cell Neurosci 2023; 17:1220030. [PMID: 37608987 PMCID: PMC10441807 DOI: 10.3389/fncel.2023.1220030] [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: 05/19/2023] [Accepted: 07/05/2023] [Indexed: 08/24/2023] Open
Abstract
At the end of the 20th century, analog systems in computer science have been widely replaced by digital systems due to their higher computing power. Nevertheless, the question keeps being intriguing until now: is the brain analog or digital? Initially, the latter has been favored, considering it as a Turing machine that works like a digital computer. However, more recently, digital and analog processes have been combined to implant human behavior in robots, endowing them with artificial intelligence (AI). Therefore, we think it is timely to compare mathematical models with the biology of computation in the brain. To this end, digital and analog processes clearly identified in cellular and molecular interactions in the Central Nervous System are highlighted. But above that, we try to pinpoint reasons distinguishing in silico computation from salient features of biological computation. First, genuinely analog information processing has been observed in electrical synapses and through gap junctions, the latter both in neurons and astrocytes. Apparently opposed to that, neuronal action potentials (APs) or spikes represent clearly digital events, like the yes/no or 1/0 of a Turing machine. However, spikes are rarely uniform, but can vary in amplitude and widths, which has significant, differential effects on transmitter release at the presynaptic terminal, where notwithstanding the quantal (vesicular) release itself is digital. Conversely, at the dendritic site of the postsynaptic neuron, there are numerous analog events of computation. Moreover, synaptic transmission of information is not only neuronal, but heavily influenced by astrocytes tightly ensheathing the majority of synapses in brain (tripartite synapse). At least at this point, LTP and LTD modifying synaptic plasticity and believed to induce short and long-term memory processes including consolidation (equivalent to RAM and ROM in electronic devices) have to be discussed. The present knowledge of how the brain stores and retrieves memories includes a variety of options (e.g., neuronal network oscillations, engram cells, astrocytic syncytium). Also epigenetic features play crucial roles in memory formation and its consolidation, which necessarily guides to molecular events like gene transcription and translation. In conclusion, brain computation is not only digital or analog, or a combination of both, but encompasses features in parallel, and of higher orders of complexity.
Collapse
Affiliation(s)
- Peter J. Gebicke-Haerter
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
5
|
Yuan J, Huang X, Zhao Y, Gu J, Yuan Y, Liu Z, Zou H, Bian J. Rat Hepatocytes Mitigate Cadmium Toxicity by Forming Annular Gap Junctions and Degrading Them via Endosome-Lysosome Pathway. Int J Mol Sci 2022; 23:ijms232415607. [PMID: 36555247 PMCID: PMC9778680 DOI: 10.3390/ijms232415607] [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: 11/11/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Gap junction protein connexin 43 (Cx43) plays a critical role in gap junction communication in rat hepatocytes. However, those located between hepatocytes are easily internalized following exposure to poisons. Herein, we investigated the potential of buffalo rat liver 3A (BRL 3A) cells to generate annular gap junctions (AGJs) proficient at alleviating cadmium (Cd) cytotoxic injury through degradation via an endosome-lysosome pathway. Our results showed that Cd-induced damage of liver microtubules promoted Cx43 internalization and increased Cx43 phosphorylation at Ser373 site. Furthermore, we established that Cd induced AGJs generation in BRL 3A cells, and AGJs were subsequently degraded through the endosome-lysosome pathway. Overall, our results suggested that Cx43 internalization and the generation of AGJs were cellular protective mechanisms to alleviate Cd toxicity in rat hepatocytes.
Collapse
Affiliation(s)
- Junzhao Yuan
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xiaoqian Huang
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yumeng Zhao
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: (H.Z.); (J.B.)
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: (H.Z.); (J.B.)
| |
Collapse
|
6
|
Casanellas I, Lagunas A, Vida Y, Pérez-Inestrosa E, Rodríguez-Pereira C, Magalhaes J, Andrades JA, Becerra J, Samitier J. Nanoscale ligand density modulates gap junction intercellular communication of cell condensates during chondrogenesis. Nanomedicine (Lond) 2022; 17:775-791. [PMID: 35642556 DOI: 10.2217/nnm-2021-0399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To unveil the influence of cell-matrix adhesions in the establishment of gap junction intercellular communication (GJIC) during cell condensation in chondrogenesis. Materials & methods: Previously developed nanopatterns of the cell adhesive ligand arginine-glycine-aspartic acid were used as cell culture substrates to control cell adhesion at the nanoscale. In vitro chondrogenesis of mesenchymal stem cells was conducted on the nanopatterns. Cohesion and GJIC were evaluated in cell condensates. Results: Mechanical stability and GJIC are enhanced by a nanopattern configuration in which 90% of the surface area presents adhesion sites separated less than 70 nm, thus providing an onset for cell signaling. Conclusion: Cell-matrix adhesions regulate GJIC of mesenchymal cell condensates during in vitro chondrogenesis from a threshold configuration at the nanoscale.
Collapse
Affiliation(s)
- Ignasi Casanellas
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science &Technology (BIST). c/Baldiri Reixac, 10-12, Barcelona, 08028, Spain.,Department of Electronics & Biomedical Engineering, University of Barcelona (UB). c/Martí i Franquès, 1, 08028, Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Anna Lagunas
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science &Technology (BIST). c/Baldiri Reixac, 10-12, Barcelona, 08028, Spain.,Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Yolanda Vida
- Universidad de Málaga-IBIMA, Dpto. Química Orgánica. Campus de Teatinos s/n, Málaga, 29071, Spain.,Centro Andaluz de Nanomedicina y Biotecnología-BIONAND. Parque Tecnológico de Andalucía, c/Severo Ochoa 35, C,ampanillas, Málaga, 29590, Spain
| | - Ezequiel Pérez-Inestrosa
- Universidad de Málaga-IBIMA, Dpto. Química Orgánica. Campus de Teatinos s/n, Málaga, 29071, Spain.,Centro Andaluz de Nanomedicina y Biotecnología-BIONAND. Parque Tecnológico de Andalucía, c/Severo Ochoa 35, C,ampanillas, Málaga, 29590, Spain
| | - Cristina Rodríguez-Pereira
- Unidad de Medicina Regenerativa, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC). c/Xubias de Arriba, 84, A Coruña, 15006, Spain
| | - Joana Magalhaes
- Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.,Unidad de Medicina Regenerativa, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC). c/Xubias de Arriba, 84, A Coruña, 15006, Spain
| | - José A Andrades
- Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.,Centro Andaluz de Nanomedicina y Biotecnología-BIONAND. Parque Tecnológico de Andalucía, c/Severo Ochoa 35, C,ampanillas, Málaga, 29590, Spain.,Department of Cell Biology, Genetics & Physiology, Universidad de Málaga (UMA), Instituto de Investigación Biomédica de Málaga (IBIMA). Av. Cervantes, 2, Málaga, 29071, Spain
| | - José Becerra
- Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.,Centro Andaluz de Nanomedicina y Biotecnología-BIONAND. Parque Tecnológico de Andalucía, c/Severo Ochoa 35, C,ampanillas, Málaga, 29590, Spain.,Department of Cell Biology, Genetics & Physiology, Universidad de Málaga (UMA), Instituto de Investigación Biomédica de Málaga (IBIMA). Av. Cervantes, 2, Málaga, 29071, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science &Technology (BIST). c/Baldiri Reixac, 10-12, Barcelona, 08028, Spain.,Department of Electronics & Biomedical Engineering, University of Barcelona (UB). c/Martí i Franquès, 1, 08028, Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| |
Collapse
|
7
|
Minogue PJ, Tong JJ, Wichmann K, Mysliwiec H, Ebihara L, Beyer EC, Berthoud VM. Cataract-linked serine mutations in the gap junction protein connexin50 expose a sorting signal that promotes its lysosomal degradation. J Biol Chem 2022; 298:101673. [PMID: 35120923 PMCID: PMC8927986 DOI: 10.1016/j.jbc.2022.101673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 01/24/2023] Open
Abstract
Many human connexin50 (Cx50) mutants have been linked to cataracts including two carboxyl terminus serine mutants that are known phosphorylation sites in the lens (Cx50S258F and Cx50S259Y). To examine the behavior of these mutants and the role of phosphorylation at these positions, we stably transfected HeLa cells with cataract-linked and phosphorylation-mimicking (Cx50S258D and Cx50S259D) Cx50 mutants. We observed that gap junctional plaques were rarely detected in Cx50S258F- and Cx50S259Y-expressing cells compared with wild-type cells. In addition, gap junction abundance and size were greatly increased for Cx50S258D and Cx50S259D mutants. Cx50S258F and Cx50S259Y supported very low levels of gap junctional coupling, whereas Cx50S258D and Cx50S259D supported extensive intercellular communication. Furthermore, Cx50 levels as detected by immunoblotting were lower in Cx50S258F and Cx50S259Y mutants than in the wild type or the aspartate substitution mutants, and chloroquine or ammonium chloride treatment significantly increased Cx50S258F and Cx50S259Y protein levels, implying participation of the lysosome in their increased degradation. Alanine substitution of amino acids within a predicted tyrosine-based sorting signal in Cx50S258F and Cx50S259Y increased levels of gap junctional plaques and intercellular transfer of Neurobiotin. These results suggest that absence of phosphorylatable serines at these positions exposes a sorting signal leading to lysosomal degradation of Cx50, whereas phosphorylation at these sites conceals this signal and allows targeting of Cx50 to the plasma membrane and stabilization of gap junction plaques. We propose that in the lens degradation of Cx50S258F and Cx50S259Y decreases Cx50 levels at the plasma membrane and consequently Cx50 function, leading to cataracts.
Collapse
Affiliation(s)
- Peter J Minogue
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | - Jun-Jie Tong
- Center of Proteomics and Molecular Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Kelly Wichmann
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | - Hubert Mysliwiec
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | - Lisa Ebihara
- Center of Proteomics and Molecular Therapeutics/Discipline of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Eric C Beyer
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | - Viviana M Berthoud
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA.
| |
Collapse
|
8
|
Morioka N, Kondo S, Harada N, Takimoto T, Tokunaga N, Nakamura Y, Hisaoka-Nakashima K, Nakata Y. Downregulation of connexin43 potentiates noradrenaline-induced expression of brain-derived neurotrophic factor in primary cultured cortical astrocytes. J Cell Physiol 2021; 236:6777-6792. [PMID: 33665818 DOI: 10.1002/jcp.30353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022]
Abstract
Decreased expression of brain-derived neurotrophic factor (BDNF) is involved in the pathology of depressive disorders. Astrocytes produce BDNF following antidepressant treatment or stimulation of adrenergic receptors. Connexin43 (Cx43) is mainly expressed in central nervous system astrocytes and its expression is downregulated in patients with major depression. How changes in Cx43 expression affect astrocyte function, including BDNF production, is poorly understood. The current study examined the effect of Cx43 knockdown on BDNF expression in cultured cortical astrocytes after stimulation of adrenergic receptors. The expression of Cx43 in rat primary cultured cortical astrocytes was downregulated with RNA interference. Levels of messenger RNAs (mRNAs) or proteins were measured by real-time PCR and western blotting, respectively. Knockdown of Cx43 potentiated noradrenaline (NA)-induced expression of BDNF mRNA in cultured astrocytes. NA treatment induced proBDNF protein expression in astrocytes transfected with small interfering RNA (siRNA) targeting Cx43, but not with control siRNA. This potentiation was mediated by the Src tyrosine kinase-extracellular signal-regulated kinase (ERK) pathway through stimulation of adrenergic α1 and β receptors. Furthermore, the Gq/11 protein-Src-ERK pathway and the G-protein coupled receptor kinase 2-Src-ERK pathway were involved in α1 and β adrenergic receptor-mediated potentiation of BDNF mRNA expression, respectively. The current studies demonstrate a novel mechanism of BDNF expression in cortical astrocytes mediated by Cx43, in which downregulation of Cx43 increases, through adrenergic receptors, the expression of BDNF. The current findings indicate a potentially novel mechanism of action of antidepressants, via regulation of astrocytic Cx43 expression and subsequent BDNF expression.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Astrocytes/drug effects
- Astrocytes/metabolism
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Cells, Cultured
- Cerebral Cortex/cytology
- Cerebral Cortex/drug effects
- Cerebral Cortex/metabolism
- Connexin 43/genetics
- Connexin 43/metabolism
- Down-Regulation
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Gene Knockdown Techniques
- Male
- Norepinephrine/pharmacology
- Primary Cell Culture
- RNA Interference
- Rats, Wistar
- Receptors, Adrenergic, alpha-1/drug effects
- Receptors, Adrenergic, alpha-1/metabolism
- Receptors, Adrenergic, beta/drug effects
- Receptors, Adrenergic, beta/metabolism
- Signal Transduction
- src-Family Kinases/metabolism
- Rats
Collapse
Affiliation(s)
- Norimitsu Morioka
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Syun Kondo
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Nanase Harada
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Tomoyo Takimoto
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Nozomi Tokunaga
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yoshihiro Nakata
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| |
Collapse
|
9
|
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: 57] [Impact Index Per Article: 19.0] [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
|
10
|
Kanapathy M, Hachach-Haram N, Bystrzonowski N, Becker DL, Mosahebi A, Richards T. Epidermal graft encourages wound healing by down-regulation of gap junctional protein and activation of wound bed without graft integration as opposed to split-thickness skin graft. Int Wound J 2021; 18:332-341. [PMID: 33751815 PMCID: PMC8244016 DOI: 10.1111/iwj.13536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 01/07/2023] Open
Abstract
Wound coverage by split-thickness skin graft (SSG) and epidermal graft (EG) shortens healing time, with comparable outcomes. However, the healing mechanism of EG is not as well understood as SSG. The difference in the healing mechanisms of EG and SSG was investigated using gap junctional proteins, proliferative marker, and cytokeratin markers. Paired punch biopsies were taken from the wound edge and wound bed from patients undergoing EG and SSG at weeks 0 and 1 to investigate wound edge keratinocyte migratory activities (connexins 43, 30, and 26), wound bed activation (Ki67), and the presence of graft integration to the wound bed (cytokeratins 14 and 6). Twenty-four paired biopsies were taken at weeks 0 and 1 (EG, n = 12; SSG, n = 12). Wound edge biopsies demonstrated down-regulation of connexins 43 (P = .023) and 30 (P = .027) after EG, indicating accelerated healing from the wound edge. At week 1, increased expression of Ki67 (P < .05) was seen after EG, indicating activation of cells within the wound bed. Keratinocytes expressing cytokeratins 6 and 14 were observed on all wounds treated with SSG but were absent at week 1 after EG, indicating the absence of graft integration following EG. Despite EG and SSG both being autologous skin grafts, they demonstrate different mechanisms of wound healing. EG accelerates wound healing from the wound edges and activates the wound bed despite not integrating into the wound bed at week 1 post-grafting as opposed to SSG, hence demonstrating properties comparable with a bioactive dressing instead of a skin substitute.
Collapse
Affiliation(s)
- Muholan Kanapathy
- Division of Surgery and Interventional Science, University College London, London, UK.,Department of Plastic and Reconstructive Surgery, Royal Free NHS Foundation Trust Hospital, London, UK
| | - Nadine Hachach-Haram
- Department of Plastic and Reconstructive Surgery, Royal Free NHS Foundation Trust Hospital, London, UK
| | - Nicola Bystrzonowski
- Department of Plastic and Reconstructive Surgery, Royal Free NHS Foundation Trust Hospital, London, UK
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Institute of Medical Biology, A*Star, Immunos, Biomedical Grove, Singapore
| | - Afshin Mosahebi
- Division of Surgery and Interventional Science, University College London, London, UK.,Department of Plastic and Reconstructive Surgery, Royal Free NHS Foundation Trust Hospital, London, UK
| | - Toby Richards
- Division of Surgery and Interventional Science, University College London, London, UK.,Department of Plastic and Reconstructive Surgery, Royal Free NHS Foundation Trust Hospital, London, UK
| |
Collapse
|
11
|
Andelova K, Egan Benova T, Szeiffova Bacova B, Sykora M, Prado NJ, Diez ER, Hlivak P, Tribulova N. Cardiac Connexin-43 Hemichannels and Pannexin1 Channels: Provocative Antiarrhythmic Targets. Int J Mol Sci 2020; 22:ijms22010260. [PMID: 33383853 PMCID: PMC7795512 DOI: 10.3390/ijms22010260] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac connexin-43 (Cx43) creates gap junction channels (GJCs) at intercellular contacts and hemi-channels (HCs) at the peri-junctional plasma membrane and sarcolemmal caveolae/rafts compartments. GJCs are fundamental for the direct cardiac cell-to-cell transmission of electrical and molecular signals which ensures synchronous myocardial contraction. The HCs and structurally similar pannexin1 (Panx1) channels are active in stressful conditions. These channels are essential for paracrine and autocrine communication through the release of ions and signaling molecules to the extracellular environment, or for uptake from it. The HCs and Panx1 channel-opening profoundly affects intracellular ionic homeostasis and redox status and facilitates via purinergic signaling pro-inflammatory and pro-fibrotic processes. These conditions promote cardiac arrhythmogenesis due to the impairment of the GJCs and selective ion channel function. Crosstalk between GJCs and HCs/Panx1 channels could be crucial in the development of arrhythmogenic substrates, including fibrosis. Despite the knowledge gap in the regulation of these channels, current evidence indicates that HCs and Panx1 channel activation can enhance the risk of cardiac arrhythmias. It is extremely challenging to target HCs and Panx1 channels by inhibitory agents to hamper development of cardiac rhythm disorders. Progress in this field may contribute to novel therapeutic approaches for patients prone to develop atrial or ventricular fibrillation.
Collapse
Affiliation(s)
- Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
| | - Tamara Egan Benova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
| | - Natalia Jorgelina Prado
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, M5500 Mendoza, Argentina; (N.J.P.); (E.R.D.)
| | - Emiliano Raul Diez
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, M5500 Mendoza, Argentina; (N.J.P.); (E.R.D.)
| | - Peter Hlivak
- Department of Arrhythmias and Pacing, National Institute of Cardiovascular Diseases, Pod Krásnou Hôrkou 1, 83348 Bratislava, Slovakia;
| | - Narcis Tribulova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 84104 Bratislava, Slovakia; (K.A.); (T.E.B.); (B.S.B.); (M.S.)
- Correspondence: ; Tel.: +421-2-32295-423
| |
Collapse
|
12
|
Lin YF, Lin HC, Tsai CL, Hsu YC. GJB2 mutation spectrum in the Taiwanese population and genotype-phenotype comparisons in patients with hearing loss carrying GJB2 c.109G>A and c.235delC mutations. Hear Res 2020; 413:108135. [PMID: 33288323 DOI: 10.1016/j.heares.2020.108135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/15/2020] [Accepted: 11/24/2020] [Indexed: 11/04/2022]
Abstract
Hearing loss, the most common sensory abnormality, is caused by the death of or damage to inner ear hair cells. Genetic mutations are the main cause of hearing loss. We used nex-generation sequencing data released by the Taiwan Biobank to investigate the GJB2 mutation spectrum in 1517 patients. We compared hearing function in Taiwanese patients with nonsyndromic hearing loss (NSHL) caused by the two most common GJB2 mutations c.109G>A (p.V37I) and c.235delC. We extracted DNA from the oral mucosa of patients with NSHL and performed Sanger sequencing to confirm the genotype. Of 240 patients with NSHL, we identified 25 with GJB2 c.109G>A and 9 with GJB2 c.235delC mutations, after excluding patients aged >10 years, in whom hearing loss may have been caused due to age-related degeneration. We investigated genotype-phenotype correlations in patients harboring GJB2 c.109G>A and c.235delC mutations. Furthermore, we described the GJB2 mutation spectrum in the Taiwanese population and identified the role of homozygous and heterozygous GJB2 mutations associated with hearing phenotypes in patients with NSHL. Thus, our study provides insights into the complexity of GJB2 genetics. Our data indicate that GJB2 c.109G>A heterozygotes had poorer hearing than did homozygotes. The mechanism underlying the more severe phenotype in heterozygotes and whether the phenotype is caused by GJB2 heterozygotes or compound heterozygotes warrant future investigation.
Collapse
Affiliation(s)
- Yi-Feng Lin
- Clinico Hearingaid Asia Co., Ltd.; Institute of Biomedical Sciences, Mackay Medical College, No. 46, Sec. 3, Zhongzheng Rd., Sanzhi Dist., 252 New Taipei City, Taiwan; Department of Audiology and Speech-Language Pathology, Mackay Medical College, New Taipei City, Taiwan
| | - Hung-Ching Lin
- Department of Audiology and Speech-Language Pathology, Mackay Medical College, New Taipei City, Taiwan; Department of Otolaryngology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chia-Ling Tsai
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Sec. 3, Zhongzheng Rd., Sanzhi Dist., 252 New Taipei City, Taiwan
| | - Yi-Chao Hsu
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Sec. 3, Zhongzheng Rd., Sanzhi Dist., 252 New Taipei City, Taiwan.
| |
Collapse
|
13
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Leila Slaoui
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Noémie Mazaré
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Marc Oudart
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Rodrigo Alvear-Perez
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Xabier Elorza-Vidal
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
14
|
Antagonistic Functions of Connexin 43 during the Development of Primary or Secondary Bone Tumors. Biomolecules 2020; 10:biom10091240. [PMID: 32859065 PMCID: PMC7565206 DOI: 10.3390/biom10091240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
Despite research and clinical advances during recent decades, bone cancers remain a leading cause of death worldwide. There is a low survival rate for patients with primary bone tumors such as osteosarcoma and Ewing’s sarcoma or secondary bone tumors such as bone metastases from prostate carcinoma. Gap junctions are specialized plasma membrane structures consisting of transmembrane channels that directly link the cytoplasm of adjacent cells, thereby enabling the direct exchange of small signaling molecules between cells. Discoveries of human genetic disorders due to genetic mutations in gap junction proteins (connexins) and experimental data using connexin knockout mice have provided significant evidence that gap-junctional intercellular communication (Gj) is crucial for tissue function. Thus, the dysfunction of Gj may be responsible for the development of some diseases. Gj is thus a main mechanism for tumor cells to communicate with other tumor cells and their surrounding microenvironment to survive and proliferate. If it is well accepted that a low level of connexin expression favors cancer cell proliferation and therefore primary tumor development, more evidence is suggesting that a high level of connexin expression stimulates various cellular process such as intravasation, extravasation, or migration of metastatic cells. If so, connexin expression would facilitate secondary tumor dissemination. This paper discusses evidence that suggests that connexin 43 plays an antagonistic role in the development of primary bone tumors as a tumor suppressor and secondary bone tumors as a tumor promoter.
Collapse
|
15
|
Angé M, Castanares-Zapatero D, De Poortere J, Dufeys C, Courtoy GE, Bouzin C, Quarck R, Bertrand L, Beauloye C, Horman S. α1AMP-Activated Protein Kinase Protects against Lipopolysaccharide-Induced Endothelial Barrier Disruption via Junctional Reinforcement and Activation of the p38 MAPK/HSP27 Pathway. Int J Mol Sci 2020; 21:ijms21155581. [PMID: 32759774 PMCID: PMC7432762 DOI: 10.3390/ijms21155581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular hyperpermeability is a determinant factor in the pathophysiology of sepsis. While, AMP-activated protein kinase (AMPK) is known to play a role in maintaining endothelial barrier function in this condition. Therefore, we investigated the underlying molecular mechanisms of this protective effect. α1AMPK expression and/or activity was modulated in human dermal microvascular endothelial cells using either α1AMPK-targeting small interfering RNA or the direct pharmacological AMPK activator 991, prior to lipopolysaccharide (LPS) treatment. Western blotting was used to analyze the expression and/or phosphorylation of proteins that compose cellular junctions (zonula occludens-1 (ZO-1), vascular endothelial cadherin (VE-Cad), connexin 43 (Cx43)) or that regulate actin cytoskeleton (p38 MAPK; heat shock protein 27 (HSP27)). Functional endothelial permeability was assessed by in vitro Transwell assays, and quantification of cellular junctions in the plasma membrane was assessed by immunofluorescence. Actin cytoskeleton remodeling was evaluated through actin fluorescent staining. We consequently demonstrate that α1AMPK deficiency is associated with reduced expression of CX43, ZO-1, and VE-Cad, and that the drastic loss of CX43 is likely responsible for the subsequent decreased expression and localization of ZO-1 and VE-Cad in the plasma membrane. Moreover, α1AMPK activation by 991 protects against LPS-induced endothelial barrier disruption by reinforcing cortical actin cytoskeleton. This is due to a mechanism that involves the phosphorylation of p38 MAPK and HSP27, which is nonetheless independent of the small GTPase Rac1. This results in a drastic decrease of LPS-induced hyperpermeability. We conclude that α1AMPK activators that are suitable for clinical use may provide a specific therapeutic intervention that limits sepsis-induced vascular leakage.
Collapse
Affiliation(s)
- Marine Angé
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
| | - Diego Castanares-Zapatero
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
- Division of Intensive Care, Cliniques Universitaires Saint Luc, 1200 Brussels, Belgium
| | - Julien De Poortere
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
| | - Cécile Dufeys
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
| | - Guillaume E. Courtoy
- IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (G.E.C.); (C.B.)
| | - Caroline Bouzin
- IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (G.E.C.); (C.B.)
| | - Rozenn Quarck
- Department of Chronic Diseases & Metabolism (CHROMETA), Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), KU Leuven, 3000 Leuven, Belgium;
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
- Division of Cardiology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.A.); (D.C.-Z.); (J.D.P.); (C.D.); (L.B.); (C.B.)
- Correspondence: ; Tel.: +32-2-764-55-66
| |
Collapse
|
16
|
Abstract
Of the 21 members of the connexin family, 4 (Cx37, Cx40, Cx43, and Cx45) are expressed in the endothelium and/or smooth muscle of intact blood vessels to a variable and dynamically regulated degree. Full-length connexins oligomerize and form channel structures connecting the cytosol of adjacent cells (gap junctions) or the cytosol with the extracellular space (hemichannels). The different connexins vary mainly with regard to length and sequence of their cytosolic COOH-terminal tails. These COOH-terminal parts, which in the case of Cx43 are also translated as independent short isoforms, are involved in various cellular signaling cascades and regulate cell functions. This review focuses on channel-dependent and -independent effects of connexins in vascular cells. Channels play an essential role in coordinating and synchronizing endothelial and smooth muscle activity and in their interplay, in the control of vasomotor actions of blood vessels including endothelial cell reactivity to agonist stimulation, nitric oxide-dependent dilation, and endothelial-derived hyperpolarizing factor-type responses. Further channel-dependent and -independent roles of connexins in blood vessel function range from basic processes of vascular remodeling and angiogenesis to vascular permeability and interactions with leukocytes with the vessel wall. Together, these connexin functions constitute an often underestimated basis for the enormous plasticity of vascular morphology and function enabling the required dynamic adaptation of the vascular system to varying tissue demands.
Collapse
Affiliation(s)
- Ulrich Pohl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany; Biomedical Centre, Cardiovascular Physiology, LMU Munich, Planegg-Martinsried, Germany; German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
17
|
Liu W, Cui Y, Wei J, Sun J, Zheng L, Xie J. Gap junction-mediated cell-to-cell communication in oral development and oral diseases: a concise review of research progress. Int J Oral Sci 2020; 12:17. [PMID: 32532966 PMCID: PMC7293327 DOI: 10.1038/s41368-020-0086-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/30/2020] [Accepted: 05/19/2020] [Indexed: 02/05/2023] Open
Abstract
Homoeostasis depends on the close connection and intimate molecular exchange between extracellular, intracellular and intercellular networks. Intercellular communication is largely mediated by gap junctions (GJs), a type of specialized membrane contact composed of variable number of channels that enable direct communication between cells by allowing small molecules to pass directly into the cytoplasm of neighbouring cells. Although considerable evidence indicates that gap junctions contribute to the functions of many organs, such as the bone, intestine, kidney, heart, brain and nerve, less is known about their role in oral development and disease. In this review, the current progress in understanding the background of connexins and the functions of gap junctions in oral development and diseases is discussed. The homoeostasis of tooth and periodontal tissues, normal tooth and maxillofacial development, saliva secretion and the integrity of the oral mucosa depend on the proper function of gap junctions. Knowledge of this pattern of cell-cell communication is required for a better understanding of oral diseases. With the ever-increasing understanding of connexins in oral diseases, therapeutic strategies could be developed to target these membrane channels in various oral diseases and maxillofacial dysplasia.
Collapse
Affiliation(s)
- Wenjing Liu
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jieya Wei
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianxun Sun
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Calhoun PJ, Phan AV, Taylor JD, James CC, Padget RL, Zeitz MJ, Smyth JW. Adenovirus targets transcriptional and posttranslational mechanisms to limit gap junction function. FASEB J 2020; 34:9694-9712. [PMID: 32485054 DOI: 10.1096/fj.202000667r] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 01/19/2023]
Abstract
Adenoviruses are responsible for a spectrum of pathogenesis including viral myocarditis. The gap junction protein connexin43 (Cx43, gene name GJA1) facilitates rapid propagation of action potentials necessary for each heartbeat. Gap junctions also propagate innate and adaptive antiviral immune responses, but how viruses may target these structures is not understood. Given this immunological role of Cx43, we hypothesized that gap junctions would be targeted during adenovirus type 5 (Ad5) infection. We find reduced Cx43 protein levels due to decreased GJA1 mRNA transcripts dependent upon β-catenin transcriptional activity during Ad5 infection, with early viral protein E4orf1 sufficient to induce β-catenin phosphorylation. Loss of gap junction function occurs prior to reduced Cx43 protein levels with Ad5 infection rapidly inducing Cx43 phosphorylation events consistent with altered gap junction conductance. Direct Cx43 interaction with ZO-1 plays a critical role in gap junction regulation. We find loss of Cx43/ZO-1 complexing during Ad5 infection by co-immunoprecipitation and complementary studies in human induced pluripotent stem cell derived-cardiomyocytes reveal Cx43 gap junction remodeling by reduced ZO-1 complexing. These findings reveal specific targeting of gap junction function by Ad5 leading to loss of intercellular communication which would contribute to dangerous pathological states including arrhythmias in infected hearts.
Collapse
Affiliation(s)
- Patrick J Calhoun
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Allen V Phan
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | | | - Carissa C James
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Rachel L Padget
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Michael J Zeitz
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
| | - James W Smyth
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| |
Collapse
|
19
|
Martin EA, Lasseigne AM, Miller AC. Understanding the Molecular and Cell Biological Mechanisms of Electrical Synapse Formation. Front Neuroanat 2020; 14:12. [PMID: 32372919 PMCID: PMC7179694 DOI: 10.3389/fnana.2020.00012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
In this review article, we will describe the recent advances made towards understanding the molecular and cell biological mechanisms of electrical synapse formation. New evidence indicates that electrical synapses, which are gap junctions between neurons, can have complex molecular compositions including protein asymmetries across joined cells, diverse morphological arrangements, and overlooked similarities with other junctions, all of which indicate new potential roles in neurodevelopmental disease. Aquatic organisms, and in particular the vertebrate zebrafish, have proven to be excellent models for elucidating the molecular mechanisms of electrical synapse formation. Zebrafish will serve as our main exemplar throughout this review and will be compared with other model organisms. We highlight the known cell biological processes that build neuronal gap junctions and compare these with the assemblies of adherens junctions, tight junctions, non-neuronal gap junctions, and chemical synapses to explore the unknown frontiers remaining in our understanding of the critical and ubiquitous electrical synapse.
Collapse
Affiliation(s)
- E Anne Martin
- Department of Biology, Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| | - Abagael M Lasseigne
- Department of Biology, Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| | - Adam C Miller
- Department of Biology, Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| |
Collapse
|
20
|
Morioka N, Nakamura Y, Zhang FF, Hisaoka-Nakashima K, Nakata Y. Role of Connexins in Chronic Pain and Their Potential as Therapeutic Targets for Next-Generation Analgesics. Biol Pharm Bull 2019; 42:857-866. [PMID: 31155584 DOI: 10.1248/bpb.b19-00195] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic pain, including inflammatory, neuropathic pain, is a serious clinical issue. There are increasing numbers of patients with chronic pain due to the growing number of elderly and it is estimated that about 25% of the global population will develop chronic pain. Chronic pain patients are refractory to medications used to treat acute pain such as opioids and non-steroidal anti-inflammatory drugs. Furthermore, the complexity and diversity of chronic pain mechanisms hinder the development of new analgesics. Thus, a better understanding of the mechanism of chronic pain is needed, which would facilitate the development of novel analgesics based on novel mechanisms. With this goal, connexins (Cxs) could be targeted for the development of new analgesics. Connexins are proteins with 20 subtypes, and function as channels, gap junctions between cells, and hemichannels that sample the extracellular space and release molecules such as neurotransmitters. Furthermore, Cxs could have functions independent of channel activity. Recent studies have shown that Cxs could be crucial in the induction and maintenance of chronic pain, and modulation of the activity or the expression of Cxs ameliorates nociceptive hypersensitivity in multiple chronic pain models. This review will cite novel findings on the role of of Cxs in the nociceptive transduction pathway under the chronic pain state and antinociceptive effects of various molecules modulating activity or expression of Cxs. Also, the potential of Cx modulation as a therapeutic strategy for intractable chronic pain will be discussed.
Collapse
Affiliation(s)
- Norimitsu Morioka
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences
| | - Yoki Nakamura
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences
| | - Fang Fang Zhang
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences.,Institute of Pharmacology, Taishan Medical University
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences
| | - Yoshihiro Nakata
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences
| |
Collapse
|
21
|
Abstract
The eutherian connexins were characterized as protein constituents of gap junctions implicated in cell-cell communications between adjoining cells in multiple cell types, regulation of major physiological processes and disease pathogeneses. However, conventional connexin gene and protein classifications could be regarded as unsuitable in descriptions of comprehensive eutherian connexin gene data sets, due to ambiguities and inconsistencies in connexin gene and protein nomenclatures. Using eutherian comparative genomic analysis protocol and 35 public eutherian reference genomic sequence data sets, the present analysis attempted to update and revise comprehensive eutherian connexin gene data sets, and address and resolve major discrepancies in their descriptions. Among 631 potential coding sequences, the tests of reliability of eutherian public genomic sequences annotated, in aggregate, 349 connexin complete coding sequences. The most comprehensive curated eutherian connexin gene data set described 21 major gene clusters, 4 of which included evidence of differential gene expansions. For example, the present gene annotations initially described human CXNK1 gene and annotated 22 human connexin genes. Phylogenetic tree calculations and calculations of pairwise nucleotide sequence identity patterns proposed revised and updated phylogenetic classification of eutherian connexin genes. Therefore, the present study integrating gene annotations, phylogenetic analysis and protein molecular evolution analysis proposed new nomenclature of eutherian connexin genes and proteins.
Collapse
|
22
|
Sarrouilhe D, Mesnil M, Dejean C. Targeting Gap Junctions: New Insights into the Treatment of Major Depressive Disorder. Curr Med Chem 2019; 26:3775-3791. [DOI: 10.2174/0929867325666180327103530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/22/2017] [Accepted: 03/21/2018] [Indexed: 01/05/2023]
Abstract
Background:Major depressive disorder (MDD) is a multifactorial chronic and debilitating mood disease with high lifetime prevalence and associated with excess mortality. Treatments for this disease are not effective in all patients showing the need to find new therapeutic targets.Objective:This review aims to update our knowledge on the involvement of astroglial gap junctions and hemichannels in MDD and to show how they have become potential targets for the treatment of this pathology.Methods:The method applied in this review includes a systematic compilation of the relevant literature.Results and Conclusion:The use of rodent models of depression, gene analysis of hippocampal tissues of MDD patients and post-mortem studies on the brains from MDD patients suggest that astrocytic gap junction dysfunction may be a part of MDD etiologies. Chronic antidepressant treatments of rats, rat cultured cortical astrocytes and human astrocytoma cell lines support the hypothesis that the up-regulation of gap junctional coupling between astrocytes could be an underlying mechanism for the therapeutic effect of antidepressants. However, two recent functional studies suggest that connexin43 hemichannel activity is a part of several antidepressants’ mode of action and that astrocyte gap junctional intercellular communication and hemichannels exert different effects on antidepressant drug response. Even if they emerge as new therapeutic targets for new and more active treatments, further studies are needed to decipher the sophisticated and respective role of astrocytic gap junctions and hemichannels in MDD.
Collapse
Affiliation(s)
- Denis Sarrouilhe
- Laboratoire de Physiologie Humaine, Faculte de Medecine et Pharmacie, Universite de Poitiers, 6 rue de la Miletrie, Bat D1, TSA 51115, 86073 Poitiers, Cedex 9, France
| | - Marc Mesnil
- STIM, ERL 7003, CNRS-Universite de Poitiers, Pole Biologie Sante, Bat B36, TSA 51106, 1 rue Georges Bonnet, 86073 Poitiers, Cedex 9, France
| | - Catherine Dejean
- Service Pharmacie, Pavillon Janet, Centre Hospitalier Henri Laborit, 370 avenue Jacques Coeur, 86021 Poitiers Cedex, France
| |
Collapse
|
23
|
Pogoda K, Kameritsch P, Mannell H, Pohl U. Connexins in the control of vasomotor function. Acta Physiol (Oxf) 2019; 225:e13108. [PMID: 29858558 DOI: 10.1111/apha.13108] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 12/13/2022]
Abstract
Vascular endothelial cells, as well as smooth muscle cells, show heterogeneity with regard to their receptor expression and reactivity. For the vascular wall to act as a functional unit, the various cells' responses require integration. Such an integration is not only required for a homogeneous response of the vascular wall, but also for the vasomotor behaviour of consecutive segments of the microvascular arteriolar tree. As flow resistances of individual sections are connected in series, sections require synchronization and coordination to allow effective changes of conductivity and blood flow. A prerequisite for the local coordination of individual vascular cells and different sections of an arteriolar tree is intercellular communication. Connexins are involved in a dual manner in this coordination. (i) By forming gap junctions between cells, they allow an intercellular exchange of signalling molecules and electrical currents. In particular, the spread of electrical currents allows for coordination of cell responses over longer distances. (ii) Connexins are able to interact with other proteins to form signalling complexes. In this way, they can modulate and integrate individual cells' responses also in a channel-independent manner. This review outlines mechanisms allowing the vascular connexins to exert their coordinating function and to regulate the vasomotor reactions of blood vessels both locally, and in vascular networks. Wherever possible, we focus on the vasomotor behaviour of small vessels and arterioles which are the main vessels determining vascular resistance, blood pressure and local blood flow.
Collapse
Affiliation(s)
- K. Pogoda
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
| | - P. Kameritsch
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
| | - H. Mannell
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
| | - U. Pohl
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
- Munich Cluster for Systems Neurology (SyNergy); Munich Germany
| |
Collapse
|
24
|
Li Q, Li QQ, Jia JN, Liu ZQ, Zhou HH, Mao XY. Targeting gap junction in epilepsy: Perspectives and challenges. Biomed Pharmacother 2018; 109:57-65. [PMID: 30396092 DOI: 10.1016/j.biopha.2018.10.068] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
Gap junctions (GJs) are multiple cellular intercellular connections that allow ions to pass directly into the cytoplasm of neighboring cells. Electrical coupling mediated by GJs plays a role in the generation of highly synchronous electrical activity. Accumulative investigations show that GJs in the brain are involved in the generation, synchronization and maintenance of seizure events. At the same time, GJ blockers exert potent curative potential on epilepsy in vivo or in vitro. This review aims to shed light on the role of GJs in epileptogenesis. Targeting GJs is likely to be served as a novel therapeutic approach on epileptic patients.
Collapse
Affiliation(s)
- Qin Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Qiu-Qi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Ji-Ning Jia
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China.
| |
Collapse
|
25
|
Lynn BD, Li X, Hormuzdi SG, Griffiths EK, McGlade CJ, Nagy JI. E3 ubiquitin ligases LNX1 and LNX2 localize at neuronal gap junctions formed by connexin36 in rodent brain and molecularly interact with connexin36. Eur J Neurosci 2018; 48:3062-3081. [PMID: 30295974 DOI: 10.1111/ejn.14198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 08/31/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022]
Abstract
Electrical synapses in the mammalian central nervous system (CNS) are increasingly recognized as highly complex structures for mediation of neuronal communication, both with respect to their capacity for dynamic short- and long-term modification in efficacy of synaptic transmission and their multimolecular regulatory and structural components. These two characteristics are inextricably linked, such that understanding of mechanisms that contribute to electrical synaptic plasticity requires knowledge of the molecular composition of electrical synapses and the functions of proteins associated with these synapses. Here, we provide evidence that the key component of gap junctions that form the majority of electrical synapses in the mammalian CNS, namely connexin36 (Cx36), directly interacts with the related E3 ubiquitin ligase proteins Ligand of NUMB protein X1 (LNX1) and Ligand of NUMB protein X2 (LNX2). This is based on immunofluorescence colocalization of LNX1 and LNX2 with Cx36-containing gap junctions in adult mouse brain versus lack of such coassociation in LNX null mice, coimmunoprecipitation of LNX proteins with Cx36, and pull-down of Cx36 with the second PDZ domain of LNX1 and LNX2. Furthermore, cotransfection of cultured cells with Cx36 and E3 ubiquitin ligase-competent LNX1 and LNX2 isoforms led to loss of Cx36-containing gap junctions between cells, whereas these junctions persisted following transfection with isoforms of these proteins that lack ligase activity. Our results suggest that a LNX protein mediates ubiquitination of Cx36 at neuronal gap junctions, with consequent Cx36 internalization, and may thereby contribute to intracellular mechanisms that govern the recently identified modifiability of synaptic transmission at electrical synapses.
Collapse
Affiliation(s)
- Bruce D Lynn
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xinbo Li
- Casey Eye Institute, Oregon Health and Science University, Portland, Oregon
| | - Sheriar G Hormuzdi
- D'Arcy Thompson Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Emily K Griffiths
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - C Jane McGlade
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - James I Nagy
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
26
|
Robertson JM. The Gliocentric Brain. Int J Mol Sci 2018; 19:ijms19103033. [PMID: 30301132 PMCID: PMC6212929 DOI: 10.3390/ijms19103033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 09/23/2018] [Accepted: 09/28/2018] [Indexed: 01/10/2023] Open
Abstract
The Neuron Doctrine, the cornerstone of research on normal and abnormal brain functions for over a century, has failed to discern the basis of complex cognitive functions. The location and mechanisms of memory storage and recall, consciousness, and learning, remain enigmatic. The purpose of this article is to critically review the Neuron Doctrine in light of empirical data over the past three decades. Similarly, the central role of the synapse and associated neural networks, as well as ancillary hypotheses, such as gamma synchrony and cortical minicolumns, are critically examined. It is concluded that each is fundamentally flawed and that, over the past three decades, the study of non-neuronal cells, particularly astrocytes, has shown that virtually all functions ascribed to neurons are largely the result of direct or indirect actions of glia continuously interacting with neurons and neural networks. Recognition of non-neural cells in higher brain functions is extremely important. The strict adherence of purely neurocentric ideas, deeply ingrained in the great majority of neuroscientists, remains a detriment to understanding normal and abnormal brain functions. By broadening brain information processing beyond neurons, progress in understanding higher level brain functions, as well as neurodegenerative and neurodevelopmental disorders, will progress beyond the impasse that has been evident for decades.
Collapse
|
27
|
Lemcke H, David R. Potential mechanisms of microRNA mobility. Traffic 2018; 19:910-917. [PMID: 30058163 DOI: 10.1111/tra.12606] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 12/29/2022]
Abstract
microRNAs (miRNAs) are important epigenetic modulators of gene expression that control cellular physiology as well as tissue homeostasis, and development. In addition to the temporal aspects of miRNA-mediated gene regulation, the intracellular localization of miRNA is crucial for its silencing activity. Recent studies indicated that miRNA is even translocated between cells via gap junctional cell-cell contacts, allowing spatiotemporal modulation of gene expression within multicellular systems. Although non coding RNA remains a focus of intense research, studies regarding the intra-and intercellular mobility of small RNAs are still largely missing. Emerging data from experimental and computational work suggest the involvement of transport mechanisms governing proper localization of miRNA in single cells and cellular syncytia. Based on these data, we discuss a model of miRNA translocation that could help to address the spatial aspects of miRNA function and the impact of miRNA molecules on the intercellular signaling network.
Collapse
Affiliation(s)
- Heiko Lemcke
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), University of Rostock, Rostock, Germany.,Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany
| | - Robert David
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), University of Rostock, Rostock, Germany.,Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany
| |
Collapse
|
28
|
A Cell Junctional Protein Network Associated with Connexin-26. Int J Mol Sci 2018; 19:ijms19092535. [PMID: 30150563 PMCID: PMC6163694 DOI: 10.3390/ijms19092535] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
GJB2 mutations are the leading cause of non-syndromic inherited hearing loss. GJB2 encodes connexin-26 (CX26), which is a connexin (CX) family protein expressed in cochlea, skin, liver, and brain, displaying short cytoplasmic N-termini and C-termini. We searched for CX26 C-terminus binding partners by affinity capture and identified 12 unique proteins associated with cell junctions or cytoskeleton (CGN, DAAM1, FLNB, GAPDH, HOMER2, MAP7, MAPRE2 (EB2), JUP, PTK2B, RAI14, TJP1, and VCL) by using mass spectrometry. We show that, similar to other CX family members, CX26 co-fractionates with TJP1, VCL, and EB2 (EB1 paralogue) as well as the membrane-associated protein ASS1. The adaptor protein CGN (cingulin) co-immuno-precipitates with CX26, ASS1, and TJP1. In addition, CGN co-immunoprecipitation with CX30, CX31, and CX43 indicates that CX association is independent on the CX C-terminus length or sequence. CX26, CGN, FLNB, and DAMM1 were shown to distribute to the organ of Corti and hepatocyte plasma membrane. In the mouse liver, CX26 and TJP1 co-localized at the plasma membrane. In conclusion, CX26 associates with components of other membrane junctions that integrate with the cytoskeleton.
Collapse
|
29
|
Nagy JI, Lynn BD. Structural and Intermolecular Associations Between Connexin36 and Protein Components of the Adherens Junction-Neuronal Gap Junction Complex. Neuroscience 2018; 384:241-261. [PMID: 29879437 DOI: 10.1016/j.neuroscience.2018.05.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 11/20/2022]
Abstract
Intimate structural and functional relationships between gap junctions and adherens junctions have been demonstrated in peripheral tissues, but have not been thoroughly examined in the central nervous system, where adherens junctions are often found in close proximity to neuronal gap junctions. Here, we used immunofluorescence approaches to document the localization of various protein components of adherens junctions in relation to those that we have previously reported to occur at electrical synapses formed by neuronal gap junctions composed of connexin36 (Cx36). The adherens junction constituents N-cadherin and nectin-1 were frequently found to localize near or overlap with Cx36-containing gap junctions in several brain regions examined. This was also true of the adherens junction-associated proteins α-catenin and β-catenin, as well as the proteins zonula occludens-1 and AF6 (aka, afadin) that were reported constituents of both adherens junctions and gap junctions. The deployment of the protein constituents of these junctions was especially striking at somatic contacts between primary afferent neurons in the mesencephalic trigeminal nucleus (MesV), where the structural components of adherens junctions appeared to be maintained in connexin36 null mice. These results support emerging views concerning the multi-molecular composition of electrical synapses and raise possibilities for various structural and functional protein-protein interactions at what now can be considered the adherens junction-neuronal gap junction complex. Further, the results point to intracellular signaling pathways that could potentially contribute to the assembly, maintenance and turnover of this complex, as well as to the dynamic nature of neuronal communication at electrical synapses.
Collapse
Affiliation(s)
- J I Nagy
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - B D Lynn
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
30
|
Direct Cell⁻Cell Interactions in the Endometrium and in Endometrial Pathophysiology. Int J Mol Sci 2018; 19:ijms19082227. [PMID: 30061539 PMCID: PMC6121364 DOI: 10.3390/ijms19082227] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Cell contacts exhibit a considerable influence on tissue physiology and homeostasis by controlling paracellular and intercellular transport processes, as well as by affecting signaling pathways. Since they maintain cell polarity, they play an important role in cell plasticity. The knowledge about the junctional protein families and their interactions has increased considerably during recent years. In contrast to most other tissues, the endometrium undergoes extensive physiological changes and reveals an extraordinary plasticity due to its crucial role in the establishment and maintenance of pregnancy. These complex changes are accompanied by changes in direct cell–cell contacts to meet the various requirements in the respective developmental stage. Impairment of this sophisticated differentiation process may lead to failure of implantation and embryo development and may be involved in the pathogenesis of endometrial diseases. In this article, we focus on the knowledge about the distribution and regulation of the different junctional proteins in the endometrium during cycling and pregnancy, as well as in pathologic conditions such as endometriosis and cancer. Decoding these sophisticated interactions should improve our understanding of endometrial physiology as well as of the mechanisms involved in pathological conditions.
Collapse
|
31
|
Connexins and Pannexins: Important Players in Tumorigenesis, Metastasis and Potential Therapeutics. Int J Mol Sci 2018; 19:ijms19061645. [PMID: 29865195 PMCID: PMC6032133 DOI: 10.3390/ijms19061645] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
Since their characterization more than five decades ago, gap junctions and their structural proteins-the connexins-have been associated with cancer cell growth. During that period, the accumulation of data and molecular knowledge about this association revealed an apparent contradictory relationship between them and cancer. It appeared that if gap junctions or connexins can down regulate cancer cell growth they can be also implied in the migration, invasion and metastatic dissemination of cancer cells. Interestingly, in all these situations, connexins seem to be involved through various mechanisms in which they can act either as gap-junctional intercellular communication mediators, modulators of signalling pathways through their interactome, or as hemichannels, which mediate autocrine/paracrine communication. This complex involvement of connexins in cancer progression is even more complicated by the fact that their hemichannel function may overlap with other gap junction-related proteins, the pannexins. Despite this complexity, the possible involvements of connexins and pannexins in cancer progression and the elucidation of the mechanisms they control may lead to use them as new targets to control cancer progression. In this review, the involvements of connexins and pannexins in these different topics (cancer cell growth, invasion/metastasis process, possible cancer therapeutic targets) are discussed.
Collapse
|
32
|
Connexin 43 regulates the expression of wound healing-related genes in human gingival and skin fibroblasts. Exp Cell Res 2018; 367:150-161. [PMID: 29596891 DOI: 10.1016/j.yexcr.2018.03.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/23/2018] [Accepted: 03/24/2018] [Indexed: 12/21/2022]
Abstract
Fibroblasts are the most abundant connective tissue cells and play an important role in wound healing. It is possible that faster and scarless wound healing in oral mucosal gingiva relative to skin may relate to the distinct phenotype of the fibroblasts residing in these tissues. Connexin 43 (Cx43) is the most ubiquitous Cx in skin (SFBLs) and gingival fibroblasts (GFBLs), and assembles into hemichannels (HCs) and gap junctions (GJs) on the cell membrane. We hypothesized that SFBLs and GFBLs display distinct expression or function of Cx43, and that this may partly underlie the different wound healing outcomes in skin and gingiva. Here we show that Cx43 distinctly formed Cx43 GJs and HCs in human skin and gingiva in vivo. However, in SFBLs, in contrast to GFBLs, only a small proportion of total Cx43 assembled into HC plaques. Using an in vivo-like 3D culture model, we further show that the GJ, HC, and channel-independent functions of Cx43 distinctly regulated wound healing-related gene expression in GFBLs and SFBLs. Therefore, the distinct wound healing outcomes in skin and gingiva may partly relate to the inherently different assembly and function of Cx43 in the resident fibroblasts.
Collapse
|
33
|
Joo HC, Choi JW, Moon H, Lee CY, Yoo KJ, Kim SW, Hwang KC. Protective effects of kenpaullone on cardiomyocytes following H 2O 2-induced oxidative stress are attributed to inhibition of connexin 43 degradation by SGSM3. Biochem Biophys Res Commun 2018; 499:368-373. [PMID: 29577900 DOI: 10.1016/j.bbrc.2018.03.166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 03/22/2018] [Indexed: 02/08/2023]
Abstract
A previous study showed that small G protein signaling modulator 3 (SGSM3) was highly correlated with Cx43 in heart functions and that high levels of SGSM3 may induce Cx43 turnover through lysosomal degradation in infarcted rat hearts. Here, we investigated the protective effects of kenpaullone on cardiomyocytes following H2O2-induced oxidative stress mediated by the interaction of SGSM3 with Cx43. We found that the gap junction protein Cx43 was significantly down-regulated in an H2O2 concentration-dependent manner, whereas expression of SGSM3 was up-regulated upon H2O2 exposure in H9c2 cells. The effect of kenpaullone pretreatment on H2O2-induced cytotoxicity was evaluated in H9c2 cells. H2O2 markedly increased the release of lactate dehydrogenase (LDH), while kenpaullone pretreatment suppressed LDH release in H9c2 cells. Moreover, kenpaullone pretreatment significantly reduced ROS fluorescence intensity and significantly down-regulated the level of apoptosis-activating genes (cleaved caspase-3, cleaved caspase-9 and cytochrome C), autophagy markers (LC3A/B), and the Cx43-interacting partner SGSM3. These results suggest that kenpaullone plays a role in protecting cardiomyocytes from oxidative stress and that the turnover of Cx43 through SGSM3-induced lysosomal degradation underlies the anti-apoptotic effect of kenpaullone.
Collapse
Affiliation(s)
- Hyun-Chel Joo
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Jung-Won Choi
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 210-701, Republic of Korea
| | - Hanbyeol Moon
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 210-701, Republic of Korea
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kyung-Jong Yoo
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 210-701, Republic of Korea; Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 404-834, Republic of Korea.
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 210-701, Republic of Korea; Catholic Kwandong University, International St. Mary's Hospital, Incheon Metropolitan City, 404-834, Republic of Korea.
| |
Collapse
|
34
|
Connexin 43 Controls the Astrocyte Immunoregulatory Phenotype. Brain Sci 2018; 8:brainsci8040050. [PMID: 29565275 PMCID: PMC5924386 DOI: 10.3390/brainsci8040050] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 01/03/2023] Open
Abstract
Astrocytes are the most abundant glial cells of the central nervous system and have recently been recognized as crucial in the regulation of brain immunity. In most neuropathological conditions, astrocytes are prone to a radical phenotypical change called reactivity, which plays a key role in astrocyte contribution to neuroinflammation. However, how astrocytes regulate brain immunity in healthy conditions is an understudied question. One of the astroglial molecule involved in these regulations might be Connexin 43 (Cx43), a gap junction protein highly enriched in astrocyte perivascular endfeet-terminated processes forming the glia limitans. Indeed, Cx43 deletion in astrocytes (Cx43KO) promotes a continuous immune recruitment and an autoimmune response against an astrocyte protein, without inducing any brain lesion. To investigate the molecular basis of this unique immune response, we characterized the polysomal transcriptome of hippocampal astrocytes deleted for Cx43. Our results demonstrate that, in the absence of Cx43, astrocytes adopt an atypical reactive status with no change in most canonical astrogliosis markers, but with an upregulation of molecules promoting immune recruitment, complement activation as well as anti-inflammatory processes. Intriguingly, while several of these upregulated transcriptional events suggested an activation of the γ-interferon pathway, no increase in this cytokine or activation of related signaling pathways were found in Cx43KO. Finally, deletion of astroglial Cx43 was associated with the upregulation of several angiogenic factors, consistent with an increase in microvascular density in Cx43KO brains. Collectively, these results strongly suggest that Cx43 controls immunoregulatory and angiogenic properties of astrocytes.
Collapse
|
35
|
Bachelot A, Gilleron J, Meduri G, Guberto M, Dulon J, Boucherie S, Touraine P, Misrahi M. A common African variant of human connexin 37 is associated with Caucasian primary ovarian insufficiency and has a deleterious effect in vitro. Int J Mol Med 2018; 41:640-648. [PMID: 29207017 PMCID: PMC5752242 DOI: 10.3892/ijmm.2017.3257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/20/2017] [Indexed: 11/06/2022] Open
Abstract
Folliculogenesis requires communication between granulosa cells and oocytes, mediated by connexin-based gap junctions. Connexin 37 (Cx37)-deficient female mice are infertile. The present study assessed Cx37 deficiency in patients with primary ovarian insufficiency (POI). A candidate gene study was performed in patients and controls from the National Genotyping Center (Evry, France) including 58 Caucasian patients with idiopathic isolated POI and 142 Caucasian controls. Direct genomic sequencing of the coding regions of the GJA4 gene (encoding Cx37) was performed with the aim to identify a deleterious variant associated with POI and absent in ethnically matched controls. A single Cx37 variant absent in the control population was identified, namely a c.946G>A heterozygous substitution leading to a p.Gly316Ser variant that was present in two POI patients. This variant was absent in all Caucasian controls from various databases, and has been observed exclusively in African populations. This variant was identified to have a dominant negative effect in HeLa cells in vitro to alter connexon function (by 67.2±7.17%), as determined by Gap-fluorescence recovery after photobleaching. The alteration principally resulted from a decrease of cell surface connexons due to altered trafficking (by 47.73±8.59%). In marked contrast to this observation, a p.Pro258Ser variant frequent in all ethnic populations in databases had no functional effect in vitro. In conclusion, the present study reported on a Cx37 variant in two Caucasian POI patients, which was absent in control Caucasian populations, and which had a deleterious effect in vitro. It is therefore suggested that in the genetic context of the Caucasian population, this variant may contribute to POI.
Collapse
Affiliation(s)
- Anne Bachelot
- AP-HP, Department of Endocrinology and Reproductive Medicine, Pitié-Salpêtrière Hospital, Reference Center for Rare Endocrine Diseases of Growth, Reference Center for Rare Gynecological Pathologies
- University Pierre and Marie Curie, University Paris 6, F-75013 Paris
| | - Jerome Gilleron
- National Institute of Health and Medical Research INSERM U1065 - University of Nice-Sophia Antipolis, Mediterranean Center for Molecular Medicine C3M, F-06000 Nice
| | - Geri Meduri
- National Institute of Health and Medical Research INSERM U1195
| | - Mihelai Guberto
- University Paris-Sud, University Paris Saclay, Medical Faculty Paris-Sud, Bicêtre Hospital, F-94275 Le Kremlin Bicêtre
| | - Jerome Dulon
- AP-HP, Department of Endocrinology and Reproductive Medicine, Pitié-Salpêtrière Hospital, Reference Center for Rare Endocrine Diseases of Growth, Reference Center for Rare Gynecological Pathologies
| | - Sylviane Boucherie
- National Institute of Health and Medical Research UMR-S 757 INSERM, University Paris-Sud, F-91400 Orsay, France
| | - Philippe Touraine
- AP-HP, Department of Endocrinology and Reproductive Medicine, Pitié-Salpêtrière Hospital, Reference Center for Rare Endocrine Diseases of Growth, Reference Center for Rare Gynecological Pathologies
- University Pierre and Marie Curie, University Paris 6, F-75013 Paris
| | - Micheline Misrahi
- University Paris-Sud, University Paris Saclay, Medical Faculty Paris-Sud, Bicêtre Hospital, F-94275 Le Kremlin Bicêtre
| |
Collapse
|
36
|
Nagy JI, Pereda AE, Rash JE. Electrical synapses in mammalian CNS: Past eras, present focus and future directions. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2018; 1860:102-123. [PMID: 28577972 PMCID: PMC5705454 DOI: 10.1016/j.bbamem.2017.05.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 12/19/2022]
Abstract
Gap junctions provide the basis for electrical synapses between neurons. Early studies in well-defined circuits in lower vertebrates laid the foundation for understanding various properties conferred by electrical synaptic transmission. Knowledge surrounding electrical synapses in mammalian systems unfolded first with evidence indicating the presence of gap junctions between neurons in various brain regions, but with little appreciation of their functional roles. Beginning at about the turn of this century, new approaches were applied to scrutinize electrical synapses, revealing the prevalence of neuronal gap junctions, the connexin protein composition of many of those junctions, and the myriad diverse neural systems in which they occur in the mammalian CNS. Subsequent progress indicated that electrical synapses constitute key elements in synaptic circuitry, govern the collective activity of ensembles of electrically coupled neurons, and in part orchestrate the synchronized neuronal network activity and rhythmic oscillations that underlie fundamental integrative processes. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- James I Nagy
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - John E Rash
- Department of Biomedical Sciences, and Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
37
|
Pogoda K, Mannell H, Blodow S, Schneider H, Schubert KM, Qiu J, Schmidt A, Imhof A, Beck H, Tanase LI, Pfeifer A, Pohl U, Kameritsch P. NO Augments Endothelial Reactivity by Reducing Myoendothelial Calcium Signal Spreading: A Novel Role for Cx37 (Connexin 37) and the Protein Tyrosine Phosphatase SHP-2. Arterioscler Thromb Vasc Biol 2017; 37:2280-2290. [PMID: 29025706 DOI: 10.1161/atvbaha.117.309913] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/26/2017] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Because of its strategic position between endothelial and smooth muscle cells in microvessels, Cx37 (Connexin 37) plays an important role in myoendothelial gap junctional intercellular communication. We have shown before that NO inhibits gap junctional intercellular communication through gap junctions containing Cx37. However, the underlying mechanism is not yet identified. APPROACH AND RESULTS Using channel-forming Cx37 mutants exhibiting partial deletions or amino acid exchanges in their C-terminal loops, we now show that the phosphorylation state of a tyrosine residue at position 332 (Y332) in the C-terminus of Cx37 controls the gap junction-dependent spread of calcium signals. Mass spectra revealed that NO protects Cx37 from dephosphorylation at Y332 by inhibition of the protein tyrosine phosphatase SHP-2. Functionally, the inhibition of gap junctional intercellular communication by NO decreased the spread of the calcium signal (induced by mechanical stimulation of individual endothelial cells) from endothelial to smooth muscle cells in intact vessels, while, at the same time, augmenting the calcium signal spreading within the endothelium. Consequently, preincubation of small resistance arteries with exogenous NO enhanced the endothelium-dependent dilator response to acetylcholine in spite of a pharmacological blockade of NO-dependent cGMP formation by the soluable guanylyl cyclase inhibitor ODQ (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one). CONCLUSIONS Our results identify a novel mechanism by which NO can increase the efficacy of calcium, rising vasoactive agonists in the microvascular endothelium.
Collapse
Affiliation(s)
- Kristin Pogoda
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Hanna Mannell
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Stephanie Blodow
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Holger Schneider
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Kai Michael Schubert
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Jiehua Qiu
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Andreas Schmidt
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Axel Imhof
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Heike Beck
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Laurentia Irina Tanase
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Alexander Pfeifer
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Ulrich Pohl
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.).
| | - Petra Kameritsch
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| |
Collapse
|
38
|
Kanapathy M, Simpson R, Madden L, Thrasivoulou C, Mosahebi A, Becker DL, Richards T. Upregulation of epidermal gap junctional proteins in patients with venous disease. Br J Surg 2017; 105:59-67. [DOI: 10.1002/bjs.10653] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/05/2017] [Accepted: 06/23/2017] [Indexed: 11/09/2022]
Abstract
Abstract
Background
Leg ulceration is a feared complication of venous insufficiency. It is not known whether varicose veins predispose skin to poor wound healing. The expression pattern of gap junctional protein connexin, a known marker of poor wound healing, was investigated across various stages of venous disease.
Methods
Patients undergoing intervention for varicose veins were assessed according to the Clinical Etiologic Anatomic Pathophysiologic (CEAP) classification of varicose veins. Paired 4-mm punch biopsies were taken from above the ankle (pathological) and above the knee (control). Tissues were stained with haematoxylin and eosin, and for connexin 43, connexin 30 and connexin 26.
Results
Forty-eight paired biopsies were taken (12 each for CEAP class C0, C2, C4 and C6). The pathological skin showed progressive epithelial hyperthickening, an increase in the number and depth of rete ridges, increased inflammation and loss of dermal architecture with disease progression from C4 onwards. The overall absolute connexin expression and mean connexin expression per cell in the pathological skin similarly increased across the CEAP classes from as early as C2. Increasing levels of connexin in control skin were also noted, indicating progression of the disease proximally. Connexin 43 expression showed the strongest positive correlation between pathological and control skin.
Conclusion
Connexins were overexpressed in patients with simple varicose veins, with a stepwise increased expression through venous eczema to ulceration. Connexin 43 is a potential biomarker for venous disease. This finding suggests that varicose veins predispose skin to poor wound healing. Surgical relevanceThe overexpression of connexins, a family of gap junctional proteins, is known to cause poor healing in venous leg ulceration. It is not known whether there is any association with superficial venous disease. Here, connexin proteins were overexpressed in patients with uncomplicated varicose veins, before histological skin changes. Connexin could be a biomarker of venous disease progression.
Collapse
Affiliation(s)
- M Kanapathy
- Division of Surgery and Interventional Science, University College London, London, UK
- Department of Plastic and Reconstructive Surgery, Royal Free London NHS Foundation Trust Hospital, London, UK
- Tissue Repair and Regeneration Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - R Simpson
- Division of Surgery and Interventional Science, University College London, London, UK
| | - L Madden
- Tissue Repair and Regeneration Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - C Thrasivoulou
- Department of Cell and Developmental Biology, University College London, London, UK
| | - A Mosahebi
- Division of Surgery and Interventional Science, University College London, London, UK
- Department of Plastic and Reconstructive Surgery, Royal Free London NHS Foundation Trust Hospital, London, UK
| | - D L Becker
- Tissue Repair and Regeneration Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Institute of Medical Biology, A*Star, Immunos, Biomedical Grove, Singapore
| | - T Richards
- Division of Surgery and Interventional Science, University College London, London, UK
| |
Collapse
|
39
|
Morioka N, Fujii S, Kondo S, Zhang FF, Miyauchi K, Nakamura Y, Hisaoka-Nakashima K, Nakata Y. Downregulation of spinal astrocytic connexin43 leads to upregulation of interleukin-6 and cyclooxygenase-2 and mechanical hypersensitivity in mice. Glia 2017; 66:428-444. [DOI: 10.1002/glia.23255] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Norimitsu Morioka
- Department of Pharmacology; Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi; Minami-ku Hiroshima 734-8553 Japan
| | - Shiori Fujii
- Department of Pharmacology; Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi; Minami-ku Hiroshima 734-8553 Japan
| | - Syun Kondo
- Department of Pharmacology; Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi; Minami-ku Hiroshima 734-8553 Japan
| | - Fang Fang Zhang
- Department of Pharmacology; Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi; Minami-ku Hiroshima 734-8553 Japan
- Institute of Pharmacology, Taishan Medical University, 619 Changcheng Road; Taian Shandong 271016 China
| | - Kazuki Miyauchi
- Department of Pharmacology; Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi; Minami-ku Hiroshima 734-8553 Japan
| | - Yoki Nakamura
- Department of Pharmacology; Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi; Minami-ku Hiroshima 734-8553 Japan
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse IRP, Triad Suite 3305, 333 Cassell Drive; Baltimore MD 21224
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology; Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi; Minami-ku Hiroshima 734-8553 Japan
| | - Yoshihiro Nakata
- Department of Pharmacology; Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi; Minami-ku Hiroshima 734-8553 Japan
| |
Collapse
|
40
|
Thévenin AF, Margraf RA, Fisher CG, Kells-Andrews RM, Falk MM. Phosphorylation regulates connexin43/ZO-1 binding and release, an important step in gap junction turnover. Mol Biol Cell 2017; 28:3595-3608. [PMID: 29021339 PMCID: PMC5706988 DOI: 10.1091/mbc.e16-07-0496] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 12/11/2022] Open
Abstract
To investigate whether connexin phosphorylation regulates the known role of zonula occludens-1 protein (ZO-1) in gap junction (GJ) function, we generated and analyzed a series of phosphomimetic and phosphorylation-dead mutants by mutating known conserved regulatory serine (S) residues 255, 279/282, 365, 368, and 373 located in the C-terminal domain of connexin43 (Cx43) into glutamic acid (E) or alanine (A) residues. All connexin mutants were translated into stable, full-length proteins and assembled into GJs when expressed in HeLa or Madin-Darby canine kidney epithelial cells. However, mutants with S residues exchanged at positions 365, 368, and 373 exhibited a significantly altered ZO-1 interaction profile, while mutants with S residues exchanged at 255 and 279/282 did not. Unlike wild-type Cx43, in which ZO-1 binding is restricted to the periphery of GJ plaques, S365A, S365E, S368A, S368E, and S373A mutants bound ZO-1 throughout the GJ plaques, while the S373E mutant did not bind ZO-1 at all. Inability to disengage from ZO-1 correlated with increased GJ plaque size and increased connexin protein half-life, while maintaining GJ channels in an open, functional state. Quantitative clathrin-binding analyses revealed no significant alterations in clathrin-binding efficiency, suggesting that the inability to disengage from ZO-1 prevented maturation of functional into nonfunctional/endocytic channels, rather than ZO-1 interfering with GJ endocytosis directly. Collectively, our results indicate that ZO-1 binding regulates channel accrual, while disengagement from ZO-1 is critical for GJ channel closure and transitioning GJ channels for endocytosis. Intriguingly, these transitional ZO-1 binding/release and channel-aging steps are mediated by a series of hierarchical phosphorylation/dephosphorylation events at S373, S365, and S368, well-known Cx43 Akt, protein kinase A, and protein kinase C phosphorylation sites located in the vicinity of the ZO-1 binding site.
Collapse
Affiliation(s)
| | - Rachel A Margraf
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015
| | - Charles G Fisher
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015
| | | | - Matthias M Falk
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015
| |
Collapse
|
41
|
Cardiotoxic Effects of Short-Term Doxorubicin Administration: Involvement of Connexin 43 in Calcium Impairment. Int J Mol Sci 2017; 18:ijms18102121. [PMID: 29019935 PMCID: PMC5666803 DOI: 10.3390/ijms18102121] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 09/29/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
The use of Doxorubicin (DOXO), a potent antineoplastic agent, is limited by the development of cardiotoxicity. DOXO-induced cardiotoxicity is multifactorial, although alterations in calcium homeostasis, seem to be involved. Since even the Connexin43 (Cx43) plays a pivotal role in these two phenomena, in this study we have analyzed the effects of DOXO on Cx43 expression and localization. Damage caused by anthracyclines on cardiomyocytes is immediate after each injection, in the present study we used a short-term model of DOXO-induced cardiomyopathy. C57BL/6j female mice were randomly divided in groups and injected with DOXO (2 or 10 mg/kg i.p.) for 1–3 or 7 days once every other day. Cardiac function was assessed by Echocardiography. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCAII) and phospholamban (PLB) expression were assessed by Western blot analysis, intracellular [Ca2+] were detected spectrofluorometrically by means of Fura-2 pentakis (acetoxymethyl) ester (FURA-2AM), and Cx43 and pCx43 expression and localization was analyzed by Western blot and confirmed by immunofluorescence analysis. DOXO induces impairment in Ca2+ homeostasis, already evident after a single administration, and affects Cx43 expression and localization. Our data suggest that DOXO-induced alterations in Ca2+ homeostasis causes in the cells the induction of compensatory mechanisms until a certain threshold, above which cardiac injury is triggered.
Collapse
|
42
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
43
|
Lee CY, Choi JW, Shin S, Lee J, Seo HH, Lim S, Lee S, Joo HC, Kim SW, Hwang KC. Interaction of small G protein signaling modulator 3 with connexin 43 contributes to myocardial infarction in rat hearts. Biochem Biophys Res Commun 2017; 491:429-435. [DOI: 10.1016/j.bbrc.2017.07.081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 11/29/2022]
|
44
|
Sato H, Coburn J. Leptospira interrogans causes quantitative and morphological disturbances in adherens junctions and other biological groups of proteins in human endothelial cells. PLoS Negl Trop Dis 2017; 11:e0005830. [PMID: 28750011 PMCID: PMC5549773 DOI: 10.1371/journal.pntd.0005830] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/08/2017] [Accepted: 07/24/2017] [Indexed: 12/21/2022] Open
Abstract
Pathogenic Leptospira transmits from animals to humans, causing the zoonotic life-threatening infection called leptospirosis. This infection is reported worldwide with higher risk in tropical regions. Symptoms of leptospirosis range from mild illness to severe illness such as liver damage, kidney failure, respiratory distress, meningitis, and fatal hemorrhagic disease. Invasive species of Leptospira rapidly disseminate to multiple tissues where this bacterium damages host endothelial cells, increasing vascular permeability. Despite the burden in humans and animals, the pathogenic mechanisms of Leptospira infection remain to be elucidated. The pathogenic leptospires adhere to endothelial cells and permeabilize endothelial barriers in vivo and in vitro. In this study, human endothelial cells were infected with the pathogenic L. interrogans serovar Copenhageni or the saprophyte L. biflexa serovar Patoc to investigate morphological changes and other distinctive phenotypes of host cell proteins by fluorescence microscopy. Among those analyzed, 17 proteins from five biological classes demonstrated distinctive phenotypes in morphology and/or signal intensity upon infection with Leptospira. The affected biological groups include: 1) extracellular matrix, 2) intercellular adhesion molecules and cell surface receptors, 3) intracellular proteins, 4) cell-cell junction proteins, and 5) a cytoskeletal protein. Infection with the pathogenic strain most profoundly disturbed the biological structures of adherens junctions (VE-cadherin and catenins) and actin filaments. Our data illuminate morphological disruptions and reduced signals of cell-cell junction proteins and filamentous actin in L. interrogans-infected endothelial cells. In addition, Leptospira infection, regardless of pathogenic status, influenced other host proteins belonging to multiple biological classes. Our data suggest that this zoonotic agent may damage endothelial cells via multiple cascades or pathways including endothelial barrier damage and inflammation, potentially leading to vascular hyperpermeability and severe illness in vivo. This work provides new insights into the pathophysiological mechanisms of Leptospira infection.
Collapse
Affiliation(s)
- Hiromi Sato
- Center for Infectious Disease Research, Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Jenifer Coburn
- Center for Infectious Disease Research, Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States of America
- * E-mail:
| |
Collapse
|
45
|
Hu Z, Shi W, Riquelme MA, Shi Q, Biswas S, Lo WK, White TW, Gu S, Jiang JX. Connexin 50 Functions as an Adhesive Molecule and Promotes Lens Cell Differentiation. Sci Rep 2017; 7:5298. [PMID: 28706245 PMCID: PMC5509658 DOI: 10.1038/s41598-017-05647-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 06/01/2017] [Indexed: 11/25/2022] Open
Abstract
Connexins play essential roles in lens homeostasis and development. Here, we identified a new role for Cx50 that mediates cell-cell adhesion function. Cx50 enhanced the adhesive capability of AQP0. Interestingly, the expression of Cx50 alone promoted cell adhesion at a comparable level to AQP0; however, this cell adhesive function was not observed with other lens connexins, Cx43 and Cx46. Moreover, the adhesive property occurred in both homotypic with Cx50 expressed in both pairing cells and heterotypic with Cx50 in only one pairing cell, and this function appears to be unrelated to its role in forming gap junction channels. Cx50 KO lenses exhibited increased intercellular spaces between lens fiber cells. The second extracellular loop domain (E2) is primarily responsible for this adhesive function. Treatment with a fusion protein containing E2 domain inhibited cell adhesion. Furthermore, disruption of cell adhesion by the E2 domains impaired primary lens cell differentiation. Five critical amino acid residues in the E2 domain primarily are involved in cell adhesive function as well as lens epithelial-fiber differentiation. Together, these results suggest that in addition to forming gap junction channels, Cx50 acts as an adhesive molecule that is critical in maintaining lens fiber integrity and epithelial-fiber differentiation.
Collapse
Affiliation(s)
- Zhengping Hu
- Departments of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen Shi
- Departments of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Manuel A Riquelme
- Departments of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Qian Shi
- Departments of Physiology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Sondip Biswas
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Woo-Kuen Lo
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Thomas W White
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Sumin Gu
- Departments of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jean X Jiang
- Departments of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
46
|
Ledee D, Kang MA, Kajimoto M, Purvine S, Brewer H, Pasa-Tolic L, Portman MA. Quantitative cardiac phosphoproteomics profiling during ischemia-reperfusion in an immature swine model. Am J Physiol Heart Circ Physiol 2017; 313:H125-H137. [PMID: 28455290 PMCID: PMC5538860 DOI: 10.1152/ajpheart.00842.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 01/26/2023]
Abstract
Ischemia-reperfusion (I/R) results in altered metabolic and molecular responses, and phosphorylation is one of the most noted regulatory mechanisms mediating signaling mechanisms during physiological stresses. To expand our knowledge of the potential phosphoproteomic changes in the myocardium during I/R, we used Isobaric Tags for Relative and Absolute Quantitation-based analyses in left ventricular samples obtained from porcine hearts under control or I/R conditions. The data are available via ProteomeXchange with identifier PXD006066. We identified 1,896 phosphopeptides within left ventricular control and I/R porcine samples. Significant differential phosphorylation between control and I/R groups was discovered in 111 phosphopeptides from 86 proteins. Analysis of the phosphopeptides using Motif-x identified five motifs: (..R..S..), (..SP..), (..S.S..), (..S…S..), and (..S.T..). Semiquantitative immunoblots confirmed site location and directional changes in phosphorylation for phospholamban and pyruvate dehydrogenase E1, two proteins known to be altered by I/R and identified by this study. Novel phosphorylation sites associated with I/R were also identified. Functional characterization of the phosphopeptides identified by our methodology could expand our understanding of the signaling mechanisms involved during I/R damage in the heart as well as identify new areas to target therapeutic strategies.NEW & NOTEWORTHY We used Isobaric Tags for Relative and Absolute Quantitation technology to investigate the phosphoproteomic changes that occur in cardiac tissue under ischemia-reperfusion conditions. The results of this study provide an extensive catalog of phosphoproteins, both predicted and novel, associated with ischemia-reperfusion, thereby identifying new pathways for investigation.
Collapse
Affiliation(s)
- Dolena Ledee
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington
- Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, Washington
| | - Min A Kang
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington
| | - Masaki Kajimoto
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington
| | - Samuel Purvine
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington; and
| | - Heather Brewer
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington; and
| | - Ljiljana Pasa-Tolic
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington; and
| | - Michael A Portman
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington;
- Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, Washington
| |
Collapse
|
47
|
Multiple and complex influences of connexins and pannexins on cell death. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017. [PMID: 28625689 DOI: 10.1016/j.bbamem.2017.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell death is a fundamental process for organogenesis, immunity and cell renewal. During the last decades a broad range of molecular tools were identified as important players for several different cell death pathways (apoptosis, pyroptosis, necrosis, autosis…). Aside from these direct regulators of cell death programs, several lines of evidence proposed connexins and pannexins as potent effectors of cell death. In the present review we discussed the potential roles played by connexins, pannexins and innexins in the different cell death programs at different scales from gap junction intercellular communication to protein-protein interactions. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
|
48
|
Roy S, Jiang JX, Li AF, Kim D. Connexin channel and its role in diabetic retinopathy. Prog Retin Eye Res 2017; 61:35-59. [PMID: 28602949 DOI: 10.1016/j.preteyeres.2017.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022]
Abstract
Diabetic retinopathy is the leading cause of blindness in the working age population. Unfortunately, there is no cure for this devastating ocular complication. The early stage of diabetic retinopathy is characterized by the loss of various cell types in the retina, namely endothelial cells and pericytes. As the disease progresses, vascular leakage, a clinical hallmark of diabetic retinopathy, becomes evident and may eventually lead to diabetic macular edema, the most common cause of vision loss in diabetic retinopathy. Substantial evidence indicates that the disruption of connexin-mediated cellular communication plays a critical role in the pathogenesis of diabetic retinopathy. Yet, it is unclear how altered communication via connexin channel mediated cell-to-cell and cell-to-extracellular microenvironment is linked to the development of diabetic retinopathy. Recent observations suggest the possibility that connexin hemichannels may play a role in the pathogenesis of diabetic retinopathy by allowing communication between cells and the microenvironment. Interestingly, recent studies suggest that connexin channels may be involved in regulating retinal vascular permeability. These cellular events are coordinated at least in part via connexin-mediated intercellular communication and the maintenance of retinal vascular homeostasis. This review highlights the effect of high glucose and diabetic condition on connexin channels and their impact on the development of diabetic retinopathy.
Collapse
Affiliation(s)
- Sayon Roy
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, Boston, MA, United States.
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - An-Fei Li
- Department of Ophthalmology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan
| | - Dongjoon Kim
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
49
|
Wang SG, Tsao DD, Vanderpool KG, Yasumura T, Rash JE, Nagy JI. Connexin36 localization to pinealocytes in the pineal gland of mouse and rat. Eur J Neurosci 2017; 45:1594-1605. [PMID: 28474748 PMCID: PMC5507615 DOI: 10.1111/ejn.13602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 01/07/2023]
Abstract
Several cell types in the pineal gland are known to establish intercellular gap junctions, but the connexin constituents of those junctions have not been fully characterized. Specifically, the expression of connexin36 (Cx36) protein and mRNA has been examined in the pineal, but the identity of cells that produce Cx36 and that form Cx36-containing gap junctions has not been determined. We used immunofluorescence and freeze fracture replica immunogold labelling (FRIL) of Cx36 to investigate the cellular and subcellular localization of Cx36 in the pineal gland of adult mouse and rat. Immunofluorescence labelling of Cx36 was visualized exclusively as puncta or short immunopositive strands that were distributed throughout the pineal, and which were absent in pineal sections from Cx36 null mice. By double immunofluorescence labelling, Cx36 was localized to tryptophan hydroxylase-positive and 5-hydroxytryptamine-positive pinealocyte cell bodies and their large initial processes, including at intersections of those processes and at sites displaying a confluence of processes. Labelling for the cell junction marker zonula occludens-1 (ZO-1) either overlapped or was closely associated with labelling for Cx36. Pinealocytes thus form Cx36-containing gap junctions that also incorporate the scaffolding protein ZO-1. FRIL revealed labelling of Cx36 at ultrastructurally defined gap junctions between pinealocytes, most of which was at gap junctions having reticular, ribbon or string configurations. The results suggest that the endocrine functions of pinealocytes and their secretion of melatonin is supported by their intercellular communication via Cx36-containing gap junctions, which may now be tested by the use of Cx36 null mice.
Collapse
Affiliation(s)
- S G Wang
- Department of Physiology, Faculty of Medicine, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB, R3E 0J9, Canada
| | - D D Tsao
- Department of Physiology, Faculty of Medicine, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB, R3E 0J9, Canada
| | - K G Vanderpool
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - T Yasumura
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - J E Rash
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - J I Nagy
- Department of Physiology, Faculty of Medicine, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB, R3E 0J9, Canada
| |
Collapse
|
50
|
Epifantseva I, Shaw RM. Intracellular trafficking pathways of Cx43 gap junction channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:40-47. [PMID: 28576298 DOI: 10.1016/j.bbamem.2017.05.018] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/19/2017] [Accepted: 05/25/2017] [Indexed: 12/11/2022]
Abstract
Gap Junction (GJ) channels, including the most common Connexin 43 (Cx43), have fundamental roles in excitable tissues by facilitating rapid transmission of action potentials between adjacent cells. For instance, synchronization during each heartbeat is regulated by these ion channels at the cardiomyocyte cell-cell border. Cx43 protein has a short half-life, and rapid synthesis and timely delivery of those proteins to particular subdomains are crucial for the cellular organization of gap junctions and maintenance of intracellular coupling. Impairment in gap junction trafficking contributes to dangerous complications in diseased hearts such as the arrhythmias of sudden cardiac death. Of recent interest are the protein-protein interactions with the Cx43 carboxy-terminus. These interactions have significant impact on the full length Cx43 lifecycle and also contribute to trafficking of Cx43 as well as possibly other functions. We are learning that many of the known non-canonical roles of Cx43 can be attributed to the recently identified six endogenous Cx43 truncated isoforms which are produced by internal translation. In general, alternative translation is a new leading edge for proteome expansion and therapeutic drug development. This review highlights recent mechanisms identified in the trafficking of gap junction channels, involvement of other proteins contributing to the delivery of channels to the cell-cell border, and understanding of possible roles of the newly discovered alternatively translated isoforms in Cx43 biology. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Irina Epifantseva
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Robin M Shaw
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.; Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA..
| |
Collapse
|