1
|
Kraujaliene L, Kraujalis T, Snipas M, Verselis VK. An Ala/Glu difference in E1 of Cx26 and Cx30 contributes to their differential anionic permeabilities. J Gen Physiol 2024; 156:e202413600. [PMID: 39302317 PMCID: PMC11415307 DOI: 10.1085/jgp.202413600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
Two closely related connexins, Cx26 and Cx30, share widespread expression in the cochlear cellular networks. Gap junction channels formed by these connexins have been shown to have different permeability profiles, with Cx30 showing a strongly reduced preference for anionic tracers. The pore-forming segment of the first extracellular loop, E1, identified by computational studies of the Cx26 crystal structure to form a parahelix and a narrowed region of the pore, differs at a single residue at position 49. Cx26 contains an Ala and Cx30, a charged Glu at this position, and cysteine scanning in hemichannels identified this position to be pore-lining. To assess whether the Ala/Glu difference affects permeability, we modeled and quantified Lucifer Yellow transfer between HeLa cell pairs expressing WT Cx26 and Cx30 and variants that reciprocally substituted Glu and Ala at position 49. Cx26(A49E) and Cx30(E49A) substitutions essentially reversed the Lucifer Yellow permeability profile when accounting for junctional conductance. Moreover, by using a calcein efflux assay in single cells, we observed a similar reduced anionic preference in undocked Cx30 hemichannels and a reversal with reciprocal Ala/Glu substitutions. Thus, our data indicate that Cx26 and Cx30 gap junction channels and undocked hemichannels retain similar permeability characteristics and that a single residue difference in their E1 domains can largely account for their differential permeabilities to anionic tracers. The higher anionic permeability of Cx26 compared with Cx30 suggests that these connexins may serve distinct signaling functions in the cochlea, perhaps reflected in the vastly higher prevalence of Cx26 mutations in human deafness.
Collapse
Affiliation(s)
- Lina Kraujaliene
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Tadas Kraujalis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Applied Informatics, Kaunas University of Technology, Kaunas, Lithuania
| | - Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Mathematical Modelling, Kaunas University of Technology, Kaunas, Lithuania
| | - Vytas K. Verselis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Kumar D, Harris AL, Luo YL. Molecular permeation through large pore channels: computational approaches and insights. J Physiol 2024:10.1113/JP285198. [PMID: 39373834 PMCID: PMC11973239 DOI: 10.1113/jp285198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Computational methods such as molecular dynamics (MD) have illuminated how single-atom ions permeate membrane channels and how selectivity among them is achieved. Much less is understood about molecular permeation through eukaryotic channels that mediate the flux of small molecules (e.g. connexins, pannexins, LRRC8s, CALHMs). Here we describe computational methods that have been profitably employed to explore the movements of molecules through wide pores, revealing mechanistic insights, guiding experiments, and suggesting testable hypotheses. This review illustrates MD techniques such as voltage-driven flux, potential of mean force, and mean first-passage-time calculations, as applied to molecular permeation through wide pores. These techniques have enabled detailed and quantitative modeling of molecular interactions and movement of permeants at the atomic level. We highlight novel contributors to the transit of molecules through these wide pathways. In particular, the flexibility and anisotropic nature of permeant molecules, coupled with the dynamics of pore-lining residues, lead to bespoke permeation dynamics. As more eukaryotic large-pore channel structures and functional data become available, these insights and approaches will be important for understanding the physical principles underlying molecular permeation and as guides for experimental design.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Biotechnology and Pharmaceutical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Andrew L. Harris
- Department of Pharmacology, Physiology, and Neuroscience. New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Yun Lyna Luo
- Department of Biotechnology and Pharmaceutical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
3
|
Gaete PS, Kumar D, Fernandez CI, Valdez Capuccino JM, Bhatt A, Jiang W, Lin YC, Liu Y, Harris AL, Luo YL, Contreras JE. Large-pore connexin hemichannels function like molecule transporters independent of ion conduction. Proc Natl Acad Sci U S A 2024; 121:e2403903121. [PMID: 39116127 PMCID: PMC11331127 DOI: 10.1073/pnas.2403903121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024] Open
Abstract
Connexin hemichannels were identified as the first members of the eukaryotic large-pore channel family that mediate permeation of both atomic ions and small molecules between the intracellular and extracellular environments. The conventional view is that their pore is a large passive conduit through which both ions and molecules diffuse in a similar manner. In stark contrast to this notion, we demonstrate that the permeation of ions and of molecules in connexin hemichannels can be uncoupled and differentially regulated. We find that human connexin mutations that produce pathologies and were previously thought to be loss-of-function mutations due to the lack of ionic currents are still capable of mediating the passive transport of molecules with kinetics close to those of wild-type channels. This molecular transport displays saturability in the micromolar range, selectivity, and competitive inhibition, properties that are tuned by specific interactions between the permeating molecules and the N-terminal domain that lies within the pore-a general feature of large-pore channels. We propose that connexin hemichannels and, likely, other large-pore channels, are hybrid channel/transporter-like proteins that might switch between these two modes to promote selective ion conduction or autocrine/paracrine molecular signaling in health and disease processes.
Collapse
Affiliation(s)
- Pablo S. Gaete
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA95616
| | - Deepak Kumar
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA91766
| | - Cynthia I. Fernandez
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA95616
| | - Juan M. Valdez Capuccino
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ07103
| | - Aashish Bhatt
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA91766
| | - Wenjuan Jiang
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA91766
| | - Yi-Chun Lin
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA91766
| | - Yu Liu
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ07103
| | - Andrew L. Harris
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ07103
| | - Yun L. Luo
- Department of Biotechnology and Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA91766
| | - Jorge E. Contreras
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA95616
| |
Collapse
|
4
|
Memo C, Parisse P, Amoriello R, Pachetti M, Palandri A, Casalis L, Ballerini C, Ballerini L. Extracellular vesicles released by LPS-stimulated spinal organotypic slices spread neuroinflammation into naïve slices through connexin43 hemichannel opening and astrocyte aberrant calcium dynamics. Front Cell Neurosci 2024; 18:1433309. [PMID: 39049826 PMCID: PMC11266295 DOI: 10.3389/fncel.2024.1433309] [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: 05/15/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Neuroinflammation is a hallmark of multiple neurodegenerative diseases, shared by all pathological processes which primarily impact on neurons, including Central Nervous System (CNS) injuries. In reactive CNS, activated glia releases extracellular vesicles (EVs), nanosized membranous particles known to play a key role in intercellular communication. EVs mediate neuroinflammatory responses and might exacerbate tissue deterioration, ultimately influencing neurodegenerative disease progression. Methods We treated spinal cord organotypic slices with LPS, a ligand extensively used to induce sEVs release, to mimic mild inflammatory conditions. We combine atomic force microscopy (AFM), nanoparticle tracking (NTA) and western blot (WB) analysis to validate the isolation and characterisation of sEVs. We further use immunofluorescence and confocal microscopy with live calcium imaging by GCaMP6f reporter to compare glial reactivity to treatments with sEVs when isolated from resting and LPS treated organ slices. Results In our study, we focus on CNS released small EVs (sEVs) and their impact on the biology of inflammatory environment. We address sEVs local signalling within the CNS tissue, in particular their involvement in inflammation spreading mechanism(s). sEVs are harvested from mouse organotypic spinal cord cultures, an in vitro model which features 3D complexity and retains spinal cord resident cells. By confocal microscopy and live calcium imaging we monitor glial responses in naïve spinal slices when exposed to sEVs isolated from resting and LPS treated organ slices. Discussion We show that sEVs, only when released during LPS neuroinflammation, recruit naïve astrocytes in the neuroinflammation cycle and we propose that such recruitment be mediated by EVs hemichannel (HC) permeability.
Collapse
Affiliation(s)
- Christian Memo
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | - Pietro Parisse
- Nanoinnovation Lab, ELETTRA Synchrotron Light Source, Basovizza, Italy
- CNR-IOM, Basovizza, Italy
| | - Roberta Amoriello
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Firenze, Italy
| | - Maria Pachetti
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | - Anabela Palandri
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| | - Loredana Casalis
- Nanoinnovation Lab, ELETTRA Synchrotron Light Source, Basovizza, Italy
| | - Clara Ballerini
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Firenze, Italy
| | - Laura Ballerini
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| |
Collapse
|
5
|
Yuan C, Gerhards L, Solov'yov IA, Dedek K. Biotin-cGMP and -cAMP are able to permeate through the gap junctions of some amacrine cells in the mouse retina despite their large size. FRONTIERS IN OPHTHALMOLOGY 2024; 3:1334602. [PMID: 38983094 PMCID: PMC11182161 DOI: 10.3389/fopht.2023.1334602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/22/2023] [Indexed: 07/11/2024]
Abstract
Gap junctions transmit electrical signals in neurons and serve metabolic coupling and chemical communication. Gap junctions are made of intercellular channels with large pores, allowing ions and small molecules to permeate. In the mammalian retina, intercellular coupling fulfills many vital functions in visual signal processing but is also implicated in promoting cell death after insults, such as excitotoxicity or hypoxia. Conversely, some studies also suggested a role for retinal gap junctions in neuroprotection. Recently, gap junctions were also advocated as conduits for therapeutic drug delivery in neurodegenerative disorders. This requires the permeation of rather large molecules through retinal gap junctions. However, the permeability of retinal networks for molecules >0.6 kDa has not been tested systematically. Here, we used the cut-loading method and probed gap junctional networks in the mouse retina for their permeability to cGMP and cAMP coupled to Biotin, using the well-characterized tracer Neurobiotin as control. Biotin-cGMP and -cAMP have a molecular weight of >0.8 kDa. We show that they cannot pass the gap junctions of horizontal cells but can permeate through the gap junctions of specific amacrine cells in the inner retina. These amacrine cells do not comprise AII amacrine cells and nitric oxide-releasing amacrine cells but some unknown type. In summary, we show that some retinal gap junctions are large enough to let molecules >0.8 kDa pass, making the intercellular delivery of therapeutic agents - already successfully exploited, for example, in cancer - also feasible in neurodegenerative diseases.
Collapse
Affiliation(s)
- Chunxu Yuan
- Animal Navigation, Institute for Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Luca Gerhards
- Institute of Physics, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Ilia A Solov'yov
- Institute of Physics, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
- CeNaD - Center for Nanoscale Dynamics, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Animal Navigation, Institute for Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
6
|
León-Fuentes IM, Salgado-Gil MG, Novoa MS, Retamal MA. Connexins in Cancer, the Possible Role of Connexin46 as a Cancer Stem Cell-Determining Protein. Biomolecules 2023; 13:1460. [PMID: 37892142 PMCID: PMC10604234 DOI: 10.3390/biom13101460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/15/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is a widespread and incurable disease caused by genetic mutations, leading to uncontrolled cell proliferation and metastasis. Connexins (Cx) are transmembrane proteins that facilitate intercellular communication via hemichannels and gap junction channels. Among them, Cx46 is found mostly in the eye lens. However, in pathological conditions, Cx46 has been observed in various types of cancers, such as glioblastoma, melanoma, and breast cancer. It has been demonstrated that elevated Cx46 levels in breast cancer contribute to cellular resistance to hypoxia, and it is an enhancer of cancer aggressiveness supporting a pro-tumoral role. Accordingly, Cx46 is associated with an increase in cancer stem cell phenotype. These cells display radio- and chemoresistance, high proliferative abilities, self-renewal, and differentiation capacities. This review aims to consolidate the knowledge of the relationship between Cx46, its role in forming hemichannels and gap junctions, and its connection with cancer and cancer stem cells.
Collapse
Affiliation(s)
| | | | | | - Mauricio A. Retamal
- Programa de Comunicación Celular en Cáncer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, República de Honduras 12740, Las Condes, Santiago 7610496, Chile; (I.M.L.-F.); (M.G.S.-G.); (M.S.N.)
| |
Collapse
|
7
|
Qi C, Lavriha P, Bayraktar E, Vaithia A, Schuster D, Pannella M, Sala V, Picotti P, Bortolozzi M, Korkhov VM. Structures of wild-type and selected CMT1X mutant connexin 32 gap junction channels and hemichannels. SCIENCE ADVANCES 2023; 9:eadh4890. [PMID: 37647412 PMCID: PMC10468125 DOI: 10.1126/sciadv.adh4890] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/27/2023] [Indexed: 09/01/2023]
Abstract
In myelinating Schwann cells, connection between myelin layers is mediated by gap junction channels (GJCs) formed by docked connexin 32 (Cx32) hemichannels (HCs). Mutations in Cx32 cause the X-linked Charcot-Marie-Tooth disease (CMT1X), a degenerative neuropathy without a cure. A molecular link between Cx32 dysfunction and CMT1X pathogenesis is still missing. Here, we describe the high-resolution cryo-electron cryo-myography (cryo-EM) structures of the Cx32 GJC and HC, along with two CMT1X-linked mutants, W3S and R22G. While the structures of wild-type and mutant GJCs are virtually identical, the HCs show a major difference: In the W3S and R22G mutant HCs, the amino-terminal gating helix partially occludes the pore, consistent with a diminished HC activity. Our results suggest that HC dysfunction may be involved in the pathogenesis of CMT1X.
Collapse
Affiliation(s)
- Chao Qi
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Pia Lavriha
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Erva Bayraktar
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Anand Vaithia
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Dina Schuster
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Micaela Pannella
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Valentina Sala
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Paola Picotti
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Mario Bortolozzi
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Volodymyr M. Korkhov
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| |
Collapse
|
8
|
Zappalà A, Romano IR, D’Angeli F, Musumeci G, Lo Furno D, Giuffrida R, Mannino G. Functional Roles of Connexins and Gap Junctions in Osteo-Chondral Cellular Components. Int J Mol Sci 2023; 24:4156. [PMID: 36835567 PMCID: PMC9967557 DOI: 10.3390/ijms24044156] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Gap junctions (GJs) formed by connexins (Cxs) play an important role in the intercellular communication within most body tissues. In this paper, we focus on GJs and Cxs present in skeletal tissues. Cx43 is the most expressed connexin, participating in the formation of both GJs for intercellular communication and hemichannels (HCs) for communication with the external environment. Through GJs in long dendritic-like cytoplasmic processes, osteocytes embedded in deep lacunae are able to form a functional syncytium not only with neighboring osteocytes but also with bone cells located at the bone surface, despite the surrounding mineralized matrix. The functional syncytium allows a coordinated cell activity through the wide propagation of calcium waves, nutrients and anabolic and/or catabolic factors. Acting as mechanosensors, osteocytes are able to transduce mechanical stimuli into biological signals that spread through the syncytium to orchestrate bone remodeling. The fundamental role of Cxs and GJs is confirmed by a plethora of investigations that have highlighted how up- and downregulation of Cxs and GJs critically influence skeletal development and cartilage functions. A better knowledge of GJ and Cx mechanisms in physiological and pathological conditions might help in developing therapeutic approaches aimed at the treatment of human skeletal system disorders.
Collapse
Affiliation(s)
- Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Ivana Roberta Romano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Floriana D’Angeli
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giuliana Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
| |
Collapse
|
9
|
Ortiz J, Bobkov YV, DeBiasse MB, Mitchell DG, Edgar A, Martindale MQ, Moss AG, Babonis LS, Ryan JF. Independent Innexin Radiation Shaped Signaling in Ctenophores. Mol Biol Evol 2023; 40:7026321. [PMID: 36740225 PMCID: PMC9949713 DOI: 10.1093/molbev/msad025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/30/2022] [Accepted: 01/25/2023] [Indexed: 02/07/2023] Open
Abstract
Innexins facilitate cell-cell communication by forming gap junctions or nonjunctional hemichannels, which play important roles in metabolic, chemical, ionic, and electrical coupling. The lack of knowledge regarding the evolution and role of these channels in ctenophores (comb jellies), the likely sister group to the rest of animals, represents a substantial gap in our understanding of the evolution of intercellular communication in animals. Here, we identify and phylogenetically characterize the complete set of innexins of four ctenophores: Mnemiopsis leidyi, Hormiphora californensis, Pleurobrachia bachei, and Beroe ovata. Our phylogenetic analyses suggest that ctenophore innexins diversified independently from those of other animals and were established early in the emergence of ctenophores. We identified a four-innexin genomic cluster, which was present in the last common ancestor of these four species and has been largely maintained in these lineages. Evidence from correlated spatial and temporal gene expression of the M. leidyi innexin cluster suggests that this cluster has been maintained due to constraints related to gene regulation. We describe the basic electrophysiological properties of putative ctenophore hemichannels from muscle cells using intracellular recording techniques, showing substantial overlap with the properties of bilaterian innexin channels. Together, our results suggest that the last common ancestor of animals had gap junctional channels also capable of forming functional innexin hemichannels, and that innexin genes have independently evolved in major lineages throughout Metazoa.
Collapse
Affiliation(s)
| | | | - Melissa B DeBiasse
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL, USA,School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Dorothy G Mitchell
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL, USA,Department of Biology, University of Florida, Gainesville, FL, USA
| | - Allison Edgar
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL, USA
| | - Mark Q Martindale
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL, USA,Department of Biology, University of Florida, Gainesville, FL, USA
| | - Anthony G Moss
- Biological Sciences Department, Auburn University, Auburn, AL, USA
| | - Leslie S Babonis
- Whitney Laboratory for Marine Bioscience, University of Florida, St Augustine, FL, USA,Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
10
|
Wei H, Moffett JR, Amanat M, Fatemi A, Tsukamoto T, Namboodiri AM, Slusher BS. The pathogenesis of, and pharmacological treatment for, Canavan disease. Drug Discov Today 2022; 27:2467-2483. [PMID: 35636725 PMCID: PMC11806932 DOI: 10.1016/j.drudis.2022.05.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/05/2022] [Accepted: 05/24/2022] [Indexed: 12/12/2022]
Abstract
Canavan disease (CD) is an inherited leukodystrophy resulting from mutations in the gene encoding aspartoacylase (ASPA). ASPA is highly expressed in oligodendrocytes and catalyzes the cleavage of N-acetylaspartate (NAA) to produce aspartate and acetate. In this review, we examine the pathologies and clinical presentation in CD, the metabolism and transportation of NAA in the brain, and the hypothetical mechanisms whereby ASPA deficiency results in dysmyelination and a failure of normal brain development. We also discuss therapeutic options that could be used for the treatment of CD.
Collapse
Affiliation(s)
- Huijun Wei
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA
| | - John R Moffett
- Department of Anatomy, Physiology and Genetics and Neuroscience Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA.
| | - Man Amanat
- Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Ali Fatemi
- Department of Neurology, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Behavioral Science, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Aryan M Namboodiri
- Department of Anatomy, Physiology and Genetics and Neuroscience Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Medicine, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
11
|
Endogenous pannexin1 channels form functional intercellular cell-cell channels with characteristic voltage-dependent properties. Proc Natl Acad Sci U S A 2022; 119:e2202104119. [PMID: 35486697 PMCID: PMC9171361 DOI: 10.1073/pnas.2202104119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pannexin1 is a glycoprotein that has been shown to form functional plasma membrane channels and mediate many cellular signaling pathways. However, the formation and function of pannexin1-based intercellular cell–cell channels in mammalian cells and vertebrate tissue is a question of substantial debate. This work provides robust electrophysiological evidence to demonstrate that endogenously expressed human pannexin1 forms cell–cell channels and lays the groundwork for studying a potential new type of electrical synapses between many mammalian cell types that endogenously express pannexin1. The occurrence of intercellular channels formed by pannexin1 has been challenged for more than a decade. Here, we provide an electrophysiological characterization of exogenous human pannexin1 (hPanx1) cell–cell channels expressed in HeLa cells knocked out for connexin45. The observed hPanx1 cell–cell channels show two phenotypes: O-state and S-state. The former displayed low transjunctional voltage (Vj) sensitivity and single-channel conductance of ∼175 pS, with a substate of ∼35 pS; the latter showed a peculiar dynamic asymmetry in Vj dependence and single-channel conductance identical to the substate conductance of the O-state. S-state hPanx1 cell–cell channels were also identified between TC620 cells, a human oligodendroglioma cell line that endogenously expresses hPanx1. In these cells, dye and electrical coupling increased with temperature and were strongly reduced after hPanx1 expression was knocked down by small interfering RNA or inhibited with Panx1 mimetic inhibitory peptide. Moreover, cell–cell coupling was augmented when hPanx1 levels were increased with a doxycycline-inducible expression system. Application of octanol, a connexin gap junction (GJ) channel inhibitor, was not sufficient to block electrical coupling between HeLa KO Cx45-hPanx1 or TC620 cell pairs. In silico studies suggest that several arginine residues inside the channel pore may be neutralized by hydrophobic interactions, allowing the passage of DAPI, consistent with dye coupling observed between TC620 cells. These findings demonstrate that endogenously expressed hPanx1 forms intercellular cell–cell channels and their unique properties resemble those described in innexin-based GJ channels. Since Panx1 is ubiquitously expressed, finding conditions to recognize Panx1 cell–cell channels in different cell types might require special attention.
Collapse
|
12
|
King DR, Sedovy MW, Leng X, Xue J, Lamouille S, Koval M, Isakson BE, Johnstone SR. Mechanisms of Connexin Regulating Peptides. Int J Mol Sci 2021; 22:ijms221910186. [PMID: 34638526 PMCID: PMC8507914 DOI: 10.3390/ijms221910186] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/22/2022] Open
Abstract
Gap junctions (GJ) and connexins play integral roles in cellular physiology and have been found to be involved in multiple pathophysiological states from cancer to cardiovascular disease. Studies over the last 60 years have demonstrated the utility of altering GJ signaling pathways in experimental models, which has led to them being attractive targets for therapeutic intervention. A number of different mechanisms have been proposed to regulate GJ signaling, including channel blocking, enhancing channel open state, and disrupting protein-protein interactions. The primary mechanism for this has been through the design of numerous peptides as therapeutics, that are either currently in early development or are in various stages of clinical trials. Despite over 25 years of research into connexin targeting peptides, the overall mechanisms of action are still poorly understood. In this overview, we discuss published connexin targeting peptides, their reported mechanisms of action, and the potential for these molecules in the treatment of disease.
Collapse
Affiliation(s)
- D. Ryan King
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
| | - Meghan W. Sedovy
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, VA 24061, USA
| | - Xinyan Leng
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
| | - Jianxiang Xue
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (J.X.); (B.E.I.)
| | - Samy Lamouille
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA 24016, USA
| | - Michael Koval
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (J.X.); (B.E.I.)
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Scott R. Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA 24016, USA; (D.R.K.); (M.W.S.); (X.L.); (S.L.)
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060, USA
- Correspondence:
| |
Collapse
|
13
|
Gaete PS, Lillo MA, López W, Liu Y, Jiang W, Luo Y, Harris AL, Contreras JE. A novel voltage-clamp/dye uptake assay reveals saturable transport of molecules through CALHM1 and connexin channels. J Gen Physiol 2021; 152:211474. [PMID: 33074302 PMCID: PMC7579738 DOI: 10.1085/jgp.202012607] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Large-pore channels permeable to small molecules such as ATP, in addition to atomic ions, are emerging as important regulators in health and disease. Nonetheless, their mechanisms of molecular permeation and selectivity remain mostly unexplored. Combining fluorescence microscopy and electrophysiology, we developed a novel technique that allows kinetic analysis of molecular permeation through connexin and CALHM1 channels in Xenopus oocytes rendered translucent. Using this methodology, we found that (1) molecular flux through these channels saturates at low micromolar concentrations, (2) kinetic parameters of molecular transport are sensitive to modulators of channel gating, (3) molecular transport and ionic currents can be differentially affected by mutation and gating, and (4) N-terminal regions of these channels control transport kinetics and permselectivity. Our methodology allows analysis of how human disease-causing mutations affect kinetic properties and permselectivity of molecular signaling and enables the study of molecular mechanisms, including selectivity and saturability, of molecular transport in other large-pore channels.
Collapse
Affiliation(s)
- Pablo S Gaete
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Mauricio A Lillo
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - William López
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Yu Liu
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Wenjuan Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA
| | - Yun Luo
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA
| | - Andrew L Harris
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| | - Jorge E Contreras
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ
| |
Collapse
|
14
|
Rode K, Langeheine M, Seeger B, Brehm R. Connexin43 in Germ Cells Seems to Be Dispensable for Murine Spermatogenesis. Int J Mol Sci 2021; 22:ijms22157924. [PMID: 34360693 PMCID: PMC8348783 DOI: 10.3390/ijms22157924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/13/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023] Open
Abstract
Testicular Connexin43 (Cx43) connects adjacent Sertoli cells (SC) and SC to germ cells (GC) in the seminiferous epithelium and plays a crucial role in spermatogenesis. However, the distinction whether this results from impaired inter-SC communication or between GC and SC is not possible, so far. Thus, the question arises, whether a GC-specific Cx43 KO has similar effects on spermatogenesis as it is general or SC-specific KO. Using the Cre/loxP recombinase system, two conditional KO mouse lines lacking Cx43 in premeiotic (pGCCx43KO) or meiotic GC (mGCCx43KO) were generated. It was demonstrated by qRT-PCR that Cx43 mRNA was significantly decreased in adult pGCCx43KO mice, while it was also reduced in mGCCx43KO mice, yet not statistically significant. Body and testis weights, testicular histology, tubular diameter, numbers of intratubular cells and Cx43 protein synthesis and localization did not show any significant differences in semi-quantitative Western blot analysis and immunohistochemistry comparing adult male KO and WT mice of both mouse lines. Male KO mice were fertile. These results indicate that Cx43 in spermatogonia/spermatids does not seem to be essential for successful termination of spermatogenesis and fertility as it is known for Cx43 in somatic SC, but SC-GC communication might rather occur via heterotypic GJ channels.
Collapse
Affiliation(s)
- Kristina Rode
- Institute of Anatomy, University of Veterinary Medicine Foundation, 30173 Hanover, Germany; (K.R.); (M.L.)
| | - Marion Langeheine
- Institute of Anatomy, University of Veterinary Medicine Foundation, 30173 Hanover, Germany; (K.R.); (M.L.)
| | - Bettina Seeger
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Foundation, 30173 Hanover, Germany;
| | - Ralph Brehm
- Institute of Anatomy, University of Veterinary Medicine Foundation, 30173 Hanover, Germany; (K.R.); (M.L.)
- Correspondence: ; Tel.: +49-511-8457215
| |
Collapse
|
15
|
GJA1 rs2071165 A > G Variant Increased Gastric Cancer Risk in Females of Northwest China: A Case-Control Study. JOURNAL OF ONCOLOGY 2021; 2021:5556303. [PMID: 34221012 PMCID: PMC8225425 DOI: 10.1155/2021/5556303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/28/2021] [Indexed: 11/18/2022]
Abstract
Gastric cancer (GC) is one of the most common malignancies, and its incidence rates vary widely between men and women. Previous studies have suggested that connexin 43 (Cx43, encoded by gap junction protein alpha 1 (GJA1)) and secretory carrier membrane protein 1 (SCAMP1) are key functional proteins in tumors. Herein, the association between GJA1 and SCAMP1 polymorphisms and GC susceptibility and prognosis was evaluated. A total of three single-nucleotide polymorphisms among 681GC patients and 756 controls were tested using the Agena MassARRAY RS1000 system, including GJA1 rs2071165, SCAMP1 rs4530741, and SCAMP1 rs6874309. The strength of the association with GC risk was assessed by the odds ratios (ORs) and 95% confidence intervals (CIs) generated from the logistic regression model. Kaplan–Meier curve, long-rank tests, and a multivariate Cox proportional hazard model were used for prognosis analysis. The expression of GJA1 was assessed by immunohistochemistry. The GJA1 rs2071165 AA/AG genotype significantly increased the risk of GC in the female Chinese population (OR = 1.55, 95% CI = 1.03–2.32, p=0.034). Furthermore, the risk effect of GJA1 rs2071165 was more evident in the subgroups of female patients with GC, stratified by age, clinical stage, tumor size, and recurrence/metastasis. However, no obvious differences in Cx43 expression in GC tissues were observed between males and females. Furthermore, no significant association between SCAMP1 rs4530741 and rs6874309 polymorphisms and GC risk or prognosis was observed. In conclusion, this study suggests for the first time that the GJA1 rs2071165 polymorphism is associated with increased GC risk in females, revealing a potential new clinical marker for assessing GC risk in females.
Collapse
|
16
|
Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int J Mol Sci 2021; 22:ijms22094413. [PMID: 33922534 PMCID: PMC8122935 DOI: 10.3390/ijms22094413] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Collapse
|
17
|
Natha CM, Vemulapalli V, Fiori MC, Chang CWT, Altenberg GA. Connexin hemichannel inhibitors with a focus on aminoglycosides. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166115. [PMID: 33711451 DOI: 10.1016/j.bbadis.2021.166115] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/06/2021] [Accepted: 02/22/2021] [Indexed: 12/31/2022]
Abstract
Connexins are membrane proteins involved directly in cell-to-cell communication through the formation of gap-junctional channels. These channels result from the head-to-head docking of two hemichannels, one from each of two adjacent cells. Undocked hemichannels are also present at the plasma membrane where they mediate the efflux of molecules that participate in autocrine and paracrine signaling, but abnormal increase in hemichannel activity can lead to cell damage in disorders such as cardiac infarct, stroke, deafness, cataracts, and skin diseases. For this reason, connexin hemichannels have emerged as a valid therapeutic target. Know small molecule hemichannel inhibitors are not ideal leads for the development of better drugs for clinical use because they are not specific and/or have toxic effects. Newer inhibitors are more selective and include connexin mimetic peptides, anti-connexin antibodies and drugs that reduce connexin expression such as antisense oligonucleotides. Re-purposed drugs and their derivatives are also promising because of the significant experience with their clinical use. Among these, aminoglycoside antibiotics have been identified as inhibitors of connexin hemichannels that do not inhibit gap-junctional channels. In this review, we discuss connexin hemichannels and their inhibitors, with a focus on aminoglycoside antibiotics and derivatives of kanamycin A that inhibit connexin hemichannels, but do not have antibiotic effect.
Collapse
Affiliation(s)
- Cristina M Natha
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Varun Vemulapalli
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Mariana C Fiori
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Cheng-Wei T Chang
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, USA
| | - Guillermo A Altenberg
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
18
|
Li C, Shi L, Peng C, Yu G, Zhang Y, Du Z. Lead-induced cardiomyocytes apoptosis by inhibiting gap junction intercellular communication via autophagy activation. Chem Biol Interact 2020; 337:109331. [PMID: 33242459 DOI: 10.1016/j.cbi.2020.109331] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/01/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022]
Abstract
Lead (Pb) is one of the most common heavy metal contaminants in the environment. Pb can cause pathophysiological changes in several organ systems, including the cardiovascular system, but the molecular mechanism remains elusive. The study aimed to study the effects of Pb on Gap junction intercellular communication (GJIC) and its role in Pb-induced apoptosis. The present study aims to determine whether Pb-induced autophagy promotes apoptosis of rat cardiac myocytes (H9c2 cells) by downregulating GJIC using CCK-8 Kit, scrape loading/dye transfer assay, Annexin V/PI assays, Western blot analysis and double-immunofluorescence experiments. The results showed that Pb elicited cytotoxicity in a time- and concentration-dependent manner and led to increased apoptosis in a concentration-dependent manner in H9c2 cells. Pb also reduced GJIC in H9c2 cells in a concentration-dependent manner through the downregulation of connexin (Cx) 43. Inhibition of gap junctions by gap junction blocker carbenoxolone disodium (CBX) resulted in increased apoptosis. Furthermore, Pb increased autophagy in a concentration-dependent manner in H9c2 cells, decreasing the distribution of Cx43 on the cell membrane, and targeted Cx43 to autophagosome via light chain 3 (LC3). However, autophagy inhibitor 3-Methyladenine (3-MA) can slow down the downregulation of Cx43 induced by Pb in H9c2 cells. In conclusion, our results provide evidence that Pb-decreased GJIC promotes apoptosis in cardiomyocytes. This is probably because of the fact that Pb-induced autophagy exacerbates GJIC inhibition and downregulation of Cx43.
Collapse
Affiliation(s)
- Chao Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China
| | - Liang Shi
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China
| | - Cheng Peng
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China; Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, 4108, Queensland, Australia
| | - Gongchang Yu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China; Laboratory Animal Center, North China University of Science and Technology, Tangshan, 063210, Hebei, China.
| | - Zhongjun Du
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China.
| |
Collapse
|
19
|
Koç Ş, Baysal SS. Practical Method for Salt Intake Follow-Up in Hypertensive Patients. Metab Syndr Relat Disord 2020; 18:353-361. [PMID: 32580624 DOI: 10.1089/met.2020.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Obese and hypertensive (HT) patients should restrict salt intake. In excessive salt intake, ouabain-like compounds inhibit Na/K-ATPase (Na+ pump), which increases intracellular Na+ and Ca2+. Ca2+ has a vasotonic effect on arteries and an inotropic effect on the heart and may cause cortical opacities in the lens. To our knowledge, there is still no practical method for salt intake follow-up. This study tested whether salt intake follow-up can be performed with the help of opacity tracking. Methods: In total, 400 HT patients (age 30-69 years) with cortical lens opacities were included in the study. Changes in opacities based on biomicroscopic examination at baseline and after 3 months were recorded digitally with the help of imaging software. Salt intake at 1 and 3 months was evaluated with a 24-hr urine Na assay. Changes in opacities were compared among group 1 (∼50% salt reduction), group 2 (∼10% salt reduction), and group 3 (∼15% salt increase). Results: Age and changes in small opacity diameter (SOD) and large opacity diameter (LOD) were the most important determinants of the 50% salt reduction in the third month. For changes in LOD, the sensitivity was 88.5% [confidence interval (95% CI) 85.2-91.7] and specificity was 95.5% (95% CI 93.1-98.7) for predicting a 50% salt restriction during the 3-month period. For SOD, the values were 85% (95% CI 82.5-87) and 95% (95% CI 92.3-97.5), respectively. Conclusions: Opacity changes are a practical method for predicting a 50% reduction in salt intake over a 3-month period in 30- to 59-year-old HT patients.
Collapse
Affiliation(s)
- Şahbender Koç
- Department of Cardiology, University of Health Sciences. Keçiören Education and Training Hospital, Ankara, Turkey
| | - Sadettin Selçuk Baysal
- Department of Cardiology, University of Health Sciences, Şanlıurfa Mehmet Akif İnan Education and Training Hospital, Urfa, Turkey
| |
Collapse
|
20
|
Brink PR, Valiunas V, White TW. Lens Connexin Channels Show Differential Permeability to Signaling Molecules. Int J Mol Sci 2020; 21:ijms21186943. [PMID: 32971763 PMCID: PMC7555617 DOI: 10.3390/ijms21186943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/12/2022] Open
Abstract
Gap junction channels mediate the direct intercellular passage of small ions as well as larger solutes such as second messengers. A family of proteins called connexins make up the subunits of gap junction channels in chordate animals. Each individual connexin forms channels that exhibit distinct permeability to molecules that influence cellular signaling, such as calcium ions, cyclic nucleotides, or inositol phosphates. In this review, we examine the permeability of connexin channels containing Cx43, Cx46, and Cx50 to signaling molecules and attempt to relate the observed differences in permeability to possible in vivo consequences that were revealed by studies of transgenic animals where these connexin genes have been manipulated. Taken together, these data suggest that differences in the permeability of individual connexin channels to larger solutes like 3',5'-cyclic adenosine monophosphate (cAMP) and inositol 1,4,5-trisphosphate (IP3) could play a role in regulating epithelial cell division, differentiation, and homeostasis in organs like the ocular lens.
Collapse
|
21
|
Valiunas V, White TW. Connexin43 and connexin50 channels exhibit different permeability to the second messenger inositol triphosphate. Sci Rep 2020; 10:8744. [PMID: 32457413 PMCID: PMC7251084 DOI: 10.1038/s41598-020-65761-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/07/2020] [Indexed: 11/12/2022] Open
Abstract
Gap junction channels made of different connexins have distinct permeability to second messengers, which could affect many cell processes, including lens epithelial cell division. Here, we have compared the permeability of IP3 and Ca2+ through channels made from two connexins, Cx43 and Cx50, that are highly expressed in vertebrate lens epithelial cells. Solute transfer was measured while simultaneously monitoring junctional conductance via dual whole-cell/perforated patch clamp. HeLa cells expressing Cx43 or Cx50 were loaded with Fluo-8, and IP3 or Ca2+ were delivered via patch pipette to one cell of a pair, or to a monolayer while fluorescence intensity changes were recorded. Cx43 channels were permeable to IP3 and Ca2+. Conversely, Cx50 channels were impermeable to IP3, while exhibiting high permeation of Ca2+. Reduced Cx50 permeability to IP3 could play a role in regulating cell division and homeostasis in the lens.
Collapse
Affiliation(s)
- Virginijus Valiunas
- The Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794, USA
| | - Thomas W White
- The Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794, USA.
| |
Collapse
|
22
|
Dominiak A, Chełstowska B, Olejarz W, Nowicka G. Communication in the Cancer Microenvironment as a Target for Therapeutic Interventions. Cancers (Basel) 2020; 12:E1232. [PMID: 32422889 PMCID: PMC7281160 DOI: 10.3390/cancers12051232] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment (TME) is a complex system composed of multiple cells, such as non-cancerous fibroblasts, adipocytes, immune and vascular cells, as well as signal molecules and mediators. Tumor cells recruit and reprogram other cells to produce factors that maintain tumor growth. Communication between cancerous and surrounding cells is a two-way process and engages a diverse range of mechanisms that, in consequence, can lead to rapid proliferation, metastasis, and drug resistance, or can serve as a tumors-suppressor, e.g., through tumor-immune cell interaction. Cross-talk within the cancer microenvironment can be direct by cell-to-cell contact via adhesion molecules, electrical coupling, and passage through gap junctions, or indirect through classical paracrine signaling by cytokines, growth factors, and extracellular vesicles. Therapeutic approaches for modulation of cell-cell communication may be a promising strategy to combat tumors. In particular, integrative approaches targeting tumor communication in combination with conventional chemotherapy seem reasonable. Currently, special attention is paid to suppressing the formation of open-ended channels as well as blocking exosome production or ablating their cargos. However, many aspects of cell-to-cell communication have yet to be clarified, and, in particular, more work is needed in regard to mechanisms of bidirectional signal transfer. Finally, it seems that some interactions in TEM can be not only cancer-specific, but also patient-specific, and their recognition would help to predict patient response to therapy.
Collapse
Affiliation(s)
- Agnieszka Dominiak
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.O.); (G.N.)
- Center for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Beata Chełstowska
- Department of Internal Medicine and Hematology, Laboratory of Hematology and Flow Cytometry, Military Institute of Medicine, 04-140 Warsaw, Poland;
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.O.); (G.N.)
- Center for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Grażyna Nowicka
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.O.); (G.N.)
- Center for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
23
|
Jourdeuil K, Taneyhill LA. The gap junction protein connexin 43 controls multiple aspects of cranial neural crest cell development. J Cell Sci 2020; 133:jcs235440. [PMID: 31964703 PMCID: PMC7044449 DOI: 10.1242/jcs.235440] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022] Open
Abstract
Gap junctions are intercellular channels between cells that facilitate cell-cell communication. Connexin 43 (Cx43; also known as GJA1), the predominant gap junction protein in vertebrates, is expressed in premigratory cranial neural crest cells and is maintained throughout the neural crest cell epithelial-to-mesenchymal transition (EMT), but its function in these cells is unknown. To this end, we used a combination of in vivo and ex vivo experiments to assess gap junction formation, and Cx43 function, in chick cranial neural crest cells. Our results demonstrate that gap junctions exist between premigratory and migratory cranial neural crest cells and depend on Cx43 for their function. In the embryo, Cx43 knockdown just prior to EMT delays the emergence of Cx43-depleted neural crest cells from the neural tube, but these cells eventually successfully emigrate and join the migratory stream. This delay can be rescued by introduction of full-length Cx43 into Cx43-depleted cells. Furthermore, Cx43 depletion reduces the size of the premigratory neural crest cell domain through an early effect on neural crest cell specification. Collectively, these data identify new roles for Cx43 in chick cranial neural crest cell development.
Collapse
Affiliation(s)
- Karyn Jourdeuil
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
24
|
Yang W, Lampe PD, Kensel-Hammes P, Hesson J, Ware CB, Crisa L, Cirulli V. Connexin 43 Functions as a Positive Regulator of Stem Cell Differentiation into Definitive Endoderm and Pancreatic Progenitors. iScience 2019; 19:450-460. [PMID: 31430690 PMCID: PMC6708988 DOI: 10.1016/j.isci.2019.07.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/04/2019] [Accepted: 07/18/2019] [Indexed: 01/05/2023] Open
Abstract
Efficient stem cell differentiation into pancreatic islet cells is of critical importance for the development of cell replacement therapies for diabetes. Here, we identify the expression pattern of connexin 43 (Cx43), a gap junction (GJ) channel protein, in human embryonic stem cell (hESC)-derived definitive endoderm (DE) and primitive gut tube cells, representing early lineages for posterior foregut (PF), pancreatic progenitors (PP), pancreatic endocrine progenitors (PE), and islet cells. As the function of GJ channels is dependent on their gating status, we tested the impact of supplementing hESC-derived PP cell cultures with AAP10, a peptide that promotes Cx43 GJ channel opening. We found that this treatment promotes the expression of DE markers FoxA2 and Sox17, leads to a more efficient derivation of DE, and improves the yield of PF, PP, and PE cells. These results demonstrate a functional involvement of GJ channels in the differentiation of embryonic stem cells into pancreatic cell lineages.
Collapse
Affiliation(s)
- Wendy Yang
- Department of Medicine, UW Diabetes Institute, University of Washington, 850 Republican Street, S475, Seattle, WA 98109, USA
| | - Paul D Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Patricia Kensel-Hammes
- Department of Medicine, UW Diabetes Institute, University of Washington, 850 Republican Street, S475, Seattle, WA 98109, USA
| | - Jennifer Hesson
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, S480, Seattle, WA 98109, USA
| | - Carol B Ware
- Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, S480, Seattle, WA 98109, USA
| | - Laura Crisa
- Department of Medicine, UW Diabetes Institute, University of Washington, 850 Republican Street, S475, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, S480, Seattle, WA 98109, USA; Department of Pharmacology, University of Washington, Seattle, WA, USA.
| | - Vincenzo Cirulli
- Department of Medicine, UW Diabetes Institute, University of Washington, 850 Republican Street, S475, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican Street, S480, Seattle, WA 98109, USA; Department of Pharmacology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
25
|
Traub RD, Whittington MA, Maier N, Schmitz D, Nagy JI. Could electrical coupling contribute to the formation of cell assemblies? Rev Neurosci 2019; 31:121-141. [DOI: 10.1515/revneuro-2019-0059] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/07/2019] [Indexed: 12/20/2022]
Abstract
Abstract
Cell assemblies and central pattern generators (CPGs) are related types of neuronal networks: both consist of interacting groups of neurons whose collective activities lead to defined functional outputs. In the case of a cell assembly, the functional output may be interpreted as a representation of something in the world, external or internal; for a CPG, the output ‘drives’ an observable (i.e. motor) behavior. Electrical coupling, via gap junctions, is critical for the development of CPGs, as well as for their actual operation in the adult animal. Electrical coupling is also known to be important in the development of hippocampal and neocortical principal cell networks. We here argue that electrical coupling – in addition to chemical synapses – may therefore contribute to the formation of at least some cell assemblies in adult animals.
Collapse
Affiliation(s)
- Roger D. Traub
- AI Foundations, IBM T.J. Watson Research Center , Yorktown Heights, NY 10598 , USA
| | | | - Nikolaus Maier
- Charité-Universitätsmedizin Berlin , Neuroscience Research Center , Charitéplatz 1 , D-10117 Berlin , Germany
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin , Neuroscience Research Center , Charitéplatz 1 , D-10117 Berlin , Germany
| | - James I. Nagy
- Department of Physiology and Pathophysiology , University of Manitoba , Winnipeg R3E OJ9, MB , Canada
| |
Collapse
|
26
|
Valiunas V, Brink PR, White TW. Lens Connexin Channels Have Differential Permeability to the Second Messenger cAMP. Invest Ophthalmol Vis Sci 2019; 60:3821-3829. [PMID: 31529078 PMCID: PMC6750889 DOI: 10.1167/iovs.19-27302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/13/2019] [Indexed: 01/14/2023] Open
Abstract
Purpose Gap junction channels exhibit connexin specific biophysical properties, including the selective intercellular passage of larger solutes, such as second messengers. Here, we have examined the cyclic nucleotide permeability of the lens connexins, which could influence events like epithelial cell division and differentiation. Methods We compared the cAMP permeability through channels composed of Cx43, Cx46, or Cx50 using simultaneous measurements of junctional conductance and intercellular transfer. For cAMP detection, the recipient cells were transfected with a cAMP sensor gene, the cyclic nucleotide-modulated channel from sea urchin sperm (SpIH). cAMP was introduced via patch pipette into the cell of the pair that did not express SpIH. SpIH-derived currents were recorded from the other cell of a pair that expressed SpIH. cAMP permeability was also directly visualized in transfected cells using a chemically modified fluorescent form of the molecule. Results cAMP transfer was observed for homotypic Cx43 channels over a wide range of junctional conductance. Homotypic Cx46 channels also transferred cAMP, but permeability was reduced compared with Cx43. In contrast, homotypic Cx50 channels exhibited extremely low permeability to cAMP, when compared with either Cx43, or Cx46. Conclusions These data show that channels made from Cx43 and Cx46 result in the intercellular delivery of cAMP in sufficient quantity to activate cyclic nucleotide-modulated channels. The data also suggest that the greatly reduced cAMP permeability of Cx50 channels could play a role in the regulation of cell division in the lens.
Collapse
Affiliation(s)
- Virginijus Valiunas
- The Renaissance Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, New York, United States
| | - Peter R. Brink
- The Renaissance Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, New York, United States
| | - Thomas W. White
- The Renaissance Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, New York, United States
| |
Collapse
|
27
|
Sciuto KJ, Deng SW, Moreno A, Zaitsev AV. Chronology of critical events in neonatal rat ventricular myocytes occurring during reperfusion after simulated ischemia. PLoS One 2019; 14:e0212076. [PMID: 30730997 PMCID: PMC6366697 DOI: 10.1371/journal.pone.0212076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/25/2019] [Indexed: 11/19/2022] Open
Abstract
While an ischemic insult poses a lethal danger to myocardial cells, a significant proportion of cardiac myocytes remain viable throughout the ischemic episode and die, paradoxically, only after the blood flow is reinstated. Despite decades of research, the actual chronology of critical events leading to cardiomyocyte death during the reperfusion phase remains poorly understood. Arguably, identification of the pivotal event in this setting is necessary to design effective strategies aimed at salvaging the myocardium after an ischemic attack. Here we used neonatal rat ventricular myocytes (NRVMs) subjected to 20–30 min of simulated ischemia followed by 1 hour of “reperfusion”. Using different combinations of spectrally-compatible fluorescent indicators, we analyzed the relative timing of the following events: (1) abnormal increase in cytoplasmic [Ca2+] (TCaCy); (2) abnormal increase in mitochondrial [Ca2+] (TCaMi); (3) loss of mitochondrial inner membrane potential (ΔΨm) indicating mitochondrial permeability transitions (TMPT); (4) sacrolemmal permeabilization (SP) to the normally impermeable small fluorophore TO-PRO3 (TSP). In additional experiments we also analyzed the timing of abnormal uptake of Zn2+ into the cytoplasm (TZnCy) relative to TCaCy and TSP. We focused on those NRVMs which survived anoxia, as evidenced by at least 50% recovery of ΔΨm and the absence of detectable SP. In these cells, we found a consistent sequence of critical events in the order, from first to last, of TCaCy, TCaMi, TMPT, TSP. After detecting TCaCy and TCaMi, abrupt switches between 1.1 mM and nominally zero [Ca2+] in the perfusate quickly propagated to the cytoplasmic and mitochondrial [Ca2+]. Depletion of the sarcoplasmic reticulum with ryanodine (5 μM)/thapsigargin (1 μM) accelerated all events without changing their order. In the presence of ZnCl2 (10–30 μM) in the perfusate we found a consistent timing sequence TCaCy < TZn ≤ TSP. In some cells ZnCl2 interfered with Ca2+ uptake, causing “steps” or “gaps” in the [Ca2+]Cy curve, a phenomenon never observed in the absence of ZnCl2. Together, these findings suggest an evolving permeabilization of NRVM’s sarcolemma during reoxygenation, in which the expansion of the pore size determines the timing of critical events, including TMPT.
Collapse
Affiliation(s)
- Katie J. Sciuto
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Steven W. Deng
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Alonso Moreno
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - Alexey V. Zaitsev
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
28
|
A Barter Economy in Tumors: Exchanging Metabolites through Gap Junctions. Cancers (Basel) 2019; 11:cancers11010117. [PMID: 30669506 PMCID: PMC6356692 DOI: 10.3390/cancers11010117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 02/07/2023] Open
Abstract
To produce physiological functions, many tissues require their cells to be connected by gap junctions. Such diffusive coupling is important in establishing a cytoplasmic syncytium through which cells can exchange signals, substrates and metabolites. Often the benefits of connectivity become apparent solely at the multicellular level, leading to the notion that cells work for a common good rather than exclusively in their self-interest. In some tumors, gap junctional connectivity between cancer cells is reduced or absent, but there are notable cases where it persists or re-emerges in late-stage disease. Diffusive coupling will blur certain phenotypic differences between cells, which may seem to go against the establishment of population heterogeneity, a central pillar of cancer that stems from genetic instability. Here, building on our previous measurements of gap junctional coupling between cancer cells, we use a computational model to simulate the role of connexin-assembled channels in exchanging lactate and bicarbonate ions down their diffusion gradients. Based on the results of these simulations, we propose that an overriding benefit of gap junctional connectivity may relate to lactate/bicarbonate exchange, which would support an elevated metabolic rate in hypoxic tumors. In this example of barter, hypoxic cancer cells provide normoxic neighbors with lactate for mitochondrial oxidation; in exchange, bicarbonate ions, which are more plentiful in normoxic cells, are supplied to hypoxic neighbors to neutralize the H+ ions co-produced glycolytically. Both cells benefit, and so does the tumor.
Collapse
|
29
|
Connexin 43 (Cx43) in cancer: Implications for therapeutic approaches via gap junctions. Cancer Lett 2018; 442:439-444. [PMID: 30472182 DOI: 10.1016/j.canlet.2018.10.043] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/30/2018] [Accepted: 10/25/2018] [Indexed: 01/11/2023]
Abstract
Gap junctions are membrane channels found in all cells of the human body that are essential to cellular physiology. Gap junctions are formed from connexin proteins and are responsible for transfer of biologically active molecules, metabolites, and salts between neighboring cells or cells and their extracellular environment. Over the last few years, aberrant connexin 43 (Cx43) expression has been associated with cancer recurrence, metastatic spread, and poor survival. Here we provide an overview of the general structure and function of gap junctions and review their roles in different cancer types. We discuss new therapeutic approaches targeting Cx43 and potential new ways of exploiting gap junction transfer for drug delivery and anti-cancer treatment. The permeability of Cx43 channels to small molecules and macromolecules makes them highly attractive targets for delivering drugs directly into the cytoplasm. Cancer cells overexpressing Cx43 may be more permeable and sensitive to chemotherapeutics. Because Cx43 can either act as a tumor suppressor or oncogene, biomarker analysis and a better understanding of how Cx43 contextually mediates cancer phenotypes will be required to develop clinically viable Cx43-based therapies.
Collapse
|
30
|
Pietak A, Levin M. Bioelectrical control of positional information in development and regeneration: A review of conceptual and computational advances. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 137:52-68. [PMID: 29626560 PMCID: PMC10464501 DOI: 10.1016/j.pbiomolbio.2018.03.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 12/16/2022]
Abstract
Positional information describes pre-patterns of morphogenetic substances that alter spatio-temporal gene expression to instruct development of growth and form. A wealth of recent data indicate bioelectrical properties, such as the transmembrane potential (Vmem), are involved as instructive signals in the spatiotemporal regulation of morphogenesis. However, the mechanistic relationships between Vmem and molecular positional information are only beginning to be understood. Recent advances in computational modeling are assisting in the development of comprehensive frameworks for mechanistically understanding how endogenous bioelectricity can guide anatomy in a broad range of systems. Vmem represents an extraordinarily strong electric field (∼1.0 × 106 V/m) active over the thin expanse of the plasma membrane, with the capacity to influence a variety of downstream molecular signaling cascades. Moreover, in multicellular networks, intercellular coupling facilitated by gap junction channels may induce directed, electrodiffusive transport of charged molecules between cells of the network to generate new positional information patterning possibilities and characteristics. Given the demonstrated role of Vmem in morphogenesis, here we review current understanding of how Vmem can integrate with molecular regulatory networks to control single cell state, and the unique properties bioelectricity adds to transport phenomena in gap junction-coupled cell networks to facilitate self-assembly of morphogen gradients and other patterns. Understanding how Vmem integrates with biochemical regulatory networks at the level of a single cell, and mechanisms through which Vmem shapes molecular positional information in multicellular networks, are essential for a deep understanding of body plan control in development, regeneration and disease.
Collapse
Affiliation(s)
| | - Michael Levin
- Allen Discovery Center at Tufts, USA; Center for Regenerative and Developmental Biology, Tufts University, Medford, MA, USA
| |
Collapse
|
31
|
Fasciani I, Pluta P, González-Nieto D, Martínez-Montero P, Molano J, Paíno CL, Millet O, Barrio LC. Directional coupling of oligodendrocyte connexin-47 and astrocyte connexin-43 gap junctions. Glia 2018; 66:2340-2352. [PMID: 30144323 DOI: 10.1002/glia.23471] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/21/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022]
Abstract
Intercellular communication via gap junction channels between oligodendrocytes and between astrocytes as well as between these cell types is essential to maintain the integrity of myelin in the central nervous system. Oligodendrocyte gap junction connexin-47 (Cx47) is a key element in this crosstalk and indeed, mutations in human Cx47 cause severe myelin disorders. However, the permeation properties of channels of Cx47 alone and in heterotypic combination with astrocyte Cx43 remain unclear. We show here that Cx47 contains three extra residues at 5' amino-terminus that play a critical role in the channel pore structure and account for relative low ionic conductivity, cationic permselectivity and voltage-gating properties of oligodendrocyte-oligodendrocyte Cx47 channels. Regarding oligodendrocyte-astrocyte coupling, heterotypic channels formed by Cx47 with Cx43 exhibit ionic and chemical rectification, which creates a directional diffusion barrier for the movement of ions and larger negatively charged molecules from cells expressing Cx47 to those with Cx43. The restrictive permeability of Cx47 channels and the diffusion barrier of Cx47-Cx43 channels was abolished by a mutation associated with leukodystrophy, the Cx47P90S, suggesting a novel pathogenic mechanism underlying myelin disorders that involves alterations in the panglial permeation.
Collapse
Affiliation(s)
- Ilaria Fasciani
- Unit of Experimental Neurology and Neurobiology, "Ramón y Cajal" Hospital-IRYCIS, Carretera de Colmenar km 9, Madrid, 28034, Spain
| | - Paula Pluta
- Structural Biology Unit of CIC bioGUNE, Bizkaia Technology Park, Building 800, Derio, 48160, Spain
| | - Daniel González-Nieto
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, and Center for Biomedical Technology, Universidad Politécnica de Madrid, Campus de Montegancedo S/N, Pozuelo de Alarcón, Madrid, 28223, Spain
| | - Paloma Martínez-Montero
- Unit of Molecular Genetics-INGEMM, Hospital "La Paz"-IDIPAZ, Paseo de la Castellana 261, 28046-Madrid, Spain
| | - Jesús Molano
- Unit of Molecular Genetics-INGEMM, Hospital "La Paz"-IDIPAZ, Paseo de la Castellana 261, 28046-Madrid, Spain
| | - Carlos L Paíno
- Unit of Experimental Neurology and Neurobiology, "Ramón y Cajal" Hospital-IRYCIS, Carretera de Colmenar km 9, Madrid, 28034, Spain
| | - Oscar Millet
- Structural Biology Unit of CIC bioGUNE, Bizkaia Technology Park, Building 800, Derio, 48160, Spain
| | - Luis C Barrio
- Unit of Experimental Neurology and Neurobiology, "Ramón y Cajal" Hospital-IRYCIS, Carretera de Colmenar km 9, Madrid, 28034, Spain
| |
Collapse
|
32
|
Li SKL, Shan SW, Li HL, Cheng AKW, Pan F, Yip SP, Civan MM, To CH, Do CW. Characterization and Regulation of Gap Junctions in Porcine Ciliary Epithelium. ACTA ACUST UNITED AC 2018; 59:3461-3468. [DOI: 10.1167/iovs.18-24682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Stanley Ka-Lok Li
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Sze-Wan Shan
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Hoi-Lam Li
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Angela King-Wah Cheng
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Feng Pan
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Shea-Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Mortimer M. Civan
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Chi-Ho To
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Chi-Wai Do
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| |
Collapse
|
33
|
Valiunas V, Cohen IS, Brink PR. Defining the factors that affect solute permeation of gap junction channels. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2018; 1860:96-101. [PMID: 28690048 PMCID: PMC5705451 DOI: 10.1016/j.bbamem.2017.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 01/22/2023]
Abstract
This review focuses on the biophysical properties and structure of the pore and vestibule of homotypic gap junction channels as they relate to channel permeability and selectivity. Gap junction channels are unique in their sole role to connect the cytoplasm of two adjacent cells. In general, these channels are considered to be poorly selective, possess open probabilities approximating unity, and exhibit mean open times ranging from milliseconds to seconds. These properties suggest that such channels can function as delivery pathways from cell to cell for solutes that are significantly larger than monovalent ions. We have taken quantitative data from published works concerning unitary conductance, ion flux, and permeability for homotypic connexin 43 (Cx43), Cx40, Cx26, Cx50, and Cx37, and performed a comparative analysis of conductance and/or ion/solute flux versus diffusion coefficient. The analysis of monovalent cation flux portrays the pore as equivalent to an aqueous space where hydrogen bonding and weak interactions with binding sites dominate. For larger solutes, size, shape and charge are also significant components in determining the permeation rate. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Virginijus Valiunas
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA.
| | - Ira S Cohen
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Peter R Brink
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
34
|
Nielsen BS, Alstrom JS, Nicholson BJ, Nielsen MS, MacAulay N. Permeant-specific gating of connexin 30 hemichannels. J Biol Chem 2017; 292:19999-20009. [PMID: 28982982 PMCID: PMC5723989 DOI: 10.1074/jbc.m117.805986] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/28/2017] [Indexed: 11/06/2022] Open
Abstract
Gap junctions confer interconnectivity of the cytoplasm in neighboring cells via docking of two connexons expressed in each of the adjacent membranes. Undocked connexons, referred to as hemichannels, may open and connect the cytoplasm with the extracellular fluid. The hemichannel configuration of connexins (Cxs) displays isoform-specific permeability profiles that are not directly determined by the size and charge of the permeant. To further explore Ca2+-mediated gating and permeability features of connexin hemichannels, we heterologously expressed Cx30 hemichannels in Xenopus laevis oocytes. The sensitivity toward divalent cation-mediated gating differed between small atomic ions (current) and fluorescent dye permeants, indicating that these permeants are distinctly gated. Three aspartate residues in Cx30 (Asp-50, Asp-172, and Asp-179) have been implicated previously in the Ca2+ sensitivity of other hemichannel isoforms. Although the aspartate at position Asp-50 was indispensable for divalent cation-dependent gating of Cx30 hemichannels, substitutions of the two other residues had no significant effect on gating, illustrating differences in the gating mechanisms between connexin isoforms. Using the substituted cysteine accessibility method (SCAM), we evaluated the role of possible pore-lining residues in the permeation of ions and ethidium through Cx30 hemichannels. Of the cysteine-substituted residues, interaction of a proposed pore-lining cysteine at position 37 with the positively charged compound [2-(trimethylammonium)ethyl] methane thiosulfonate bromide (MTS-ET) increased Cx30-mediated currents with unperturbed ethidium permeability. In summary, our results demonstrate that the permeability of hemichannels is regulated in a permeant-specific manner and underscores that hemichannels are selective rather than non-discriminating and freely diffusable pores.
Collapse
Affiliation(s)
| | | | - Bruce J Nicholson
- Department of Biochemistry, School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Morten Schak Nielsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | | |
Collapse
|
35
|
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: 179] [Impact Index Per Article: 22.4] [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
|
36
|
Decrock E, Hoorelbeke D, Ramadan R, Delvaeye T, De Bock M, Wang N, Krysko DV, Baatout S, Bultynck G, Aerts A, Vinken M, Leybaert L. Calcium, oxidative stress and connexin channels, a harmonious orchestra directing the response to radiotherapy treatment? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1099-1120. [DOI: 10.1016/j.bbamcr.2017.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/02/2017] [Accepted: 02/04/2017] [Indexed: 02/07/2023]
|
37
|
Zhong G, Akoum N, Appadurai DA, Hayrapetyan V, Ahmed O, Martinez AD, Beyer EC, Moreno AP. Mono-Heteromeric Configurations of Gap Junction Channels Formed by Connexin43 and Connexin45 Reduce Unitary Conductance and Determine both Voltage Gating and Metabolic Flux Asymmetry. Front Physiol 2017; 8:346. [PMID: 28611680 PMCID: PMC5447054 DOI: 10.3389/fphys.2017.00346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/11/2017] [Indexed: 11/29/2022] Open
Abstract
In cardiac tissues, the expression of multiple connexins (Cx40, Cx43, Cx45, and Cx30.2) is a requirement for proper development and function. Gap junctions formed by these connexins have distinct permeability and gating mechanisms. Since a single cell can express more than one connexin isoform, the formation of hetero-multimeric gap junction channels provides a tissue with an enormous repertoire of combinations to modulate intercellular communication. To study further the perm-selectivity and gating properties of channels containing Cx43 and Cx45, we studied two monoheteromeric combinations in which a HeLa cell co-transfected with Cx43 and Cx45 was paired with a cell expressing only one of these connexins. Macroscopic measurements of total conductance between cell pairs indicated a drastic reduction in total conductance for mono-heteromeric channels. In terms of Vj dependent gating, Cx43 homomeric connexons facing heteromeric connexons only responded weakly to voltage negativity. Cx45 homomeric connexons exhibited no change in Vj gating when facing heteromeric connexons. The distributions of unitary conductances (γj) for both mono-heteromeric channels were smaller than predicted, and both showed low permeability to the fluorescent dyes Lucifer yellow and Rhodamine123. For both mono-heteromeric channels, we observed flux asymmetry regardless of dye charge: flux was higher in the direction of the heteromeric connexon for MhetCx45 and in the direction of the homomeric Cx43 connexon for MhetCx43. Thus, our data suggest that co-expression of Cx45 and Cx43 induces the formation of heteromeric connexons with greatly reduced permeability and unitary conductance. Furthermore, it increases the asymmetry for voltage gating for opposing connexons, and it favors asymmetric flux of molecules across the junction that depends primarily on the size (not the charge) of the crossing molecules.
Collapse
Affiliation(s)
- Guoqiang Zhong
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical UniversityGuangxi, China
| | - Nazem Akoum
- University Medical Center, University of WashingtonSeattle, WA, United States
| | | | | | - Osman Ahmed
- Atlanta Heart SpecialistsAtlanta, GA, United States
| | - Agustin D Martinez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| | - Eric C Beyer
- Department of Pediatrics, University of ChicagoChicago, IL, United States
| | - Alonso P Moreno
- Cardiovascular Research and Training Institute (CVRTI), Department of Bioengineering, University of UtahSalt Lake Citiy, UT, United States
| |
Collapse
|
38
|
Rodriguez-Grande B, Konsman JP. Gas Diffusion in the CNS. J Neurosci Res 2017; 96:207-218. [PMID: 28504343 DOI: 10.1002/jnr.24077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/23/2017] [Accepted: 04/10/2017] [Indexed: 12/21/2022]
Abstract
Gases have been long known to have essential physiological functions in the CNS such as respiration or regulation of vascular tone. Since gases have been classically considered to freely diffuse, research in gas biology has so far focused on mechanisms of gas synthesis and gas reactivity, rather than gas diffusion and transport. However, the discovery of gas pores during the last two decades and the characterization of diverse diffusion patterns through different membranes has raised the possibility that modulation of gas diffusion is also a physiologically relevant parameter. Here we review the means of gas movement into and within the brain through "free" diffusion and gas pores, notably aquaporins, discussing the role that gas diffusion may play in the modulation of gas function. We highlight how diffusion is relevant to neuronal signaling, volume transmission, and cerebrovascular control in the case of NO, one of the most extensively studied gases. We point out how facilitated transport can be especially relevant for gases with low permeability in lipid membranes like NH3 and discuss the possible implications of NH3 -permeable channels in physiology and hyperammonemic encephalopathy. We identify novel research questions about how modulation of gas diffusion could intervene in CNS pathologies. This emerging area of research can provide novel and interesting insights in the field of gas biology.
Collapse
|
39
|
Mondal A, Sachse FB, Moreno AP. Modulation of Asymmetric Flux in Heterotypic Gap Junctions by Pore Shape, Particle Size and Charge. Front Physiol 2017; 8:206. [PMID: 28428758 PMCID: PMC5382223 DOI: 10.3389/fphys.2017.00206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/20/2017] [Indexed: 01/26/2023] Open
Abstract
Gap junction channels play a vital role in intercellular communication by connecting cytoplasm of adjoined cells through arrays of channel-pores formed at the common membrane junction. Their structure and properties vary depending on the connexin isoform(s) involved in forming the full gap junction channel. Lack of information on the molecular structure of gap junction channels has limited the development of computational tools for single channel studies. Currently, we rely on cumbersome experimental techniques that have limited capabilities. We have earlier reported a simplified Brownian dynamics gap junction pore model and demonstrated that variations in pore shape at the single channel level can explain some of the differences in permeability of heterotypic channels observed in in vitro experiments. Based on this computational model, we designed simulations to study the influence of pore shape, particle size and charge in homotypic and heterotypic pores. We simulated dye diffusion under whole cell voltage clamping. Our simulation studies with pore shape variations revealed a pore shape with maximal flux asymmetry in a heterotypic pore. We identified pore shape profiles that match the in silico flux asymmetry results to the in vitro results of homotypic and heterotypic gap junction formed out of Cx43 and Cx45. Our simulation results indicate that the channel's pore-shape established flux asymmetry and that flux asymmetry is primarily regulated by the sizes of the conical and/or cylindrical mouths at each end of the pore. Within the set range of particle size and charge, flux asymmetry was found to be independent of particle size and directly proportional to charge magnitude. While particle charge was vital to creating flux asymmetry, charge magnitude only scaled the observed flux asymmetry. Our studies identified the key factors that help predict asymmetry. Finally, we suggest the role of such flux asymmetry in creating concentration imbalances of messenger molecules in cardiomyocytes. We also assess the potency of fibroblasts in aggravating such imbalances through Cx43-Cx45 heterotypic channels in fibrotic heart tissue.
Collapse
Affiliation(s)
- Abhijit Mondal
- Department of Bioengineering, University of UtahSalt Lake City, UT, USA.,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake City, UT, USA
| | - Frank B Sachse
- Department of Bioengineering, University of UtahSalt Lake City, UT, USA.,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake City, UT, USA
| | - Alonso P Moreno
- Department of Bioengineering, University of UtahSalt Lake City, UT, USA.,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake City, UT, USA.,Department of Internal Medicine, Cardiology, University of UtahSalt Lake City, UT, USA
| |
Collapse
|
40
|
Thomas NM, Gray R, Fry CH, Desplantez T, Peters NS, Severs NJ, Macleod KT, Dupont E. Functional consequences of co-expressing connexin40 or connexin45 with connexin43 on intercellular electrical coupling. Biochem Biophys Res Commun 2017; 483:191-196. [PMID: 28034749 DOI: 10.1016/j.bbrc.2016.12.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 12/25/2016] [Indexed: 11/26/2022]
Abstract
The functional characteristics of the co-expression of connexin43, connexin40, and connexin45 proteins in human myocardium are thought to play an important role in governing normal propagation of the cardiac electrical impulse and in generating the myocardial substrate for some arrhythmias and conduction disturbances. A rat liver epithelial cell line, that endogenously expresses connexin43, was used to induce also expression of connexin40 or connexin45 after stable transfection using an inducible ecdysone system. Electrical coupling was estimated from measurement of the input resistance of transfected cells using an intracellular microelectrode to inject current and record changes to membrane potential. However, varied expression of the transfected connexin40 or connexin45 did not change electrical coupling, although connexin43/40 co-expression led to better coupling than connexin43/45 co-expression. Quantification of endogenous connexin43 expression, at both mRNA and protein levels, showed that it was altered in a manner dependent on the transfected connexin isotype. The data using rat liver epithelial cells indicate an increased electrical coupling upon expression of connexin40 and connexin43 but decreased coupling with connexin45 and connexin43 co-expression.
Collapse
Affiliation(s)
- Neil M Thomas
- Myocardial Function, National Heart and Lung Institute, Imperial College London, London, UK.
| | | | - Christopher H Fry
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK.
| | - Thomas Desplantez
- Myocardial Function, National Heart and Lung Institute, Imperial College London, London, UK.
| | - Nicholas S Peters
- Myocardial Function, National Heart and Lung Institute, Imperial College London, London, UK.
| | - Nicholas J Severs
- Myocardial Function, National Heart and Lung Institute, Imperial College London, London, UK.
| | - Kenneth T Macleod
- Myocardial Function, National Heart and Lung Institute, Imperial College London, London, UK.
| | - Emmanuel Dupont
- Myocardial Function, National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
41
|
Computational Studies of Molecular Permeation through Connexin26 Channels. Biophys J 2017; 110:584-599. [PMID: 26840724 DOI: 10.1016/j.bpj.2015.11.3528] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/21/2015] [Accepted: 11/23/2015] [Indexed: 11/22/2022] Open
Abstract
A signal property of connexin channels is the ability to mediate selective diffusive movement of molecules through plasma membrane(s), but the energetics and determinants of molecular movement through these channels have yet to be understood. Different connexin channels have distinct molecular selectivities that cannot be explained simply on the basis of size or charge of the permeants. To gain insight into the forces and interactions that underlie selective molecular permeation, we investigated the energetics of two uncharged derivatized sugars, one permeable and one impermeable, through a validated connexin26 (Cx26) channel structural model, using molecular dynamics and associated analytic tools. The system is a Cx26 channel equilibrated in explicit membrane/solvent, shown by Brownian dynamics to reproduce key conductance characteristics of the native channel. The results are consistent with the known difference in permeability to each molecule. The energetic barriers extend through most of the pore length, rather than being highly localized as in ion-specific channels. There is little evidence for binding within the pore. Force decomposition reveals how, for each tested molecule, interactions with water and the Cx26 protein vary over the length of the pore and reveals a significant contribution from hydrogen bonding and interaction with K(+). The flexibility of the pore width varies along its length, and the tested molecules have differential effects on pore width as they pass through. Potential sites of interaction within the pore are defined for each molecule. The results suggest that for the tested molecules, differences in hydrogen bonding and entropic factors arising from permeant flexibility substantially contribute to the energetics of permeation. This work highlights factors involved in selective molecular permeation that differ from those that define selectivity among atomic ions.
Collapse
|
42
|
Computational simulations of asymmetric fluxes of large molecules through gap junction channel pores. J Theor Biol 2016; 412:61-73. [PMID: 27590324 DOI: 10.1016/j.jtbi.2016.08.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 06/30/2016] [Accepted: 08/30/2016] [Indexed: 02/05/2023]
Abstract
Gap junction channels are formed out of connexin isoforms, which enable molecule and ion selective diffusion amongst neighboring cells. HeLa cells expressing distinct connexins (Cx) allow the formation of heterotypic channels, where we observed a molecular charge-independent preferential flux of large fluorescent molecules in the Cx45 to Cx43 direction. We hypothesize that the pore's shape is a significant factor along-side charge and transjunctional voltages for this asymmetric flux. To test this hypothesis, we developed a 3D computational model simulating Brownian diffusion of large molecules in a gap junction channel pore. The basic pore contour was derived from x-ray crystallographic structures of Cx43 and Cx26 and approximated using basic geometric shapes. Lucifer yellow dye molecules and cesium counter-ions were modeled as spheres using their respective Stokes radii. Our simulation results from simple diffusion and constant concentration gradient experiments showed that only charged particles yield asymmetric fluxes in heterotypic pores. While increasing the inner mouth size resulted in a near-quadratic rise in flux, the rise was asymptotic for outer mouth radii increase. Probability maps and average force per particle per pore section explain the asymmetric flux with variation in pore shape. Furthermore, the simulation results are in agreement with our in vitro experimental results with HeLa cells in Cx43-Cx45 heterotypic configurations. The presence of asymmetric fluxes can help us to understand effects of the molecular structure of the pore and predict potential differences in vivo.
Collapse
|
43
|
Li S, Peng W, Chen X, Geng X, Zhan W, Sun J. Expression and role of gap junction protein connexin43 in immune challenge-induced extracellular ATP release in Japanese flounder (Paralichthys olivaceus). FISH & SHELLFISH IMMUNOLOGY 2016; 55:348-357. [PMID: 27291350 DOI: 10.1016/j.fsi.2016.06.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/02/2016] [Accepted: 06/08/2016] [Indexed: 06/06/2023]
Abstract
Connexin43 (Cx43) is the best characterized gap junction protein that allows the direct exchange of signaling molecules during cell-to-cell communications. The immunological functions and ATP permeable properties of Cx43 have been insensitively examined in mammals. The similar biological significance of Cx43 in lower vertebrates, however, is not yet understood. In the present study we identified and characterized a Cx43 ortholog (termed PoCx43) from Japanese flounder (Paralichthys olivaceus) and investigated its role in immune challenge-induced extracellular ATP release. PoCx43 mRNA transcripts are widely distributed in all tested normal tissues and cells with predominant expression in the brain, and are significantly up-regulated by LPS, poly(I:C) and zymosan challenges and Edwardsiella tarda infections as well, suggesting that PoCx43 expression was modulated by the inflammatory stresses. In addition, cyclic AMP (cAMP), an essential second messenger, also plays an important role in regulating PoCx43 gene expression, by which the PoCx43-mediated gap junctional communication may be regulated. Furthermore, overexpression of PoCx43 in Japanese flounder FG-9307 cells significantly potentiates the LPS- and poly(I:C)-induced extracellular ATP release and this enhanced ATP release was attenuated by pre-incubation with Cx43 inhibitor carbenoxolone. In a complementary experiment, down-regulation of PoCx43 endogenous expression in FG-9307 cells with small interfering RNA also significantly reduced the PAMP-induced extracellular ATP release, suggesting that PoCx43 is an important ATP release conduit under the immune challenge conditions. Finally, we showed that extracellular ATP stimulation led to an increased PoCx43 expression which probably provides a feedback mechanism in regulating PoCx43 expression at the transcriptional level. These findings suggest that PoCx43 is an inducible immune response gene and an important conduit for immune challenge-induced extracellular ATP release in fish.
Collapse
Affiliation(s)
- Shuo Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China.
| | - Weijiao Peng
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Xiaoli Chen
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China
| | - Xuyun Geng
- Tianjin Center for Control and Prevention of Aquatic Animal Infectious Disease, 442 South Jiefang Road, Hexi District, Tianjin 300221, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, LMMEC, Ocean University of China, Qingdao 266003, China
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin 300387, China.
| |
Collapse
|
44
|
Rash JE, Vanderpool KG, Yasumura T, Hickman J, Beatty JT, Nagy JI. KV1 channels identified in rodent myelinated axons, linked to Cx29 in innermost myelin: support for electrically active myelin in mammalian saltatory conduction. J Neurophysiol 2016; 115:1836-59. [PMID: 26763782 PMCID: PMC4869480 DOI: 10.1152/jn.01077.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/04/2016] [Indexed: 11/22/2022] Open
Abstract
Saltatory conduction in mammalian myelinated axons was thought to be well understood before recent discoveries revealed unexpected subcellular distributions and molecular identities of the K(+)-conductance pathways that provide for rapid axonal repolarization. In this study, we visualize, identify, localize, quantify, and ultrastructurally characterize axonal KV1.1/KV1.2 channels in sciatic nerves of rodents. With the use of light microscopic immunocytochemistry and freeze-fracture replica immunogold labeling electron microscopy, KV1.1/KV1.2 channels are localized to three anatomically and compositionally distinct domains in the internodal axolemmas of large myelinated axons, where they form densely packed "rosettes" of 9-nm intramembrane particles. These axolemmal KV1.1/KV1.2 rosettes are precisely aligned with and ultrastructurally coupled to connexin29 (Cx29) channels, also in matching rosettes, in the surrounding juxtaparanodal myelin collars and along the inner mesaxon. As >98% of transmembrane proteins large enough to represent ion channels in these specialized domains, ∼500,000 KV1.1/KV1.2 channels define the paired juxtaparanodal regions as exclusive membrane domains for the voltage-gated K(+)conductance that underlies rapid axonal repolarization in mammals. The 1:1 molecular linkage of KV1 channels to Cx29 channels in the apposed juxtaparanodal collars, plus their linkage to an additional 250,000-400,000 Cx29 channels along each inner mesaxon in every large-diameter myelinated axon examined, supports previously proposed K(+)conductance directly from juxtaparanodal axoplasm into juxtaparanodal myeloplasm in mammalian axons. With neither Cx29 protein nor myelin rosettes detectable in frog myelinated axons, these data showing axon-to-myelin linkage by abundant KV1/Cx29 channels in rodent axons support renewed consideration of an electrically active role for myelin in increasing both saltatory conduction velocity and maximum propagation frequency in mammalian myelinated axons.
Collapse
Affiliation(s)
- John E Rash
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado; Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, Colorado; and
| | - Kimberly G Vanderpool
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Thomas Yasumura
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jordan Hickman
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jonathan T Beatty
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - James I Nagy
- Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
45
|
Steele S, Radlinski L, Taft-Benz S, Brunton J, Kawula TH. Trogocytosis-associated cell to cell spread of intracellular bacterial pathogens. eLife 2016; 5. [PMID: 26802627 PMCID: PMC4786427 DOI: 10.7554/elife.10625] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 01/22/2016] [Indexed: 11/13/2022] Open
Abstract
Macrophages are myeloid-derived phagocytic cells and one of the first immune cell types to respond to microbial infections. However, a number of bacterial pathogens are resistant to the antimicrobial activities of macrophages and can grow within these cells. Macrophages have other immune surveillance roles including the acquisition of cytosolic components from multiple types of cells. We hypothesized that intracellular pathogens that can replicate within macrophages could also exploit cytosolic transfer to facilitate bacterial spread. We found that viable Francisella tularensis, as well as Salmonella enterica bacteria transferred from infected cells to uninfected macrophages along with other cytosolic material through a transient, contact dependent mechanism. Bacterial transfer occurred when the host cells exchanged plasma membrane proteins and cytosol via a trogocytosis related process leaving both donor and recipient cells intact and viable. Trogocytosis was strongly associated with infection in mice, suggesting that direct bacterial transfer occurs by this process in vivo.
Collapse
Affiliation(s)
- Shaun Steele
- University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Lauren Radlinski
- University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Sharon Taft-Benz
- University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Jason Brunton
- University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Thomas H Kawula
- University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
46
|
An electrostatic mechanism for Ca(2+)-mediated regulation of gap junction channels. Nat Commun 2016; 7:8770. [PMID: 26753910 PMCID: PMC4730032 DOI: 10.1038/ncomms9770] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 10/01/2015] [Indexed: 01/07/2023] Open
Abstract
Gap junction channels mediate intercellular signalling that is crucial in tissue development, homeostasis and pathologic states such as cardiac arrhythmias, cancer and trauma. To explore the mechanism by which Ca(2+) blocks intercellular communication during tissue injury, we determined the X-ray crystal structures of the human Cx26 gap junction channel with and without bound Ca(2+). The two structures were nearly identical, ruling out both a large-scale structural change and a local steric constriction of the pore. Ca(2+) coordination sites reside at the interfaces between adjacent subunits, near the entrance to the extracellular gap, where local, side chain conformational rearrangements enable Ca(2+)chelation. Computational analysis revealed that Ca(2+)-binding generates a positive electrostatic barrier that substantially inhibits permeation of cations such as K(+) into the pore. Our results provide structural evidence for a unique mechanism of channel regulation: ionic conduction block via an electrostatic barrier rather than steric occlusion of the channel pore.
Collapse
|
47
|
Qiao M, Sanes JR. Genetic Method for Labeling Electrically Coupled Cells: Application to Retina. Front Mol Neurosci 2016; 8:81. [PMID: 26778956 PMCID: PMC4703850 DOI: 10.3389/fnmol.2015.00081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 12/11/2015] [Indexed: 01/08/2023] Open
Abstract
Understanding how the nervous system functions requires mapping synaptic connections between neurons. Several methods are available for imaging neurons connected by chemical synapses, but few enable marking neurons connected by electrical synapses. Here, we demonstrate that a peptide transporter, Pept2, can be used for this purpose. Pept2 transports a gap junction-permeable fluorophore-coupled dipeptide, beta-alanine-lysine-N-7-amino-4-methyl coumarin-3-acid (βALA). Cre-dependent expression of pept2 in specific neurons followed by incubation in βALA labeled electrically coupled synaptic partners. Using this method, we analyze light-dependent modulation of electrical connectivity among retinal horizontal cells.
Collapse
Affiliation(s)
- Mu Qiao
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, USA
| |
Collapse
|
48
|
Electroporation Loading and Dye Transfer: A Safe and Robust Method to Probe Gap Junctional Coupling. Methods Mol Biol 2016; 1437:155-69. [PMID: 27207293 DOI: 10.1007/978-1-4939-3664-9_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intercellular communication occurring via gap junction channels is considered a key mechanism for synchronizing physiological functions of cells and for the maintenance of tissue homeostasis. Gap junction channels are protein channels that are situated between neighboring cells and that provide a direct, yet selective route for the passage of small hydrophilic biomolecules and ions. Here, an electroporation method is described to load a localized area within an adherent cell monolayer with a gap junction-permeable fluorescent reporter dye. The technique results in a rapid and efficient labeling of a small patch of cells within the cell culture, without affecting cellular viability. Dynamic and quantitative information on gap junctional communication can subsequently be extracted by tracing the intercellular movement of the dye via time-lapse microscopy.
Collapse
|
49
|
Relating specific connexin co-expression ratio to connexon composition and gap junction function. J Mol Cell Cardiol 2015; 89:195-202. [PMID: 26550940 DOI: 10.1016/j.yjmcc.2015.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 10/05/2015] [Accepted: 11/04/2015] [Indexed: 12/15/2022]
Abstract
Cardiac connexin 43 (Cx43), Cx40 and Cx45 are co-expressed at distinct ratios in myocytes. This pattern is considered a key factor in regulating the gap junction channels composition, properties and function and remains poorly understood. This work aims to correlate gap junction function with the connexin composition of the channels at accurate ratios Cx43:Cx40 and Cx43:Cx45. Rat liver epithelial cells that endogenously express Cx43 were stably transfected to induce expression of accurate levels of Cx40 or Cx45 that may be present in various areas of the heart (e.g. atria and ventricular conduction system). Induction of Cx40 does not increase the amounts of junctional connexins (Cx43 and Cx40), whereas induction of Cx45 increases the amounts of junctional connexins (Cx43 and Cx45). Interestingly, the non-junctional fraction of Cx43 remains unaffected upon induction of Cx40 and Cx45. Co-immunoprecipitation studies show low level of Cx40/Cx43 heteromerisation and undetectable Cx45/Cx43 heteromerisation. Functional characterisation shows that induction of Cx40 and Cx45 decreases Lucifer Yellow transfer. Electrical coupling is decreased by Cx45 induction, whereas it is decreased at low induction of Cx40 and increased at high induction. These data indicate a fine regulation of the gap junction channel make-up in function of the type and the ratio of co-expressed Cxs that specifically regulates chemical and electrical coupling. This reflects specific gap junction function in regulating impulse propagation in the healthy heart, and a pro-arrhythmic potential of connexin remodelling in the diseased heart.
Collapse
|
50
|
Santa Cruz A, Meşe G, Valiuniene L, Brink PR, White TW, Valiunas V. Altered conductance and permeability of Cx40 mutations associated with atrial fibrillation. J Gen Physiol 2015; 146:387-98. [PMID: 26503720 PMCID: PMC4621748 DOI: 10.1085/jgp.201511475] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/23/2015] [Indexed: 11/20/2022] Open
Abstract
Gap junctions ensure the rapid propagation of the action potential throughout the myocardium. Three mutant forms of connexin40 (Cx40; A96S, M163V, and G38D), the primary component of the atrial gap junction channel, are associated with atrial fibrillation and retain the ability to form functional channels. We determined the biophysical properties of these mutant gap junctions in transiently transfected HeLa and N2A cells. All three mutants showed macroscopic junctional conductances over the range of 0.5 to 40 nS, and voltage dependences comparable to those of wild-type (WT) Cx40. However, the unitary conductance of G38D channels was ∼1.6-fold higher than that of WT Cx40 channels (∼220 vs. ∼135 pS), whereas the unitary conductances of the A96S and M163V mutants were similar to that of WT Cx40. Furthermore, the M163V and G38D channels exhibited approximately two- and approximately fivefold higher permeability to the anionic dye Lucifer yellow (LY) relative to K+ (LY/K+) compared with that of WT Cx40, whereas A96S LY transfer was similar to that of WT (G38D > M163V > A96S ≈ Cx40WT). In contrast, G38D channels were almost impermeable to cationic ethidium bromide (EtBr), suggesting that G38D alters channel selectivity. Conversely, A96S and M163V channels showed enhanced EtBr permeability relative to WT Cx40, with the following permeability order: M163V > A96S > Cx40WT > G38D. Altered conductive and permeability properties of mutant channels suggest an essential role for Cx40-mediated biochemical and electrical coupling in cardiac tissues. The altered properties of the three single-base substitution mutants may play a role in mechanisms of reentry arrhythmias.
Collapse
Affiliation(s)
- Ana Santa Cruz
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Gülistan Meşe
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Laima Valiuniene
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Peter R Brink
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Thomas W White
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Virginijus Valiunas
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| |
Collapse
|