1
|
Chen H, Li YX, Wong RS, Esseltine JL, Bai D. Genetically engineered human embryonic kidney cells as a novel vehicle for dual patch clamp study of human gap junction channels. Biochem J 2024; 481:741-758. [PMID: 38752978 PMCID: PMC11346430 DOI: 10.1042/bcj20240016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/29/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
Mutations in more than half of human connexin genes encoding gap junction (GJ) subunits have been linked to inherited human diseases. Functional studies of human GJ channels are essential for revealing mechanistic insights into the etiology of disease-linked connexin mutants. However, the commonly used Xenopus oocytes, N2A, HeLa, and other model cells for recombinant expression of human connexins have different and significant limitations. Here we developed a human cell line (HEK293) with each of the endogenous connexins (Cx43 and Cx45) knocked out using the CRISPR-Cas9 system. Double knockout HEK293 cells showed no background GJ coupling, were easily transfected with several human connexin genes (such as those encoding Cx46, Cx50, Cx37, Cx45, Cx26, and Cx36) which successfully formed functional GJs and were readily accessible for dual patch clamp analysis. Single knockout Cx43 or Cx45 HEK cell lines could also be used to characterize human GJ channels formed by Cx45 or Cx43, respectively, with an expression level suitable for studying macroscopic and single channel GJ channel properties. A cardiac arrhythmia linked Cx45 mutant R184G failed to form functional GJs in DKO HEK293 cells with impaired localizations. These genetically engineered HEK293 cells are well suited for patch clamp study of human GJ channels.
Collapse
Affiliation(s)
- Honghong Chen
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Yi X. Li
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Robert S. Wong
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Jessica L. Esseltine
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada A1B 3V6
| | - Donglin Bai
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada N6A 5C1
| |
Collapse
|
2
|
Mickus R, Raškevičius V, Sarapinienė I, Mikalayeva V, Prekeris R, Skeberdis VA. Phosphorylation-dependent allosteric regulation of Cx43 gap junction inhibitor potency. Biomed Pharmacother 2024; 174:116550. [PMID: 38593702 DOI: 10.1016/j.biopha.2024.116550] [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: 02/09/2024] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024] Open
Abstract
Physiological and pathological processes such as homeostasis, embryogenesis, development, tumorigenesis, and cell movement depend on the intercellular communication through gap junctions (GJIC). Connexin (Cx)-based GJ channels are formed of two apposing hemichannels in the contiguous cells and provide a direct pathway for electrical and metabolic intercellular communication. The main modulators of GJ conductance are transjunctional voltage, intracellular pH, Ca2+, Mg2+, and phosphorylation. Chemical modulators of GJIC are being used in cases of various intercellular communication-dependent diseases. In this study, we used molecular docking, dual whole-cell patch-clamp, and Western blotting to investigate the impact of connexin phosphorylation on GJ chemical gating by α-pinene and other GJ inhibitors (octanol, carbenoxolone, mefloquine, intracellular pH, glycyrrhetinic acid, and sevoflurane) in HeLa cells expressing exogenous Cx43 (full length and truncated at amino acid 258) and other connexins typical of heart and/or nervous system (Cx36, Cx40, Cx45, and Cx47), and in cells expressing endogenous Cx43 (Novikoff and U-87). We found that Ca2+-regulated kinases, such as Ca2+/calmodulin-dependent kinase II, atypical protein kinase C, cyclin-dependent kinase, and Pyk2 kinase may allosterically modulate the potency of α-pinene through phosphorylation of Cx43 C-terminus. The identified new phenomenon was Cx isoform-, inhibitor-, and cell type-dependent. Overall, these results suggest that compounds, the potency of which depends on receptor phosphorylation, might be of particular interest in developing targeted therapies for diseases accompanied by high kinase activity, such as cardiac arrhythmias, epilepsy, stroke, essential tremor, inflammation, and cancer.
Collapse
Affiliation(s)
- Rokas Mickus
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas LT-50162, Lithuania
| | - Vytautas Raškevičius
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas LT-50162, Lithuania
| | - Ieva Sarapinienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas LT-50162, Lithuania
| | - Valeryia Mikalayeva
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas LT-50162, Lithuania
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80015, USA
| | | |
Collapse
|
3
|
Jagielnicki M, Kucharska I, Bennett BC, Harris AL, Yeager M. Connexin Gap Junction Channels and Hemichannels: Insights from High-Resolution Structures. BIOLOGY 2024; 13:298. [PMID: 38785780 PMCID: PMC11117596 DOI: 10.3390/biology13050298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 05/25/2024]
Abstract
Connexins (Cxs) are a family of integral membrane proteins, which function as both hexameric hemichannels (HCs) and dodecameric gap junction channels (GJCs), behaving as conduits for the electrical and molecular communication between cells and between cells and the extracellular environment, respectively. Their proper functioning is crucial for many processes, including development, physiology, and response to disease and trauma. Abnormal GJC and HC communication can lead to numerous pathological states including inflammation, skin diseases, deafness, nervous system disorders, and cardiac arrhythmias. Over the last 15 years, high-resolution X-ray and electron cryomicroscopy (cryoEM) structures for seven Cx isoforms have revealed conservation in the four-helix transmembrane (TM) bundle of each subunit; an αβ fold in the disulfide-bonded extracellular loops and inter-subunit hydrogen bonding across the extracellular gap that mediates end-to-end docking to form a tight seal between hexamers in the GJC. Tissue injury is associated with cellular Ca2+ overload. Surprisingly, the binding of 12 Ca2+ ions in the Cx26 GJC results in a novel electrostatic gating mechanism that blocks cation permeation. In contrast, acidic pH during tissue injury elicits association of the N-terminal (NT) domains that sterically blocks the pore in a "ball-and-chain" fashion. The NT domains under physiologic conditions display multiple conformational states, stabilized by protein-protein and protein-lipid interactions, which may relate to gating mechanisms. The cryoEM maps also revealed putative lipid densities within the pore, intercalated among transmembrane α-helices and between protomers, the functions of which are unknown. For the future, time-resolved cryoEM of isolated Cx channels as well as cryotomography of GJCs and HCs in cells and tissues will yield a deeper insight into the mechanisms for channel regulation. The cytoplasmic loop (CL) and C-terminal (CT) domains are divergent in sequence and length, are likely involved in channel regulation, but are not visualized in the high-resolution X-ray and cryoEM maps presumably due to conformational flexibility. We expect that the integrated use of synergistic physicochemical, spectroscopic, biophysical, and computational methods will reveal conformational dynamics relevant to functional states. We anticipate that such a wealth of results under different pathologic conditions will accelerate drug discovery related to Cx channel modulation.
Collapse
Affiliation(s)
- Maciej Jagielnicki
- The Phillip and Patricia Frost Institute for Chemistry and Molecular Science, Department of Chemistry, University of Miami, 1201 Memorial Drive, Miami, FL 33146, USA; (M.J.); (I.K.)
| | - Iga Kucharska
- The Phillip and Patricia Frost Institute for Chemistry and Molecular Science, Department of Chemistry, University of Miami, 1201 Memorial Drive, Miami, FL 33146, USA; (M.J.); (I.K.)
| | - Brad C. Bennett
- Department of Biological and Environmental Sciences, Howard College of Arts and Sciences, Samford University, Birmingham, AL 35229, USA;
| | - Andrew L. Harris
- Rutgers New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Newark, NJ 07103, USA;
| | - Mark Yeager
- The Phillip and Patricia Frost Institute for Chemistry and Molecular Science, Department of Chemistry, University of Miami, 1201 Memorial Drive, Miami, FL 33146, USA; (M.J.); (I.K.)
- The Phillip and Patricia Frost Institute for Chemistry and Molecular Science, Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL 33146, USA
| |
Collapse
|
4
|
Snipas M, Gudaitis L, Kraujaliene L, Kraujalis T, Verselis VK. Modeling and analysis of voltage gating of gap junction channels at a single-channel level. Biophys J 2023; 122:4176-4193. [PMID: 37766427 PMCID: PMC10645554 DOI: 10.1016/j.bpj.2023.09.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/20/2023] [Accepted: 09/25/2023] [Indexed: 09/29/2023] Open
Abstract
The advancement of single-channel-level recording via the patch-clamp technique has provided a powerful means of assessing the detailed behaviors of various types of ion channels in native and exogenously expressed cellular environments. However, such recordings of gap junction (GJ) channels are hampered by unique challenges that are related to their unusual intercellular configuration and natural clustering into densely packed plaques. Thus, the methods for reliable cross-correlation of data recorded at macroscopic and single-channel levels are lacking in studies of GJs. To address this issue, we combined our previously published four-state model (4SM) of GJ channel gating by voltage with maximum likelihood estimation (MLE)-based analyses of electrophysiological recordings of GJ channel currents. First, we consider evaluation of single-channel characteristics and the methods for efficient stochastic simulation of single GJ channels from the kinetic scheme described by 4SM using data obtained from macroscopic recordings. We then present an MLE-based methodology for extraction of information about transition rates for GJ channels and, ultimately, gating parameters defined in 4SM from recordings with visible unitary events. The validity of the proposed methodology is illustrated using stochastic simulations of single GJ channels and is extended to electrophysiological data recorded in cells expressing connexin 43 tagged with enhanced green fluorescent protein.
Collapse
Affiliation(s)
- Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Department of Mathematical Modelling, Kaunas University of Technology, Kaunas, Lithuania.
| | - Lukas Gudaitis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - 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
| | - Vytas K Verselis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
5
|
Increased Hemichannel Activity Displayed by a Connexin43 Mutation Causing a Familial Connexinopathy Exhibiting Hypotrichosis with Follicular Keratosis and Hyperostosis. Int J Mol Sci 2023; 24:ijms24032222. [PMID: 36768546 PMCID: PMC9916973 DOI: 10.3390/ijms24032222] [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: 12/19/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Mutations in the GJA1 gene that encodes connexin43 (Cx43) cause several rare genetic disorders, including diseases affecting the epidermis. Here, we examined the in vitro functional consequences of a Cx43 mutation, Cx43-G38E, linked to a novel human phenotype of hypotrichosis, follicular keratosis and hyperostosis. We found that Cx43-G38E was efficiently translated in Xenopus oocytes and localized to gap junction plaques in transfected HeLa cells. Cx43-G38E formed functional gap junction channels with the same efficiency as wild-type Cx43 in Xenopus oocytes, although voltage gating of the gap junction channels was altered. Notably, Cx43-G38E significantly increased membrane current flow through the formation of active hemichannels when compared to wild-type Cx43. These data demonstrate the association of increased hemichannel activity to a connexin mutation linked to a skeletal-cutaneous phenotype, suggesting that augmented hemichannel activity could play a role in skin and skeletal disorders caused by human Cx43 mutations.
Collapse
|
6
|
Garré JM, Bukauskas FF, Bennett MVL. Single channel properties of pannexin-1 and connexin-43 hemichannels and P2X7 receptors in astrocytes cultured from rodent spinal cords. Glia 2022; 70:2260-2275. [PMID: 35915989 PMCID: PMC9560969 DOI: 10.1002/glia.24250] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/11/2022]
Abstract
Astrocytes express surface channels involved in purinergic signaling. Among these channels, pannexin-1 (Px1) and connexin-43 (Cx43) hemichannels (HCs) release ATP that acts directly, or through its derivatives, on neurons and glia via purinergic receptors. Although HCs are functional, that is, open and close under physiological and pathological conditions, single channel properties of Px1 HCs in astrocytes have not been defined. Here, we developed a dual voltage clamp technique in HeLa cells expressing human Px1-YFP, and then applied this system to rodent spinal astrocytes to compare their single channel properties with other surface channels, that is, Cx43 HCs and P2X7 receptors (P2X7Rs). Channels were recorded in cell attached patches and evoked with ramp cycles applied through another pipette in whole cell voltage clamp. The mean unitary conductances of Px1 HCs were comparable in HeLa Px1-YFP cells and spinal astrocytes, ~42 and ~48 pS, respectively. Based on their unitary conductance, voltage-dependence, and unitary activity after pharmacological and gene silencing, Px1 HCs in astrocytes could be distinguished from Cx43 HCs and P2X7Rs. Channel activity of Px1 HCs and P2X7Rs was greater than that of Cx43 HCs in control astrocytes during ramps. Unitary activity of Px1 HCs was decreased and that of Cx43 HCs and P2X7Rs increased in astrocytes treated with fibroblast growth factor 1 (FGF-1). In summary, we resolved single channel properties of three different surface channels involved in purinergic signaling in spinal astrocytes, which were differentially modulated by FGF-1, a growth factor involved in neurodevelopment, inflammation and repair.
Collapse
Affiliation(s)
- Juan Mauricio Garré
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Feliksas F Bukauskas
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
7
|
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: 3.0] [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
|
8
|
Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int J Mol Sci 2021; 22:ijms22094413. [PMID: 33922534 PMCID: PMC8122935 DOI: 10.3390/ijms22094413] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Collapse
|
9
|
Snipas M, Kraujalis T, Maciunas K, Kraujaliene L, Gudaitis L, Verselis VK. Four-State Model for Simulating Kinetic and Steady-State Voltage-Dependent Gating of Gap Junctions. Biophys J 2020; 119:1640-1655. [PMID: 32950074 DOI: 10.1016/j.bpj.2020.08.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 07/03/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022] Open
Abstract
Gap junction (GJ) channels, formed of connexin (Cx) proteins, provide a direct pathway for metabolic and electrical cell-to-cell communication. These specialized channels are not just passive conduits for the passage of ions and metabolites but have been shown to gate robustly in response to transjunctional voltage, Vj, the voltage difference between two coupled cells. Voltage gating of GJs could play a physiological role, particularly in excitable cells, which can generate large transients in membrane potential during the propagation of action potentials. We present a mathematical/computational model of GJ channel voltage gating to assess properties of GJ channels that takes into account contingent gating of two series hemichannels and the distribution of Vj across each hemichannel. From electrophysiological recordings in cell cultures expressing Cx43 or Cx45, the principal isoforms expressed in cardiac tissue, various data sets were fitted simultaneously using global optimization. The results showed that the model is capable of describing both steady-state and kinetic properties of homotypic and heterotypic GJ channels composed of these Cxs. Moreover, mathematical analyses showed that the model can be simplified to a reversible two-state system and solved analytically using a rapid equilibrium assumption. Given that excitable cells are arranged in interconnected networks, the equilibrium assumption allows for a substantial reduction in computation time, which is useful in simulations of large clusters of coupled cells. Overall, this model can serve as a tool for the studying of GJ channel gating and its effects on the spread of excitation in networks of electrically coupled cells.
Collapse
Affiliation(s)
- Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Department of Mathematical Modelling, Kaunas University of Technology, Kaunas, Lithuania.
| | - Tadas Kraujalis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Department of Applied Informatics, Kaunas University of Technology, Kaunas, Lithuania
| | - Kestutis Maciunas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Lina Kraujaliene
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Lukas Gudaitis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vytas K Verselis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
10
|
Lissoni A, Wang N, Nezlobinskii T, De Smet M, Panfilov AV, Vandersickel N, Leybaert L, Witschas K. Gap19, a Cx43 Hemichannel Inhibitor, Acts as a Gating Modifier That Decreases Main State Opening While Increasing Substate Gating. Int J Mol Sci 2020; 21:ijms21197340. [PMID: 33027889 PMCID: PMC7583728 DOI: 10.3390/ijms21197340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/12/2020] [Accepted: 09/25/2020] [Indexed: 02/05/2023] Open
Abstract
Cx43 hemichannels (HCs) are electrically and chemically gated transmembrane pores with low open probability and multiple conductance states, which makes kinetic studies of channel gating in large datasets challenging. Here, we developed open access software, named HemiGUI, to analyze HC gating transitions and investigated voltage-induced HC opening based on up to ≈4000 events recorded in HeLa-Cx43-overexpressing cells. We performed a detailed characterization of Cx43 HC gating profiles and specifically focused on the role of the C-terminal tail (CT) domain by recording the impact of adding an EGFP tag to the Cx43 CT end (Cx43-EGFP) or by supplying the Cx43 HC-inhibiting peptide Gap19 that interferes with CT interaction with the cytoplasmic loop (CL). We found that Gap19 not only decreased HC opening activity to the open state (≈217 pS) but also increased the propensity of subconductance (≈80 pS) transitions that additionally became slower as compared to the control. The work demonstrates that large sample transition analysis allows detailed investigations on Cx43 HC gating and shows that Gap19 acts as a HC gating modifier by interacting with the CT that forms a crucial gating element.
Collapse
Affiliation(s)
- Alessio Lissoni
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
| | - Nan Wang
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
| | - Timur Nezlobinskii
- Department of Physics and Astronomy, Ghent University, 9000 Ghent, Belgium; (T.N.); (A.V.P.); (N.V.)
| | - Maarten De Smet
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
| | - Alexander V. Panfilov
- Department of Physics and Astronomy, Ghent University, 9000 Ghent, Belgium; (T.N.); (A.V.P.); (N.V.)
- Laboratory of Computational Biology and Medicine, Ural Federal University, 620075 Ekaterinburg, Russia
| | - Nele Vandersickel
- Department of Physics and Astronomy, Ghent University, 9000 Ghent, Belgium; (T.N.); (A.V.P.); (N.V.)
| | - Luc Leybaert
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
- Correspondence: (L.L.); (K.W.); Tel.: +32-9-332-3366 (L.L.); +32-9-332-6944 (K.W.)
| | - Katja Witschas
- Department of Basic and Applied Medical Sciences—Physiology Group, Ghent University, 9000 Ghent, Belgium; (A.L.); (N.W.); (M.D.S.)
- Correspondence: (L.L.); (K.W.); Tel.: +32-9-332-3366 (L.L.); +32-9-332-6944 (K.W.)
| |
Collapse
|
11
|
Qian S, Tarte E. Finite element modelling of discontinuous action potential propagation in larval zebrafish and human cardiac tissue. Phys Biol 2019; 17:016001. [PMID: 31610528 DOI: 10.1088/1478-3975/ab4d62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recently the larval zebrafish has emerged as a model organism which is used to assist in the studies of human cardiac electrophysiology. Although they share many similar electrophysiological characteristics, it has been found that the conduction velocity (CV) of action potential (AP) propagation in larval zebrafish heart is up to two orders of magnitude smaller than in the adult mammalian heart. To address this difference, we have developed three dimensional discrete models of larval zebrafish ventricular fibres (LZVF) in order to simulate AP propagation, taking into account the cellular nature of the tissues and intercellular conduction via gap junctions. Since our ultimate goal is to simulate a whole larval zebrafish heart, we have used the phenomenological Fitzhugh Nagumo (FHN) equations to describe transmembrane currents, and manually adjusted the FHN parameters, to fit published AP shapes for larval zebrafish ventricular cells. This has the benefit of reduced computational load compared to approaches based on biophysical ion current models. We have created models for 48 and 72 h post fertilisation LZVF tissue using published AP and cell size data for zebrafish embryos and used mammalian values for passive electrical parameters. Using the gap junction resistivity per myocyte as an adjustable parameter, we were able to obtain CVs in both of our LZVF models which agree with experimental observations. In order to validate our approach, we have applied it to a human ventricular fibre (HVF) model similar in structure and parameters to other models of the mammalian heart, but adjusting the FHN parameters to fit published AP shapes for human ventricular cells. We find good agreement with the human models. The gap junction resistivities used in the LZVF models are significantly higher than in the HVF case and are consistent with a lower density of gap junctions connecting cells.
Collapse
Affiliation(s)
- Shuang Qian
- School of Engineering, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
12
|
Jæger KH, Edwards AG, McCulloch A, Tveito A. Properties of cardiac conduction in a cell-based computational model. PLoS Comput Biol 2019; 15:e1007042. [PMID: 31150383 PMCID: PMC6561587 DOI: 10.1371/journal.pcbi.1007042] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 06/12/2019] [Accepted: 04/23/2019] [Indexed: 11/18/2022] Open
Abstract
The conduction of electrical signals through cardiac tissue is essential for maintaining the function of the heart, and conduction abnormalities are known to potentially lead to life-threatening arrhythmias. The properties of cardiac conduction have therefore been the topic of intense study for decades, but a number of questions related to the mechanisms of conduction still remain unresolved. In this paper, we demonstrate how the so-called EMI model may be used to study some of these open questions. In the EMI model, the extracellular space, the cell membrane, the intracellular space and the cell connections are all represented as separate parts of the computational domain, and the model therefore allows for study of local properties that are hard to represent in the classical homogenized bidomain or monodomain models commonly used to study cardiac conduction. We conclude that a non-uniform sodium channel distribution increases the conduction velocity and decreases the time delays over gap junctions of reduced coupling in the EMI model simulations. We also present a theoretical optimal cell length with respect to conduction velocity and consider the possibility of ephaptic coupling (i.e. cell-to-cell coupling through the extracellular potential) acting as an alternative or supporting mechanism to gap junction coupling. We conclude that for a non-uniform distribution of sodium channels and a sufficiently small intercellular distance, ephaptic coupling can influence the dynamics of the sodium channels and potentially provide cell-to-cell coupling when the gap junction connection is absent. The electrochemical wave traversing the heart during every beat is essential for cardiac pumping function and supply of blood to the body. Understanding the stability of this wave is crucial to understanding how lethal arrhythmias are generated. Despite this importance, our knowledge of the physical determinants of wave propagation are still evolving. One particular challenge has been the lack of accurate mathematical models of conduction at the cellular level. Because cardiac muscle is an electrical syncytium, in which direct charge transfer between cells drives wave propagation, classical bidomain and monodomain tissue models employ a homogenized approximation of this process. This approximation is not valid at the length scale of single cells, and prevents any analysis of how cellular structures impact cardiac conduction. Instead, so-called microdomain models must be used for these questions. Here we utilize a recently developed modelling framework that is well suited to represent small collections of cells. By applying this framework, we show that concentration of sodium channels at the longitudinal borders of myocytes accelerates cardiac conduction. We also demonstrate that when juxtaposed cells are sufficiently close, this non-uniform distribution induces large ephaptic currents, which contribute to intercellular coupling.
Collapse
Affiliation(s)
| | | | - Andrew McCulloch
- Department of Bioengineering, University of California, San Diego, California, United States of America
| | - Aslak Tveito
- Simula Research Laboratory, Oslo, Norway
- * E-mail:
| |
Collapse
|
13
|
Santos-Miranda A, Noureldin M, Bai D. Effects of temperature on transjunctional voltage-dependent gating kinetics in Cx45 and Cx40 gap junction channels. J Mol Cell Cardiol 2019; 127:185-193. [PMID: 30594539 DOI: 10.1016/j.yjmcc.2018.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/29/2018] [Accepted: 12/26/2018] [Indexed: 02/01/2023]
Abstract
Gap junctions (GJs) are intercellular channels directly linking neighbouring cells and are dodecamers of connexins. In the human heart, connexin40 (Cx40), Cx43, and Cx45 are expressed in different regions of the heart forming GJs ensuring rapid propagation of action potentials in the myocardium. Two of these connexins, Cx40 and Cx45, formed functional GJs with prominent transjunctional voltage-dependent gating (Vj-gating), which can be a mechanism to down regulate coupling conductance (Gj). It is not clear the effects of temperature on Vj-gating properties. We expressed Cx40 or Cx45 in N2A cells to study the Vj-gating extent, the kinetics of deactivation, and the recovery time course from deactivation at 22 °C, 28 °C, and 32 °C. Dynamic uncoupling between cell pairs were evaluated at different temperatures, junctional delays, and/or repeating frequencies. Cx40 or Cx45 GJs showed little changes in the extent of Vj-gating, but in both cases with a faster deactivation kinetics at high temperatures. The recovery from deactivation was faster at higher temperatures for Cx45 GJs, but not for Cx40 GJs. Cx45 GJs, but not Cx40 GJs, were dynamically uncoupled when sufficient junctional delays and/or repeating frequency in all tested temperatures. Gap junction specific dynamic uncoupling could play an important role in regulating action potential propagation speed in Cx45 enriched nodal cells in the heart.
Collapse
Affiliation(s)
- Artur Santos-Miranda
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Mahmoud Noureldin
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Donglin Bai
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
14
|
Connexin43 mutations linked to skin disease have augmented hemichannel activity. Sci Rep 2019; 9:19. [PMID: 30631135 PMCID: PMC6328547 DOI: 10.1038/s41598-018-37221-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/04/2018] [Indexed: 01/22/2023] Open
Abstract
Mutations in the gene (GJA1) encoding connexin43 (Cx43) are responsible for several rare genetic disorders, including non-syndromic skin-limited diseases. Here we used two different functional expression systems to characterize three Cx43 mutations linked to palmoplantar keratoderma and congenital alopecia-1, erythrokeratodermia variabilis et progressiva, or inflammatory linear verrucous epidermal nevus. In HeLa cells and Xenopus oocytes, we show that Cx43-G8V, Cx43-A44V and Cx43-E227D all formed functional gap junction channels with the same efficiency as wild-type Cx43, with normal voltage gating and a unitary conductance of ~110 pS. In HeLa cells, all three mutations also localized to regions of cell-cell contact and displayed a punctate staining pattern. In addition, we show that Cx43-G8V, Cx43-A44V and Cx43-E227D significantly increase membrane current flow through formation of active hemichannels, a novel activity that was not displayed by wild-type Cx43. The increased membrane current was inhibited by either 2 mM calcium, or 5 µM gadolinium, mediated by hemichannels with a unitary conductance of ~250 pS, and was not due to elevated mutant protein expression. The three Cx43 mutations all showed the same gain of function activity, suggesting that augmented hemichannel activity could play a role in skin-limited diseases caused by human Cx43 mutations.
Collapse
|
15
|
Moreau A, Chahine M. A New Cardiac Channelopathy: From Clinical Phenotypes to Molecular Mechanisms Associated With Na v1.5 Gating Pores. Front Cardiovasc Med 2018; 5:139. [PMID: 30356750 PMCID: PMC6189448 DOI: 10.3389/fcvm.2018.00139] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/19/2018] [Indexed: 12/19/2022] Open
Abstract
Voltage gated sodium channels (NaV) are broadly expressed in the human body. They are responsible for the initiation of action potentials in excitable cells. They also underlie several physiological processes such as cognitive, sensitive, motor, and cardiac functions. The NaV1.5 channel is the main NaV expressed in the heart. A dysfunction of this channel is usually associated with the development of pure electrical disorders such as long QT syndrome, Brugada syndrome, sinus node dysfunction, atrial fibrillation, and cardiac conduction disorders. However, mutations of Nav1.5 have recently been linked to the development of an atypical clinical entity combining complex arrhythmias and dilated cardiomyopathy. Although several Nav1.5 mutations have been linked to dilated cardiomyopathy phenotypes, their pathogenic mechanisms remain to be elucidated. The gating pore may constitute a common biophysical defect for all NaV1.5 mutations located in the channel's VSDs. The creation of such a gating pore may disrupt the ionic homeostasis of cardiomyocytes, affecting electrical signals, cell morphology, and cardiac myocyte function. The main objective of this article is to review the concept of gating pores and their role in structural heart diseases and to discuss potential pharmacological treatments.
Collapse
Affiliation(s)
- Adrien Moreau
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Mohamed Chahine
- CERVO Research Centre, Institut Universitaire en Santé Mentale de Québec, Quebec City, QC, Canada.,Department of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
16
|
A leaky voltage sensor domain of cardiac sodium channels causes arrhythmias associated with dilated cardiomyopathy. Sci Rep 2018; 8:13804. [PMID: 30218094 PMCID: PMC6138662 DOI: 10.1038/s41598-018-31772-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/21/2018] [Indexed: 11/18/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a structural heart disease that causes dilatation of cardiac chambers and impairs cardiac contractility. The SCN5A gene encodes Nav1.5, the predominant cardiac sodium channel alpha subunit. SCN5A mutations have been identified in patients with arrhythmic disorders associated with DCM. The characterization of Nav1.5 mutations located in the voltage sensor domain (VSD) and associated with DCM revealed divergent biophysical defects that do not fully explain the pathologies observed in these patients. The purpose of this study was to characterize the pathological consequences of a gating pore in the heart arising from the Nav1.5/R219H mutation in a patient with complex cardiac arrhythmias and DCM. We report its properties using cardiomyocytes derived from patient-specific human induced pluripotent stem cells. We showed that this mutation generates a proton leak (called gating pore current). We also described disrupted ionic homeostasis, altered cellular morphology, electrical properties, and contractile function, most probably linked to the proton leak. We thus propose a novel link between SCN5A mutation and the complex pathogenesis of cardiac arrhythmias and DCM. Furthermore, we suggest that leaky channels would constitute a common pathological mechanism underlying several neuronal, neuromuscular, and cardiac pathologies.
Collapse
|
17
|
Polyamines preserve connexin 43-mediated gap junctional communication during intracellular hypercalcemia and acidosis. Neuroreport 2018; 28:208-213. [PMID: 28134630 DOI: 10.1097/wnr.0000000000000746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Changes in the regulation, formation, and gating of connexin-based gap junction channels occur in various disorders. It has been shown that H and Ca are involved in the regulation of gap junctional communication. Ischemia-induced intracellular acidification and Ca overload lead to closure of gap junctions and inhibit an exchange by ions and small molecules throughout the network of cells in the heart, brain, and other tissues. In this study, we examined the role of the polyamines in the regulation of connexin 43 (Cx43)-based gap junction channels under elevated intracellular concentrations of hydrogen ([H]i) and calcium ([Ca]i) ions. Experiments, conducted in Novikoff and A172 human glioblastoma cells, which endogenously express Cx43, showed that polyamines prevent downregulation of Cx43-mediated gap junctional communication caused by elevated [Ca]i and [H]i, accompanying ischemic and other pathological conditions. siRNA knockdown of Cx43 significantly reduces gap junctional communication, indicating that Cx43 gap junctions are the targets for spermine regulation.
Collapse
|
18
|
Garciarena CD, Malik A, Swietach P, Moreno AP, Vaughan-Jones RD. Distinct moieties underlie biphasic H + gating of connexin43 channels, producing a pH optimum for intercellular communication. FASEB J 2018; 32:1969-1981. [PMID: 29183963 PMCID: PMC5893178 DOI: 10.1096/fj.201700876r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Most mammalian cells can intercommunicate via connexin-assembled, gap-junctional channels. To regulate signal transmission, connexin (Cx) channel permeability must respond dynamically to physiological and pathophysiological stimuli. One key stimulus is intracellular pH (pHi), which is modulated by a tissue’s metabolic and perfusion status. Our understanding of the molecular mechanism of H+ gating of Cx43 channels—the major isoform in the heart and brain—is incomplete. To interrogate the effects of acidic and alkaline pHi on Cx43 channels, we combined voltage-clamp electrophysiology with pHi imaging and photolytic H+ uncaging, performed over a range of pHi values. We demonstrate that Cx43 channels expressed in HeLa or N2a cell pairs are gated biphasically by pHivia a process that consists of activation by H+ ions at alkaline pHi and inhibition at more acidic pHi. For Cx43 channel–mediated solute/ion transmission, the ensemble of these effects produces a pHi optimum, near resting pHi. By using Cx43 mutants, we demonstrate that alkaline gating involves cysteine residues of the C terminus and is independent of motifs previously implicated in acidic gating. Thus, we present a molecular mechanism by which cytoplasmic acid–base chemistry fine tunes intercellular communication and establishes conditions for the optimal transmission of solutes and signals in tissues, such as the heart and brain.—Garciarena, C. D., Malik, A., Swietach, P., Moreno, A. P., Vaughan-Jones, R. D. Distinct moieties underlie biphasic H+ gating of connexin43 channels, producing a pH optimum for intercellular communication.
Collapse
Affiliation(s)
- Carolina D Garciarena
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, United Kingdom.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Akif Malik
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, United Kingdom
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, United Kingdom
| | - Alonso P Moreno
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - Richard D Vaughan-Jones
- Department of Physiology, Anatomy and Genetics, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Abstract
Voltage-gated sodium channels belong to the superfamily of voltage-gated cation channels. Their structure is based on domains comprising a voltage sensor domain (S1-S4 segments) and a pore domain (S5-S6 segments). Mutations in positively charged residues of the S4 segments may allow protons or cations to pass directly through the gating pore constriction of the voltage sensor domain; these anomalous currents are referred to as gating pore or omega (ω) currents. In the skeletal muscle disorder hypokalemic periodic paralysis, and in arrhythmic dilated cardiomyopathy, inherited mutations of S4 arginine residues promote omega currents that have been shown to be a contributing factor in the pathogenesis of these sodium channel disorders. Characterization of gating pore currents in these channelopathies and with artificial mutations has been possible by measuring the voltage-dependence and selectivity of these leak currents. The basis of gating pore currents and the structural basis of S4 movement through the gating pore has also been studied extensively with molecular dynamics. These simulations have provided valuable insight into the nature of S4 translocation and the physical basis for the effects of mutations that promote permeation of protons or cations through the gating pore.
Collapse
Affiliation(s)
- J R Groome
- Department of Biological Sciences, Idaho State University, Pocatello, ID, 83209, USA.
| | - A Moreau
- Institut NeuroMyogene, ENS de Lyon, Site MONOD, Lyon, France
| | - L Delemotte
- Science for Life Laboratory, Department of Physics, KTH Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| |
Collapse
|
20
|
Weinberg SH. Ephaptic coupling rescues conduction failure in weakly coupled cardiac tissue with voltage-gated gap junctions. CHAOS (WOODBURY, N.Y.) 2017; 27:093908. [PMID: 28964133 DOI: 10.1063/1.4999602] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Electrical conduction in cardiac tissue is usually considered to be primarily facilitated by gap junctions, providing a pathway between the intracellular spaces of neighboring cells. However, recent studies have highlighted the role of coupling via extracellular electric fields, also known as ephaptic coupling, particularly in the setting of reduced gap junction expression. Further, in the setting of reduced gap junctional coupling, voltage-dependent gating of gap junctions, an oft-neglected biophysical property in computational studies, produces a positive feedback that promotes conduction failure. We hypothesized that ephaptic coupling can break the positive feedback loop and rescue conduction failure in weakly coupled cardiac tissue. In a computational tissue model incorporating voltage-gated gap junctions and ephaptic coupling, we demonstrate that ephaptic coupling can rescue conduction failure in weakly coupled tissue. Further, ephaptic coupling increased conduction velocity in weakly coupled tissue, and importantly, reduced the minimum gap junctional coupling necessary for conduction, most prominently at fast pacing rates. Finally, we find that, although neglecting gap junction voltage-gating results in negligible differences in well coupled tissue, more significant differences occur in weakly coupled tissue, greatly underestimating the minimal gap junctional coupling that can maintain conduction. Our study suggests that ephaptic coupling plays a conduction-preserving role, particularly at rapid heart rates.
Collapse
Affiliation(s)
- S H Weinberg
- Virginia Commonwealth University, 401 West Main Street, Richmond, Virginia 23284, USA
| |
Collapse
|
21
|
Snipas M, Rimkute L, Kraujalis T, Maciunas K, Bukauskas FF. Functional asymmetry and plasticity of electrical synapses interconnecting neurons through a 36-state model of gap junction channel gating. PLoS Comput Biol 2017; 13:e1005464. [PMID: 28384220 PMCID: PMC5398722 DOI: 10.1371/journal.pcbi.1005464] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 04/20/2017] [Accepted: 03/09/2017] [Indexed: 11/18/2022] Open
Abstract
We combined the Hodgkin–Huxley equations and a 36-state model of gap junction channel gating to simulate electrical signal transfer through electrical synapses. Differently from most previous studies, our model can account for dynamic modulation of junctional conductance during the spread of electrical signal between coupled neurons. The model of electrical synapse is based on electrical properties of the gap junction channel encompassing two fast and two slow gates triggered by the transjunctional voltage. We quantified the influence of a difference in input resistances of electrically coupled neurons and instantaneous conductance–voltage rectification of gap junctions on an asymmetry of cell-to-cell signaling. We demonstrated that such asymmetry strongly depends on junctional conductance and can lead to the unidirectional transfer of action potentials. The simulation results also revealed that voltage spikes, which develop between neighboring cells during the spread of action potentials, can induce a rapid decay of junctional conductance, thus demonstrating spiking activity-dependent short-term plasticity of electrical synapses. This conclusion was supported by experimental data obtained in HeLa cells transfected with connexin45, which is among connexin isoforms expressed in neurons. Moreover, the model allowed us to replicate the kinetics of junctional conductance under different levels of intracellular concentration of free magnesium ([Mg2+]i), which was experimentally recorded in cells expressing connexin36, a major neuronal connexin. We demonstrated that such [Mg2+]i-dependent long-term plasticity of the electrical synapse can be adequately reproduced through the changes of slow gate parameters of the 36-state model. This suggests that some types of chemical modulation of gap junctions can be executed through the underlying mechanisms of voltage gating. Overall, the developed model accounts for direction-dependent asymmetry, as well as for short- and long-term plasticity of electrical synapses. Our modeling results demonstrate that such complex behavior of the electrical synapse is important in shaping the response of coupled neurons. In most computational models of neuronal networks, it is assumed that electrical synapses have a constant and ohmic conductance. However, numerous experimental studies demonstrate that connexin-based channels expressed in neuronal gap junctions can change their conductance in response to a transjunctional voltage or various chemical reagents. In addition, electrical synapses may exhibit direction-dependent asymmetry of signal transfer. To account for all these phenomena, we combined a 36-state model of gap junction channel gating with Hodgkin–Huxley equations, which describes neuronal excitability. The combined model (HH-36SM) allowed us to evaluate the kinetics of junctional conductance during the spread of electrical signal or in response to chemical factors. Our modeling results, which were based on experimental data, demonstrated that electrical synapses exhibit a complex behavior that can strongly affect the response of coupled neurons. We suggest that the proposed modeling approach is also applicable to describe the behavior of cardiac or other excitable cell networks interconnected through gap junction channels.
Collapse
Affiliation(s)
- Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Mathematical Modeling, Kaunas University of Technology, Kaunas, Lithuania
- * E-mail:
| | - Lina Rimkute
- 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
| | - Kestutis Maciunas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Feliksas F. Bukauskas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York City, New York, United States of America
| |
Collapse
|
22
|
Snipas M, Kraujalis T, Paulauskas N, Maciunas K, Bukauskas FF. Stochastic Model of Gap Junctions Exhibiting Rectification and Multiple Closed States of Slow Gates. Biophys J 2016; 110:1322-33. [PMID: 27028642 DOI: 10.1016/j.bpj.2016.01.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/03/2016] [Accepted: 01/27/2016] [Indexed: 12/26/2022] Open
Abstract
Gap-junction (GJ) channels formed from connexin (Cx) proteins provide direct pathways for electrical and metabolic cell-cell communication. Earlier, we developed a stochastic 16-state model (S16SM) of voltage gating of the GJ channel containing two pairs of fast and slow gates, each operating between open (o) and closed (c) states. However, experimental data suggest that gates may in fact contain two or more closed states. We developed a model in which the slow gate operates according to a linear reaction scheme, o↔c1↔c2, where c1 and c2 are initial-closed and deep-closed states that both close the channel fully, whereas the fast gate operates between the open state and the closed state and exhibits a residual conductance. Thus, we developed a stochastic 36-state model (S36SM) of GJ channel gating that is sensitive to transjunctional voltage (Vj). To accelerate simulation and eliminate noise in simulated junctional conductance (gj) records, we transformed an S36SM into a Markov chain 36-state model (MC36SM) of GJ channel gating. This model provides an explanation for well-established experimental data, such as delayed gj recovery after Vj gating, hysteresis of gj-Vj dependence, and the low ratio of functional channels to the total number of GJ channels clustered in junctional plaques, and it has the potential to describe chemically mediated gating, which cannot be reflected using an S16SM. The MC36SM, when combined with global optimization algorithms, can be used for automated estimation of gating parameters including probabilities of c1↔c2 transitions from experimental gj-time and gj-Vj dependencies.
Collapse
Affiliation(s)
- Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Department of Mathematical Modelling, Kaunas University of Technology, Kaunas, Lithuania
| | - Tadas Kraujalis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Nerijus Paulauskas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Kestutis Maciunas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Feliksas F Bukauskas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York.
| |
Collapse
|
23
|
Connexin26 Mutations Causing Palmoplantar Keratoderma and Deafness Interact with Connexin43, Modifying Gap Junction and Hemichannel Properties. J Invest Dermatol 2016; 136:225-235. [PMID: 26763442 PMCID: PMC4731051 DOI: 10.1038/jid.2015.389] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 09/03/2015] [Accepted: 09/21/2015] [Indexed: 12/23/2022]
Abstract
Mutations in GJB2 (Cx26) cause either deafness, or deafness associated with skin diseases. That different disorders can be caused by distinct mutations within the same gene suggests that unique channel activities are influenced by each class of mutation. We have examined the functional characteristics of two human mutations, Cx26-H73R and Cx26-S183F, causing palmoplantar keratoderma (PPK) and deafness. Both failed to form gap junction channels or hemichannels when expressed alone. Co-expression of the mutants with wild-type Cx43 showed a trans-dominant inhibition of Cx43 gap junction channels, without reductions in Cx43 protein synthesis. In addition, the presence of mutant Cx26 shifted Cx43 channel gating and kinetics towards a more Cx26-like behavior. Co-immunoprecipitation showed Cx43 being pulled down more efficiently with mutant Cx26, than wild-type, confirming the enhanced formation of heteromeric connexons. Finally, the formation of heteromeric connexons resulted in significantly increased Cx43 hemichannel activity in the presence of Cx26 mutants. These findings suggest a common mechanism whereby Cx26 mutations causing PPK and deafness trans-dominantly influence multiple functions of wild-type Cx43. They also implicate a role for aberrant hemichannel activity in the pathogenesis of PPK, and further highlight an emerging role for Cx43 in genetic skin diseases.
Collapse
|
24
|
Abstract
Polyamines (PAs), such as spermine and spermidine, modulate the activity of numerous receptors and channels in the central nervous system (CNS) and are stored in glial cells; however, little attention has been paid to their role in the regulation of connexin (Cx)-based gap junction channels. We have previously shown that PAs facilitate diffusion of Lucifer Yellow through astrocytic gap junctions in acute brain slices; therefore, we hypothesized that spermine can regulate Cx43-mediated (as the most abundant Cx in astrocytes) gap junctional communication. We used electrophysiological patch-clamp recording from paired Novikoff cells endogenously expressing Cx43 and HeLaCx43-EGFP transfectants to study pH-dependent modulation of cell–cell coupling in the presence or absence of PAs. Our results showed (i) a higher increase in gap junctional communication at higher concentrations of cytoplasmic spermine, and (ii) that spermine prevented uncoupling of gap junctions at low intracellular pH. Taken together, we conclude that spermine enhances Cx43-mediated gap junctional communication and may preserve neuronal excitability during ischemia and trauma when pH in the brain acidifies. We, therefore, suggest a new role of spermine in the regulation of a Cx43-based network under (patho)physiological conditions.
Collapse
|
25
|
Moreau A, Gosselin-Badaroudine P, Boutjdir M, Chahine M. Mutations in the Voltage Sensors of Domains I and II of Nav1.5 that are Associated with Arrhythmias and Dilated Cardiomyopathy Generate Gating Pore Currents. Front Pharmacol 2015; 6:301. [PMID: 26733869 PMCID: PMC4689871 DOI: 10.3389/fphar.2015.00301] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/09/2015] [Indexed: 12/19/2022] Open
Abstract
Voltage gated sodium channels (Nav) are transmembrane proteins responsible for action potential initiation. Mutations mainly located in the voltage sensor domain (VSD) of Nav1.5, the cardiac sodium channel, have been associated with the development of arrhythmias combined with dilated cardiomyopathy. Gating pore currents have been observed with three unrelated mutations associated with similar clinical phenotypes. However, gating pores have never been associated with mutations outside the first domain of Nav1.5. The aim of this study was to explore the possibility that gating pore currents might be caused by the Nav1.5 R225P and R814W mutations (R3, S4 in DI and DII, respectively), which are associated with rhythm disturbances and dilated cardiomyopathy. Nav1.5 WT and mutant channels were transiently expressed in tsA201 cells. The biophysical properties of the alpha pore currents and the presence of gating pore currents were investigated using the patch-clamp technique. We confirmed the previously reported gain of function of the alpha pores of the mutant channels, which mainly consisted of increased window currents mostly caused by shifts in the voltage dependence of activation. We also observed gating pore currents associated with the R225P and R814W mutations. This novel permeation pathway was open under depolarized conditions and remained temporarily open at hyperpolarized potentials after depolarization periods. Gating pore currents could represent a molecular basis for the development of uncommon electrical abnormalities and changes in cardiac morphology. We propose that this biophysical defect be routinely evaluated in the case of Nav1.5 mutations on the VSD.
Collapse
Affiliation(s)
- Adrien Moreau
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec City QC, Canada
| | | | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn NY, USA
| | - Mohamed Chahine
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Quebec CityQC, Canada; Department of Medicine, Université Laval, Quebec CityQC, Canada
| |
Collapse
|
26
|
Moreau A, Gosselin-Badaroudine P, Delemotte L, Klein ML, Chahine M. Gating pore currents are defects in common with two Nav1.5 mutations in patients with mixed arrhythmias and dilated cardiomyopathy. ACTA ACUST UNITED AC 2015; 145:93-106. [PMID: 25624448 PMCID: PMC4306709 DOI: 10.1085/jgp.201411304] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nav1.5 channels bearing voltage-sensor domain mutations associated with atypical cardiac arrhythmias and dilated cardiomyopathy generate gating pore currents. The gating pore current, also called omega current, consists of a cation leak through the typically nonconductive voltage-sensor domain (VSD) of voltage-gated ion channels. Although the study of gating pore currents has refined our knowledge of the structure and the function of voltage-gated ion channels, their implication in cardiac disorders has not been established. Two Nav1.5 mutations (R222Q and R225W) located in the VSD are associated with atypical clinical phenotypes involving complex arrhythmias and dilated cardiomyopathy. Using the patch-clamp technique, in silico mutagenesis, and molecular dynamic simulations, we tested the hypothesis that these two mutations may generate gating pore currents, potentially accounting for their clinical phenotypes. Our findings suggest that the gating pore current generated by the R222Q and R225W mutations could constitute the underlying pathological mechanism that links Nav1.5 VSD mutations with human cardiac arrhythmias and dilatation of cardiac chambers.
Collapse
Affiliation(s)
- Adrien Moreau
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec, Québec City, Québec G1J 2G3, Canada
| | - Pascal Gosselin-Badaroudine
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec, Québec City, Québec G1J 2G3, Canada
| | - Lucie Delemotte
- Institute of Computational Molecular Science, Temple University, Philadelphia, PA 19122
| | - Michael L Klein
- Institute of Computational Molecular Science, Temple University, Philadelphia, PA 19122
| | - Mohamed Chahine
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec, Québec City, Québec G1J 2G3, Canada Department of Medicine, Université Laval, Québec City, Québec G1K 7P4, Canada
| |
Collapse
|
27
|
Antanavičiūtė I, Ereminienė E, Vysockas V, Račkauskas M, Skipskis V, Rysevaitė K, Treinys R, Benetis R, Jurevičius J, Skeberdis VA. Exogenous connexin43-expressing autologous skeletal myoblasts ameliorate mechanical function and electrical activity of the rabbit heart after experimental infarction. Int J Exp Pathol 2014; 96:42-53. [PMID: 25529770 DOI: 10.1111/iep.12109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 10/26/2014] [Indexed: 12/19/2022] Open
Abstract
Acute myocardial infarction is one of the major causes of mortality worldwide. For regeneration of the rabbit heart after experimentally induced infarction we used autologous skeletal myoblasts (SMs) due to their high proliferative potential, resistance to ischaemia and absence of immunological and ethical concerns. The cells were characterized with muscle-specific and myogenic markers. Cell transplantation was performed by injection of cell suspension (0.5 ml) containing approximately 6 million myoblasts into the infarction zone. The animals were divided into four groups: (i) no injection; (ii) sham injected; (iii) injected with wild-type SMs; and (iv) injected with SMs expressing connexin43 fused with green fluorescent protein (Cx43EGFP). Left ventricular ejection fraction (LVEF) was evaluated by 2D echocardiography in vivo before infarction, when myocardium has stabilized after infarction, and 3 months after infarction. Electrical activity in the healthy and infarction zones of the heart was examined ex vivo in Langendorff-perfused hearts by optical mapping using di-4-ANEPPS, a potential sensitive fluorescent dye. We demonstrate that SMs in the coculture can couple electrically not only to abutted but also to remote acutely isolated allogenic cardiac myocytes through membranous tunnelling tubes. The beneficial effect of cellular therapy on LVEF and electrical activity was observed in the group of animals injected with Cx43EGFP-expressing SMs. L-type Ca(2+) current amplitude was approximately fivefold smaller in the isolated SMs compared to healthy myocytes suggesting that limited recovery of LVEF may be related to inadequate expression or function of L-type Ca(2+) channels in transplanted differentiating SMs.
Collapse
Affiliation(s)
- Ieva Antanavičiūtė
- Institute of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Li TC, Wang WH, Li C, Yang JJ. Association between mutations in the gap junction β4 gene and nonsyndromic hearing loss: genotype-phenotype correlation patterns. Mol Med Rep 2014; 11:619-24. [PMID: 25333454 DOI: 10.3892/mmr.2014.2725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 09/12/2014] [Indexed: 11/06/2022] Open
Abstract
Numerous studies have confirmed that gap junctions, composed of connexin (Cx) protein, are essential for auditory function. However, few studies have investigated the correlation between variants in the gap junction β4 (GJB4) gene and phenotype in patients with nonsyndromic hearing loss. Our previous study identified 11 patients with GJB4 gene variants in 253 unrelated patients with nonsyndromic hearing loss. In the present study, the phenotype-genotype correlation was examined in the 11 deaf patients with the different variants of GJB4. Analytical results revealed that the majority of probands had congenital hearing loss, which was bilateral, stable and without associated dermatological manifestations or morphological changes of the inner ear. An audiometric profile, including the observed consistency with severe-profound and flat shape dominance, may enable screening for variants of GJB4. On the basis of the above results, it was hypothesized that GJB4 may be a genetic risk factor for the development of nonsyndromic hearing loss and the data from the present study can be used to direct the clinical evaluation and effectively manage the care of families of children with GJB4.
Collapse
Affiliation(s)
- Tung-Cheng Li
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan, R.O.C
| | - Wen-Hung Wang
- Department of Otolaryngology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan, R.O.C
| | - Chuan Li
- Department of Biomedical Sciences, Chung Shan Medical University Hospital, Taichung 402, Taiwan, R.O.C
| | - Jiann-Jou Yang
- Department of Biomedical Sciences, Chung Shan Medical University Hospital, Taichung 402, Taiwan, R.O.C
| |
Collapse
|
29
|
Palacios-Prado N, Chapuis S, Panjkovich A, Fregeac J, Nagy JI, Bukauskas FF. Molecular determinants of magnesium-dependent synaptic plasticity at electrical synapses formed by connexin36. Nat Commun 2014; 5:4667. [PMID: 25135336 PMCID: PMC4142521 DOI: 10.1038/ncomms5667] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/11/2014] [Indexed: 01/28/2023] Open
Abstract
Neuronal gap junction (GJ) channels composed of connexin36 (Cx36) play an important role in neuronal synchronization and network dynamics. Here we show that Cx36-containing electrical synapses between inhibitory neurons of the thalamic reticular nucleus are bidirectionally modulated by changes in intracellular free magnesium concentration ([Mg(2+)]i). Chimeragenesis demonstrates that the first extracellular loop of Cx36 contains a Mg(2+)-sensitive domain, and site-directed mutagenesis shows that the pore-lining residue D47 is critical in determining high Mg(2+)-sensitivity. Single-channel analysis of Mg(2+)-sensitive chimeras and mutants reveals that [Mg(2+)]i controls the strength of electrical coupling mostly via gating mechanisms. In addition, asymmetric transjunctional [Mg(2+)]i induces strong instantaneous rectification, providing a novel mechanism for electrical rectification in homotypic Cx36 GJs. We suggest that Mg(2+)-dependent synaptic plasticity of Cx36-containing electrical synapses could underlie neuronal circuit reconfiguration via changes in brain energy metabolism that affects neuronal levels of intracellular ATP and [Mg(2+)]i.
Collapse
Affiliation(s)
- Nicolás Palacios-Prado
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, NY 10461, USA
- Grass Laboratory, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Sandrine Chapuis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, NY 10461, USA
| | - Alejandro Panjkovich
- European Molecular Biology Laboratory, Hamburg Outstation, 22603 Hamburg, Germany
| | - Julien Fregeac
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, NY 10461, USA
| | - James I. Nagy
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Feliksas F. Bukauskas
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, NY 10461, USA
| |
Collapse
|
30
|
Pannexins form gap junctions with electrophysiological and pharmacological properties distinct from connexins. Sci Rep 2014; 4:4955. [PMID: 24828343 PMCID: PMC4021813 DOI: 10.1038/srep04955] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 04/22/2014] [Indexed: 11/17/2022] Open
Abstract
Stable expression of pannexin 1 (Panx1) and pannexin 3 (Panx3) resulted in functional gap junctions (GJs) in HeLa cells, but not in Neuro-2a (N2a) or PC-12 cells. The glycosylation pattern of expressed Panx1 varied greatly among different cell lines. In contrast to connexin (Cx) containing GJs (Cx-GJs), junctional conductance (Gj) of pannexin GJs (Panx-GJs) is very less sensitive to junctional voltage. Both Panx1 and Panx3 junctions favoured anionic dyes over cations to permeate. Though, carbenoxolone (CBX) and probenecid blocked Panx1 hemichannel activity, they had no effect on Panx1-GJs or Panx3-GJs. Extracellular loop 1 (E1) of Panx1 possibly bears the binding pocket. The Cx-GJ blocker heptanol blocked neither Panx1 hemichannel nor Panx-GJs. Unlike the GJs formed by most Cxs, CO2 did not uncouple Panx-GJs completely. Oxygen and glucose deprivation (OGD) caused lesser uncoupling of Panx-GJs compared to Cx43-GJs. These findings demonstrate properties of Panx-GJs that are distinctly different from Cx-GJs.
Collapse
|
31
|
Moreau A, Gosselin-Badaroudine P, Chahine M. Biophysics, pathophysiology, and pharmacology of ion channel gating pores. Front Pharmacol 2014; 5:53. [PMID: 24772081 PMCID: PMC3982104 DOI: 10.3389/fphar.2014.00053] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 03/12/2014] [Indexed: 12/19/2022] Open
Abstract
Voltage sensor domains (VSDs) are a feature of voltage gated ion channels (VGICs) and voltage sensitive proteins. They are composed of four transmembrane (TM) segments (S1–S4). Currents leaking through VSDs are called omega or gating pore currents. Gating pores are caused by mutations of the highly conserved positively charged amino acids in the S4 segment that disrupt interactions between the S4 segment and the gating charge transfer center (GCTC). The GCTC separates the intracellular and extracellular water crevices. The disruption of S4–GCTC interactions allows these crevices to communicate and create a fast activating and non-inactivating alternative cation-selective permeation pathway of low conductance, or a gating pore. Gating pore currents have recently been shown to cause periodic paralysis phenotypes. There is also increasing evidence that gating pores are linked to several other familial diseases. For example, gating pores in Nav1.5 and Kv7.2 channels may underlie mixed arrhythmias associated with dilated cardiomyopathy (DCM) phenotypes and peripheral nerve hyperexcitability (PNH), respectively. There is little evidence for the existence of gating pore blockers. Moreover, it is known that a number of toxins bind to the VSD of a specific domain of Na+ channels. These toxins may thus modulate gating pore currents. This focus on the VSD motif opens up a new area of research centered on developing molecules to treat a number of cell excitability disorders such as epilepsy, cardiac arrhythmias, and pain. The purpose of the present review is to summarize existing knowledge of the pathophysiology, biophysics, and pharmacology of gating pore currents and to serve as a guide for future studies aimed at improving our understanding of gating pores and their pathophysiological roles.
Collapse
Affiliation(s)
- Adrien Moreau
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec Quebec City, QC, Canada
| | | | - Mohamed Chahine
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec Quebec City, QC, Canada ; Department of Medicine, Université Laval Quebec City, QC, Canada
| |
Collapse
|
32
|
Harris AL, Contreras JE. Motifs in the permeation pathway of connexin channels mediate voltage and Ca (2+) sensing. Front Physiol 2014; 5:113. [PMID: 24744733 PMCID: PMC3978323 DOI: 10.3389/fphys.2014.00113] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/06/2014] [Indexed: 11/13/2022] Open
Abstract
Connexin channels mediate electrical coupling, intercellular molecular signaling, and extracellular release of signaling molecules. Connexin proteins assemble intracellularly as hexamers to form plasma membrane hemichannels. The docking of two hemichannels in apposed cells forms a gap junction channel that allows direct electrical and selective cytoplasmic communication between adjacent cells. Hemichannels and junctional channels are gated by voltage, but extracellular Ca (2+) also gates unpaired plasma membrane hemichannels. Unlike other ion channels, connexin channels do not contain discrete voltage- or Ca (2+)-sensing modules linked to a separate pore-forming module. All studies to date indicate that voltage and Ca (2+) sensing are predominantly mediated by motifs that lie within or are exposed to the pore lumen. The sensors appear to be integral components of the gates, imposing an obligatory structural linkage between sensing and gating not commonly present in other ion channels, in which the sensors are semi-independent domains distinct from the pore. Because of this, the structural and electrostatic features that define connexin channel gating also define pore permeability properties, and vice versa; analysis/mutagenesis of gating and of permeability properties are linked. This offers unique challenges and opportunities for elucidating mechanisms of ligand and voltage-driven gating.
Collapse
Affiliation(s)
- Andrew L Harris
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers University Newark, NJ, USA
| | - Jorge E Contreras
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers University Newark, NJ, USA
| |
Collapse
|
33
|
Nielsen MS, Axelsen LN, Sorgen PL, Verma V, Delmar M, Holstein-Rathlou NH. Gap junctions. Compr Physiol 2013; 2:1981-2035. [PMID: 23723031 DOI: 10.1002/cphy.c110051] [Citation(s) in RCA: 301] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gap junctions are essential to the function of multicellular animals, which require a high degree of coordination between cells. In vertebrates, gap junctions comprise connexins and currently 21 connexins are known in humans. The functions of gap junctions are highly diverse and include exchange of metabolites and electrical signals between cells, as well as functions, which are apparently unrelated to intercellular communication. Given the diversity of gap junction physiology, regulation of gap junction activity is complex. The structure of the various connexins is known to some extent; and structural rearrangements and intramolecular interactions are important for regulation of channel function. Intercellular coupling is further regulated by the number and activity of channels present in gap junctional plaques. The number of connexins in cell-cell channels is regulated by controlling transcription, translation, trafficking, and degradation; and all of these processes are under strict control. Once in the membrane, channel activity is determined by the conductive properties of the connexin involved, which can be regulated by voltage and chemical gating, as well as a large number of posttranslational modifications. The aim of the present article is to review our current knowledge on the structure, regulation, function, and pharmacology of gap junctions. This will be supported by examples of how different connexins and their regulation act in concert to achieve appropriate physiological control, and how disturbances of connexin function can lead to disease.
Collapse
Affiliation(s)
- Morten Schak Nielsen
- Department of Biomedical Sciences and The Danish National Research Foundation Centre for Cardiac Arrhythmia, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
34
|
Fast structural responses of gap junction membrane domains to AB5 toxins. Proc Natl Acad Sci U S A 2013; 110:E4125-33. [PMID: 24133139 DOI: 10.1073/pnas.1315850110] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Gap junctions (GJs) represent connexin-rich membrane domains that connect interiors of adjoining cells in mammalian tissues. How fast GJs can respond to bacterial pathogens has not been known previously. Using Bessel beam plane illumination and confocal spinning disk microscopy, we found fast (~500 ms) formation of connexin-depleted regions (CDRs) inside GJ plaques between cells exposed to AB5 toxins. CDR formation appears as a fast redistribution of connexin channels within GJ plaques with minor changes in outline or geometry. CDR formation does not depend on membrane trafficking or submembrane cytoskeleton and has no effect on GJ conductance. However, CDR responses depend on membrane lipids, can be modified by cholesterol-clustering agents and extracellular K(+) ion concentration, and influence cAMP signaling. The CDR response of GJ plaques to bacterial toxins is a phenomenon observed for all tested connexin isoforms. Through signaling, the CDR response may enable cells to sense exposure to AB5 toxins. CDR formation may reflect lipid-phase separation events in the biological membrane of the GJ plaque, leading to increased connexin packing and lipid reorganization. Our data demonstrate very fast dynamics (in the millisecond-to-second range) within GJ plaques, which previously were considered to be relatively stable, long-lived structures.
Collapse
|
35
|
Ambrosi C, Walker AE, DePriest AD, Cone AC, Lu C, Badger J, Skerrett IM, Sosinsky GE. Analysis of trafficking, stability and function of human connexin 26 gap junction channels with deafness-causing mutations in the fourth transmembrane helix. PLoS One 2013; 8:e70916. [PMID: 23967136 PMCID: PMC3744544 DOI: 10.1371/journal.pone.0070916] [Citation(s) in RCA: 22] [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: 03/27/2013] [Accepted: 06/25/2013] [Indexed: 01/24/2023] Open
Abstract
Human Connexin26 gene mutations cause hearing loss. These hereditary mutations are the leading cause of childhood deafness worldwide. Mutations in gap junction proteins (connexins) can impair intercellular communication by eliminating protein synthesis, mis-trafficking, or inducing channels that fail to dock or have aberrant function. We previously identified a new class of mutants that form non-functional gap junction channels and hemichannels (connexons) by disrupting packing and inter-helix interactions. Here we analyzed fourteen point mutations in the fourth transmembrane helix of connexin26 (Cx26) that cause non-syndromic hearing loss. Eight mutations caused mis-trafficking (K188R, F191L, V198M, S199F, G200R, I203K, L205P, T208P). Of the remaining six that formed gap junctions in mammalian cells, M195T and A197S formed stable hemichannels after isolation with a baculovirus/Sf9 protein purification system, while C202F, I203T, L205V and N206S formed hemichannels with varying degrees of instability. The function of all six gap junction-forming mutants was further assessed through measurement of dye coupling in mammalian cells and junctional conductance in paired Xenopus oocytes. Dye coupling between cell pairs was reduced by varying degrees for all six mutants. In homotypic oocyte pairings, only A197S induced measurable conductance. In heterotypic pairings with wild-type Cx26, five of the six mutants formed functional gap junction channels, albeit with reduced efficiency. None of the mutants displayed significant alterations in sensitivity to transjunctional voltage or induced conductive hemichannels in single oocytes. Intra-hemichannel interactions between mutant and wild-type proteins were assessed in rescue experiments using baculovirus expression in Sf9 insect cells. Of the four unstable mutations (C202F, I203T, L205V, N206S) only C202F and N206S formed stable hemichannels when co-expressed with wild-type Cx26. Stable M195T hemichannels displayed an increased tendency to aggregate. Thus, mutations in TM4 cause a range of phenotypes of dysfunctional gap junction channels that are discussed within the context of the X-ray crystallographic structure.
Collapse
Affiliation(s)
- Cinzia Ambrosi
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - Amy E. Walker
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - Adam D. DePriest
- Biology Department, State University of New York Buffalo State, Buffalo, New York, United States of America
| | - Angela C. Cone
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - Connie Lu
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
| | - John Badger
- DeltaG Technologies, San Diego, California, United States of America
| | - I. Martha Skerrett
- Biology Department, State University of New York Buffalo State, Buffalo, New York, United States of America
| | - Gina E. Sosinsky
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California San Diego, La Jolla, California, United States of America
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
36
|
Huang T, Shao Q, MacDonald A, Xin L, Lorentz R, Bai D, Laird DW. Autosomal recessive GJA1 (Cx43) gene mutations cause oculodentodigital dysplasia by distinct mechanisms. J Cell Sci 2013; 126:2857-66. [PMID: 23606748 DOI: 10.1242/jcs.123315] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oculodentodigital dysplasia (ODDD) is mainly an autosomal dominant human disease caused by mutations in the GJA1 gene, which encodes the gap junction protein connexin43 (Cx43). Surprisingly, there have been two autosomal recessive mutations reported that cause ODDD: a single amino acid substitution (R76H) and a premature truncation mutation (R33X). When expressed in either gap junctional intercellular communication (GJIC)-deficient HeLa cells or Cx43-expressing NRK cells, the R76H mutant trafficked to the plasma membrane to form gap junction-like plaques, whereas the R33X mutant remained diffusely localized throughout the cell, including the nucleus. As expected, the R33X mutant failed to form functional channels. In the case of the R76H mutant, dye transfer studies in HeLa cells and electrical conductance analysis in GJIC-deficient N2a cells revealed that this mutant could form functional gap junction channels, albeit with reduced macroscopic and single channel conductance. Alexa 350 dye transfer studies further revealed that the R76H mutant had no detectable negative effect on the function of co-expressed Cx26, Cx32, Cx37 or Cx40, whereas the R33X mutant exhibited significant dominant or trans-dominant effects on Cx43 and Cx40 as manifested by a reduction in wild-type connexin gap junction plaques. Taken together, our results suggest that the trans-dominant effect of R33X together with its complete inability to form a functional channel may explain why patients harboring this autosomal recessive R33X mutant exhibit greater disease burden than patients harboring the R76H mutant.
Collapse
Affiliation(s)
- Tao Huang
- Department of Anatomy and Cell Biology, University of Western Ontario, London ON N6A-5C1, Canada
| | | | | | | | | | | | | |
Collapse
|
37
|
Marandykina A, Palacios-Prado N, Rimkutė L, Skeberdis VA, Bukauskas FF. Regulation of connexin36 gap junction channels by n-alkanols and arachidonic acid. J Physiol 2013; 591:2087-101. [PMID: 23420660 PMCID: PMC3634521 DOI: 10.1113/jphysiol.2013.250910] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 02/13/2013] [Indexed: 12/18/2022] Open
Abstract
We examined junctional conductance (gj) and its dependence on transjunctional voltage in gap junction (GJ) channels formed of wild-type connexin36 (Cx36) or its fusion form with green fluorescent protein (Cx36-EGFP) transfected in HeLa cells or endogenously expressed in primary culture of pancreatic β-cells. Only a very small fraction (∼0.8%) of Cx36-EGFP channels assembled into junctional plaques of GJs were open under control conditions. We found that short carbon chain n-alkanols (SCCAs) increased gj, while long carbon chain n-alkanols resulted in full uncoupling; cutoff is between heptanol and octanol. The fraction of functional channels and gj increased several fold under an exposure to SCCAs, or during reduction of endogenous levels of arachidonic acid (AA) by exposure to fatty acid-free BSA or cytosolic phospholipase A2 inhibitors. Moreover, uncoupling caused by exogenously applied AA can be rescued by BSA, which binds AA and other polyunsaturated fatty acids (PUFAs), but not by BSA modified with 1,2-cyclohexanedione, which does not bind AA and other PUFAs. We propose that under control conditions, Cx36 GJ channels in HeLa transfectants and β-cells are inhibited by endogenous AA, which stabilizes a closed conformational state of the channel that leads to extremely low fraction of functional channels. In addition, SCCAs increase gj by interfering with endogenous AA-dependent inhibition, increasing open probability and the fraction of functional channels.
Collapse
Affiliation(s)
- Alina Marandykina
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
38
|
Sahu G, Bera AK. Contribution of intracellular calcium and pH in ischemic uncoupling of cardiac gap junction channels formed of connexins 43, 40, and 45: a critical function of C-terminal domain. PLoS One 2013; 8:e60506. [PMID: 23536911 PMCID: PMC3607587 DOI: 10.1371/journal.pone.0060506] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/26/2013] [Indexed: 11/19/2022] Open
Abstract
Ischemia is known to inhibit gap junction (GJ) mediated intercellular communication. However the detail mechanisms of this inhibition are largely unknown. In the present study, we determined the vulnerability of different cardiac GJ channels formed of connexins (Cxs) 43, 40, and 45 to simulated ischemia, by creating oxygen glucose deprived (OGD) condition. 5 minutes of OGD decreased the junctional conductance (Gj) of Cx43, Cx40 and Cx45 by 53±3%, 64±1% and 85±2% respectively. Reduction of Gj was prevented completely by restricting the change of both intracellular calcium ([Ca(2+)]i) and pH (pHi) with potassium phosphate buffer. Clamping of either [Ca(2+)]i or pHi, through BAPTA (2 mM) or HEPES (80 mM) respectively, offered partial resistance to ischemic uncoupling. Anti-calmodulin antibody attenuated the uncoupling of Cx43 and Cx45 significantly but not of Cx40. Furthermore, OGD could reduce only 26±2% of Gj in C-terminus (CT) truncated Cx43 (Cx43-Δ257). Tethering CT of Cx43 to the CT-truncated Cx40 (Cx40-Δ249), and Cx45 (Cx45-Δ272) helped to resist OGD mediated uncoupling. Moreover, CT domain played a significant role in determining the junction current density and plaque diameter. Our results suggest; OGD mediated uncoupling of GJ channels is primarily due to elevated [Ca(2+)]i and acidic pHi, though the latter contributes more. Among Cx43, Cx40 and Cx45, Cx43 is the most resistant to OGD while Cx45 is the most sensitive one. CT of Cx43 has major necessary elements for OGD induced uncoupling and it can complement CT of Cx40 and Cx45.
Collapse
Affiliation(s)
- Giriraj Sahu
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Amal Kanti Bera
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- * E-mail:
| |
Collapse
|
39
|
Abrams CK, Islam M, Mahmoud R, Kwon T, Bargiello TA, Freidin MM. Functional requirement for a highly conserved charged residue at position 75 in the gap junction protein connexin 32. J Biol Chem 2013; 288:3609-19. [PMID: 23209285 PMCID: PMC3561579 DOI: 10.1074/jbc.m112.392670] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 11/28/2012] [Indexed: 01/10/2023] Open
Abstract
Charcot Marie Tooth disease (CMT) is a group of inherited disorders characterized clinically by exclusively or predominantly peripheral nerve dysfunction. CMT1X, the most common form of X-linked CMT is caused by mutations in connexin 32 (Cx32). In this work, we used dual whole cell patch clamp recording to examine the functional effects of mutations at the Arg(75) position. This residue is highly conserved among members of the connexin family, and disease-causing mutations have been identified at this (or the corresponding) position in Cx26, Cx43, and Cx46. Thus, a better understanding of the effects of mutations of this position in Cx32 may have relevance to pathogenesis of a number of different human diseases. All three mutants associated with CMT1X (R75P, R75Q, and R75W) showed very low levels of coupling similar to those of the cells transfected with vector alone. Heterotypic pairing with Cx32 WT showed that the absence of coupling for these mutants in the homotypic configuration could be explained by shifts in their hemichannel G(j)-V(j) relations. Examination of the expression levels and gating characteristics of seven additional mutants (R75A, R75D, R75E, R75H, R75K, R75L, and R75V) at this position suggest that the positive charge at position 75 in Cx32 is required for normal channel function but not for gap junction assembly. Our studies also suggest that disease treatment strategies for CMT1X, which correct trafficking abnormalities in Cx32, may be ineffective for the group of mutations also conferring changes in gating properties of Cx32 channels.
Collapse
Affiliation(s)
- Charles K Abrams
- Department of Neurology, State University of New York, Downstate Medical Center, Brooklyn, New York 11203, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Electrophysiology of single and aggregate Cx43 hemichannels. PLoS One 2012; 7:e47775. [PMID: 23112846 PMCID: PMC3480394 DOI: 10.1371/journal.pone.0047775] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 09/17/2012] [Indexed: 11/20/2022] Open
Abstract
Connexin43 (Cx43) is the most ubiquitous gap junction protein in the human body and is essential for cell-to-cell communication in a variety of organs and organ systems. As a result, Cx43 is responsible for mediating both electrical and chemical signals, passing dissolved solutes and small signaling molecules between cells in a coordinated fashion. Here, we explore the electrophysiological properties of hemichannels formed from Cx43 and Cx43 fused to eGFP (Cx43eGFP) and their interactions in a planar lipid membrane (BLM). Unlike in vivo patch clamp experiments, Cx43 was purified and isolated from other membrane constituents allowing elucidation of individual protein responses to various electrical and chemical stimuli. Using this system, we examined hemichannel electrophysiology and the roles of several well-known gap junction blockers, namely: lanthanum, heptanol, carbenoxalone and lindane. We also observed a critical number of hemichannels required for an accelerated conductance increase, an emergent electrical signature indicative of plaque formation.
Collapse
|
41
|
Bond SR, Wang N, Leybaert L, Naus CC. Pannexin 1 Ohnologs in the Teleost Lineage. J Membr Biol 2012; 245:483-93. [DOI: 10.1007/s00232-012-9497-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 07/31/2012] [Indexed: 02/04/2023]
|
42
|
Wan CK, Shaikh SB, Green CR, Nicholson LFB. Comparison of bidirectional and bicistronic inducible systems for coexpression of connexin genes and fluorescent reporters. Anal Biochem 2012; 431:90-5. [PMID: 22929700 DOI: 10.1016/j.ab.2012.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 08/17/2012] [Accepted: 08/18/2012] [Indexed: 11/18/2022]
Abstract
Gene expression studies often require inducible coexpression of both a gene of interest and a reporter gene. Fusion of fluorescent reporters can, however, modify protein structure and function. We have generated inducible expression systems for two connexin genes: Cx30 and Cx43. It has been reported recently that reporter fusion to connexins can modify their function. Therefore, we compared two methods of independent reporter coexpression and examined colocalization with induced connexin expression. Identical levels of connexin expression were observed for both the bidirectional and bicistronic expression systems. In contrast, however, reporter gene expression by the bidirectional promoter provided brighter average fluorescent pixel intensity than expression of a reporter gene in a bicistronic transcript. Moreover, as a result of this difference in reporter expression, bidirectional expression systems provided equal or better colocalization between the connexins and reporter gene fluorescence. The results of our study indicate that bidirectional reporter expression provides a robust indicator of transfection and gene expression and, therefore, may favor the use of bidirectional over bicistronic reporters in the design of expression systems where the gene of interest, such as a connexin gene, contains translational motifs or long intronic regions.
Collapse
Affiliation(s)
- Carthur K Wan
- Department of Anatomy with Radiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 92-019, New Zealand.
| | | | | | | |
Collapse
|
43
|
Kronengold J, Srinivas M, Verselis VK. The N-terminal half of the connexin protein contains the core elements of the pore and voltage gates. J Membr Biol 2012; 245:453-63. [PMID: 22825713 PMCID: PMC3735448 DOI: 10.1007/s00232-012-9457-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 06/20/2012] [Indexed: 11/21/2022]
Abstract
Connexins form channels with large aqueous pores that mediate fluxes of inorganic ions and biological signaling molecules. Studies aimed at identifying the connexin pore now include a crystal structure that provides details of putative pore-lining residues that need to be verified using independent biophysical approaches. Here we extended our initial cysteine-scanning studies of the TM1/E1 region of Cx46 hemichannels to include TM2 and TM3 transmembrane segments. No evidence of reactivity was observed in either TM2 or TM3 probed with small or large thiol-modifying reagents. Several identified pore residues in E1 of Cx46 have been verified in different Cx isoforms. Use of variety of thiol reagents indicates that the connexin hemichannel pore is large and flexible enough, at least in the extracellular part of the pore funnel, to accommodate uncommonly large side chains. We also find that that gating characteristics are largely determined by the same domains that constitute the pore. These data indicate that biophysical and structural studies are converging towards a view that the N-terminal half of the Cx protein contains the principal components of the pore and gating elements, with NT, TM1 and E1 forming the pore funnel.
Collapse
Affiliation(s)
- Jack Kronengold
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | |
Collapse
|
44
|
Paulauskas N, Pranevicius H, Mockus J, Bukauskas FF. Stochastic 16-state model of voltage gating of gap-junction channels enclosing fast and slow gates. Biophys J 2012; 102:2471-80. [PMID: 22713562 DOI: 10.1016/j.bpj.2012.04.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/23/2012] [Accepted: 04/25/2012] [Indexed: 11/25/2022] Open
Abstract
Gap-junction (GJ) channels formed of connexin (Cx) proteins provide a direct pathway for electrical and metabolic cell-cell interaction. Each hemichannel in the GJ channel contains fast and slow gates that are sensitive to transjunctional voltage (Vj). We developed a stochastic 16-state model (S16SM) that details the operation of two fast and two slow gates in series to describe the gating properties of homotypic and heterotypic GJ channels. The operation of each gate depends on the fraction of Vj that falls across the gate (VG), which varies depending on the states of three other gates in series, as well as on parameters of the fast and slow gates characterizing 1), the steepness of each gate's open probability on VG; 2), the voltage at which the open probability of each gate equals 0.5; 3), the gating polarity; and 4), the unitary conductances of the gates and their rectification depending on VG. S16SM allows for the simulation of junctional current dynamics and the dependence of steady-state junctional conductance (gj,ss) on Vj. We combined global coordinate optimization algorithms with S16SM to evaluate the gating parameters of fast and slow gates from experimentally measured gj,ss-Vj dependencies in cells expressing different Cx isoforms and forming homotypic and/or heterotypic GJ channels.
Collapse
Affiliation(s)
- Nerijus Paulauskas
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, USA
| | | | | | | |
Collapse
|
45
|
Gosselin-Badaroudine P, Keller DI, Huang H, Pouliot V, Chatelier A, Osswald S, Brink M, Chahine M. A proton leak current through the cardiac sodium channel is linked to mixed arrhythmia and the dilated cardiomyopathy phenotype. PLoS One 2012; 7:e38331. [PMID: 22675453 PMCID: PMC3365008 DOI: 10.1371/journal.pone.0038331] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 05/03/2012] [Indexed: 12/19/2022] Open
Abstract
Cardiac Na+ channels encoded by the SCN5A gene are essential for initiating heart beats and maintaining a regular heart rhythm. Mutations in these channels have recently been associated with atrial fibrillation, ventricular arrhythmias, conduction disorders, and dilated cardiomyopathy (DCM). We investigated a young male patient with a mixed phenotype composed of documented conduction disorder, atrial flutter, and ventricular tachycardia associated with DCM. Further family screening revealed DCM in the patient's mother and sister and in three of the mother's sisters. Because of the complex clinical phenotypes, we screened SCN5A and identified a novel mutation, R219H, which is located on a highly conserved region on the fourth helix of the voltage sensor domain of Nav1.5. Three family members with DCM carried the R219H mutation. The wild-type (WT) and mutant Na+ channels were expressed in a heterologous expression system, and intracellular pH (pHi) was measured using a pH-sensitive electrode. The biophysical characterization of the mutant channel revealed an unexpected selective proton leak with no effect on its biophysical properties. The H+ leak through the mutated Nav1.5 channel was not related to the Na+ permeation pathway but occurred through an alternative pore, most probably a proton wire on the voltage sensor domain. We propose that acidification of cardiac myocytes and/or downstream events may cause the DCM phenotype and other electrical problems in affected family members. The identification of this clinically significant H+ leak may lead to the development of more targeted treatments.
Collapse
Affiliation(s)
| | - Dagmar I. Keller
- Cardiology Department, University Hospital Zurich, Zurich, Switzerland
- Cardiology Department, University Hospital Basel, Basel, Switzerland
- Cardiobiology Research Laboratories, University Hospital Basel, Basel, Switzerland
| | - Hai Huang
- Laval University Robert-Giffard Research Centre, Quebec City, Quebec, Canada
| | - Valérie Pouliot
- Laval University Robert-Giffard Research Centre, Quebec City, Quebec, Canada
| | - Aurélien Chatelier
- Laval University Robert-Giffard Research Centre, Quebec City, Quebec, Canada
| | - Stefan Osswald
- Cardiology Department, University Hospital Basel, Basel, Switzerland
| | - Marijke Brink
- Cardiobiology Research Laboratories, University Hospital Basel, Basel, Switzerland
| | - Mohamed Chahine
- Laval University Robert-Giffard Research Centre, Quebec City, Quebec, Canada
- Department of Medicine, Laval University, Quebec City, Quebec, Canada
- * E-mail:
| |
Collapse
|
46
|
Ek-Vitorin JF, Burt JM. Structural basis for the selective permeability of channels made of communicating junction proteins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:51-68. [PMID: 22342665 DOI: 10.1016/j.bbamem.2012.02.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 01/24/2012] [Accepted: 02/01/2012] [Indexed: 01/08/2023]
Abstract
The open state(s) of gap junction channels is evident from their permeation by small ions in response to an applied intercellular (transjunctional/transchannel) voltage gradient. That an open channel allows variable amounts of current to transit from cell-to-cell in the face of a constant intercellular voltage difference indicates channel open/closing can be complete or partial. The physiological significance of such open state options is, arguably, the main concern of junctional regulation. Because gap junctions are permeable to many substances, it is sensible to inquire whether and how each open state influences the intercellular diffusion of molecules as valuable as, but less readily detected than current-carrying ions. Presumably, structural changes perceived as shifts in channel conductivity would significantly alter the transjunctional diffusion of molecules whose limiting diameter approximates the pore's limiting diameter. Moreover, changes in junctional permeability to some molecules might occur without evident changes in conductivity, either at macroscopic or single channel level. Open gap junction channels allow the exchange of cytoplasmic permeants between contacting cells by simple diffusion. The identity of such permeants, and the functional circumstances and consequences of their junctional exchange presently constitute the most urgent (and demanding) themes of the field. Here, we consider the necessity for regulating this exchange, the possible mechanism(s) and structural elements likely involved in such regulation, and how regulatory phenomena could be perceived as changes in chemical vs. electrical coupling; an overall reflection on our collective knowledge of junctional communication is then applied to suggest new avenues of research. This article is part of a Special Issue entitled: The Communicating junctions, roles and dysfunctions.
Collapse
|
47
|
Glycogen synthase kinase 3β transfers cytoprotective signaling through connexin 43 onto mitochondrial ATP-sensitive K+ channels. Proc Natl Acad Sci U S A 2012; 109:E242-51. [PMID: 22238425 DOI: 10.1073/pnas.1107479109] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Despite compelling evidence supporting key roles for glycogen synthase kinase 3β (GSK3β), mitochondrial adenosine triphosphate-sensitive K(+) (mitoK(ATP)) channels, and mitochondrial connexin 43 (Cx43) in cytoprotection, it is not clear how these signaling modules are linked mechanistically. By patch-clamping the inner membrane of murine cardiac mitochondria, we found that inhibition of GSK3β activated mitoK(ATP). PKC activation and protein phosphatase 2a inhibition increased the open probability of mitoK(ATP) channels through GSK3β, and this GSK3β signal was mediated via mitochondrial Cx43. Moreover, (i) PKC-induced phosphorylation of mitochondrial Cx43 was reduced in GSK3β-S9A mice; (ii) Cx43 and GSK3β proteins associated in mitochondria; and (iii) SB216763-mediated reduction of infarct size was abolished in Cx43 KO mice in vivo, consistent with the notion that GSK3β inhibition results in mitoK(ATP) opening via mitochondrial Cx43. We therefore directly targeted mitochondrial Cx43 by the Cx43 C-terminal binding peptide RRNYRRNY for cardioprotection, circumventing further upstream pathways. RRNYRRNY activated mitoK(ATP) channels via Cx43. We directly recorded mitochondrial Cx43 channels that were activated by RRNYRRNY and blocked by the Cx43 mimetic peptide (43)GAP27. RRNYRRNY rendered isolated cardiomyocytes in vitro and the heart in vivo resistant to ischemia/reperfusion injury, indicating that mitochondrial Cx43- and/or mitoK(ATP)-mediated reduction of infarct size was not undermined by RRNYRRNY-related opening of sarcolemmal Cx43 channels. Our results demonstrate that GSK3β transfers cytoprotective signaling through mitochondrial Cx43 onto mitoK(ATP) channels and that Cx43 functions as a channel in mitochondria, being an attractive target for drug treatment against cardiomyocyte injury.
Collapse
|
48
|
Palacios-Prado N, Bukauskas FF. Modulation of metabolic communication through gap junction channels by transjunctional voltage; synergistic and antagonistic effects of gating and ionophoresis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1884-94. [PMID: 21930112 DOI: 10.1016/j.bbamem.2011.09.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 08/25/2011] [Accepted: 09/02/2011] [Indexed: 01/28/2023]
Abstract
Gap junction (GJ) channels assembled from connexin (Cx) proteins provide a structural basis for direct electrical and metabolic cell-cell communication. Here, we focus on gating and permeability properties of Cx43/Cx45 heterotypic GJs exhibiting asymmetries of both voltage-gating and transjunctional flux (J(j)) of fluorescent dyes depending on transjunctional voltage (V(j)). Relatively small differences in the resting potential of communicating cells can substantially reduce or enhance this flux at relative negativity or positivity on Cx45 side, respectively. Similarly, series of V(j) pulses resembling bursts of action potentials (APs) reduce J(j) when APs initiate in the cell expressing Cx43 and increase J(j) when APs initiate in the cell expressing Cx45. J(j) of charged fluorescent dyes is affected by ionophoresis and V(j)-gating and the asymmetry of J(j)-V(j) dependence in heterotypic GJs is enhanced or reduced when ionophoresis and V(j)-gating work in a synergistic or antagonistic manner, respectively. Modulation of cell-to-cell transfer of metabolites and signaling molecules by V(j) may occur in excitable as well as non-excitable tissues and may be more expressed in the border between normal and pathological regions where intercellular gradients of membrane potential and concentration of ions are substantially altered. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.
Collapse
Affiliation(s)
- Nicolás Palacios-Prado
- Dominick P.Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | |
Collapse
|
49
|
Scemes E. Nature of plasmalemmal functional "hemichannels". BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1880-3. [PMID: 21703226 DOI: 10.1016/j.bbamem.2011.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/04/2011] [Accepted: 06/07/2011] [Indexed: 12/28/2022]
Abstract
The molecular identity of the protein forming "hemichannels" at non-junctional membranes is disputed. The family of gap junction proteins, innexins, connexins, and pannexins share several common features, including permeability characteristics and sensitivity to blocking agents. Such overlap in properties renders the identification of which of these protein species actually establishes the non-junctional membrane conductance and permeability quite complicated, especially because in vertebrates pannexins and connexins have largely overlapping distributions in tissues. Recently, attempts to establish criteria to identify events that are "hemichannel" mediated and those to allow the distinction between connexin- from pannexin-mediated events have been proposed. Here, I present an update on that topic and discuss the most recent findings related to the nature of functional "hemichannels" focusing on connexin43 and pannexin1. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.
Collapse
Affiliation(s)
- Eliana Scemes
- Dominick P.Purpura Department of Neuroscience, Albert Einstein Collegeof Medicine, Bronx, NY, USA.
| |
Collapse
|
50
|
Hilgen G, von Maltzahn J, Willecke K, Weiler R, Dedek K. Subcellular distribution of connexin45 in OFF bipolar cells of the mouse retina. J Comp Neurol 2011; 519:433-50. [PMID: 21192077 DOI: 10.1002/cne.22526] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In the mouse retina, connexin45 (Cx45) participates in the gap junction between ON cone bipolar cells and AII amacrine cells, which constitutes an essential element of the primary rod pathway. Although it has been shown that Cx45 is also expressed in OFF bipolar cells, its subcellular localization and functional role in these cells are unknown. Here, we analyzed the localization of Cx45 on OFF bipolar cells in the mouse retina. For this, we used wild-type mice and a transgenic mouse line that expressed, in addition to native Cx45, a fusion protein consisting of Cx45 and the enhanced green fluorescent protein (EGFP). Cx45-EGFP expression generates an EGFP signal at gap junctions containing Cx45. Combining immunohistochemistry with intracellular injections, we found that Cx45 was present on dendrites and axon terminals of all OFF bipolar cell types. Cx45 was not found at intersections of two terminal processes of the same type, suggesting that Cx45 might not form gap junctions between axon terminals of the same OFF bipolar cell type but rather might connect OFF bipolar cells to amacrine or ganglion cells. In OFF bipolar cell dendrites, Cx45 was found predominantly in the proximal outer plexiform layer (OPL), well below the cone pedicles. Cx45 did not colocalize with Cx36, which is found predominantly in the distal OPL. We conclude that Cx45 is expressed on OFF bipolar cell dendrites, presumably forming gap junctions with cells of the same type, and on OFF bipolar cell axon terminals, presumably forming heterologous gap junctions with other retinal neurons.
Collapse
Affiliation(s)
- Gerrit Hilgen
- Department of Neurobiology, University of Oldenburg, Germany
| | | | | | | | | |
Collapse
|