1
|
Liu Y, Li C, Freites JA, Tobias DJ, Voth GA. Quantitative insights into the mechanism of proton conduction and selectivity for the human voltage-gated proton channel Hv1. Proc Natl Acad Sci U S A 2024; 121:e2407479121. [PMID: 39259593 PMCID: PMC11420211 DOI: 10.1073/pnas.2407479121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 08/09/2024] [Indexed: 09/13/2024] Open
Abstract
Human voltage-gated proton (hHv1) channels are crucial for regulating essential biological processes such as immune cell respiratory burst, sperm capacitation, and cancer cell migration. Despite the significant concentration difference between protons and other ions in physiological conditions, hHv1 demonstrates remarkable proton selectivity. Our calculations of single-proton, cation, and anion permeation free energy profiles quantitatively demonstrate that the proton selectivity of the wild-type channel originates from its strong proton affinity via the titration of the key residues D112 and D174, although the channel imposes similar kinetic blocking effects for protons compared to other ions. A two-proton knock-on model is proposed to mathematically explain the electrophysiological measurements of the pH-dependent proton conductance in the conductive state. Moreover, it is shown that the anion selectivity of the D112N mutant channel is tied to impaired proton transport and substantial anion leakage.
Collapse
Affiliation(s)
- Yu Liu
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics, and James Frank Institute, University of Chicago, Chicago, IL 60637
| | - Chenghan Li
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics, and James Frank Institute, University of Chicago, Chicago, IL 60637
| | | | - Douglas J Tobias
- Department of Chemistry, University of California, Irvine, CA 92697
| | - Gregory A Voth
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics, and James Frank Institute, University of Chicago, Chicago, IL 60637
| |
Collapse
|
2
|
Kuwabara MF, Klemptner J, Muth J, De Martino E, Oliver D, Berger TK. Zinc inhibits the voltage-gated proton channel HCNL1. Biophys J 2024:S0006-3495(24)00565-4. [PMID: 39210595 DOI: 10.1016/j.bpj.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/31/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Voltage-gated ion channels allow ion flux across biological membranes in response to changes in the membrane potential. HCNL1 is a recently discovered voltage-gated ion channel that selectively conducts protons through its voltage-sensing domain (VSD), reminiscent of the well-studied depolarization-activated Hv1 proton channel. However, HCNL1 is activated by hyperpolarization, allowing the influx of protons, which leads to an intracellular acidification in zebrafish sperm. Zinc ions (Zn2+) are important cofactors in many proteins and essential for sperm physiology. Proton channels such as Hv1 and Otopetrin1 are inhibited by Zn2+. We investigated the effect of Zn2+ on heterologously expressed HCNL1 channels using electrophysiological and fluorometric techniques. Extracellular Zn2+ inhibits HCNL1 currents with an apparent half-maximal inhibition (IC50) of 26 μM. Zn2+ slows voltage-dependent current kinetics, shifts the voltage-dependent activation to more negative potentials, and alters hyperpolarization-induced conformational changes of the voltage sensor. Our data suggest that extracellular Zn2+ inhibits HCNL1 currents by multiple mechanisms, including modulation of channel gating. Two histidine residues located at the extracellular side of the VSD might weakly contribute to Zn2+ coordination: mutants with either histidine replaced with alanine show modest shifts of the IC50 values to higher concentrations. Interestingly, Zn2+ inhibits HCNL1 at even lower concentrations from the intracellular side (IC50 ≈ 0.5 μM). A histidine residue at the intracellular end of S1 (position 50) is important for Zn2+ binding: much higher Zn2+ concentrations are required to inhibit the mutant HCNL1-H50A (IC50 ≈ 106 μM). We anticipate that Zn2+ will be a useful ion to study the structure-function relationship of HCNL1 as well as the physiological role of HCNL1 in zebrafish sperm.
Collapse
Affiliation(s)
- Makoto F Kuwabara
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany
| | - Joschua Klemptner
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany
| | - Julia Muth
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany
| | - Emilia De Martino
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany
| | - Dominik Oliver
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany
| | - Thomas K Berger
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
3
|
Shen M, Huang Y, Cai Z, Cherny VV, DeCoursey TE, Shen J. Interior pH-sensing residue of human voltage-gated proton channel H v1 is histidine 168. Biophys J 2024:S0006-3495(24)00486-7. [PMID: 39054673 DOI: 10.1016/j.bpj.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/07/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
The molecular mechanisms governing the human voltage-gated proton channel hHv1 remain elusive. Here, we used membrane-enabled hybrid-solvent continuous constant pH molecular dynamics (CpHMD) simulations with pH replica exchange to further evaluate the structural models of hHv1 in the closed (hyperpolarized) and open (depolarized) states recently obtained with MD simulations and explore potential pH-sensing residues. The CpHMD titration at a set of symmetric pH conditions revealed three residues that can gain or lose protons upon channel depolarization. Among them, residue H168 at the intracellular end of the S3 helix switches from the deprotonated to the protonated state and its protonation is correlated with the increased tilting of the S3 helix during the transition from the closed to the open state. Thus, the simulation data suggest H168 as an interior pH sensor, in support of a recent finding based on electrophysiological experiments of Hv1 mutants. We propose that protonation of H168 acts as a key that unlocks the closed channel configuration by increasing the flexibility of the S2-S3 linker, which increases the tilt angle of S3 and enhances the mobility of the S4 helix, thus promoting channel opening. Our work represents an important step toward deciphering the pH-dependent gating mechanism of hHv1.
Collapse
Affiliation(s)
- Mingzhe Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Yandong Huang
- College of Computer Engineering, Jimei University, Xiamen, Fujian Province, China.
| | - Zhitao Cai
- College of Computer Engineering, Jimei University, Xiamen, Fujian Province, China
| | - Vladimir V Cherny
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, Illinois
| | - Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, Illinois
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland.
| |
Collapse
|
4
|
Chávez JC, Carrasquel-Martínez G, Hernández-Garduño S, Matamoros Volante A, Treviño CL, Nishigaki T, Darszon A. Cytosolic and Acrosomal pH Regulation in Mammalian Sperm. Cells 2024; 13:865. [PMID: 38786087 PMCID: PMC11120249 DOI: 10.3390/cells13100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
As in most cells, intracellular pH regulation is fundamental for sperm physiology. Key sperm functions like swimming, maturation, and a unique exocytotic process, the acrosome reaction, necessary for gamete fusion, are deeply influenced by pH. Sperm pH regulation, both intracellularly and within organelles such as the acrosome, requires a coordinated interplay of various transporters and channels, ensuring that this cell is primed for fertilization. Consistent with the pivotal importance of pH regulation in mammalian sperm physiology, several of its unique transporters are dependent on cytosolic pH. Examples include the Ca2+ channel CatSper and the K+ channel Slo3. The absence of these channels leads to male infertility. This review outlines the main transport elements involved in pH regulation, including cytosolic and acrosomal pH, that participate in these complex functions. We present a glimpse of how these transporters are regulated and how distinct sets of them are orchestrated to allow sperm to fertilize the egg. Much research is needed to begin to envision the complete set of players and the choreography of how cytosolic and organellar pH are regulated in each sperm function.
Collapse
Affiliation(s)
- Julio C. Chávez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Gabriela Carrasquel-Martínez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
- CITMER, Medicina Reproductiva, México City 11520, Mexico
| | - Sandra Hernández-Garduño
- Departamento de Morfología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), México City 04510, Mexico;
| | - Arturo Matamoros Volante
- Department of Electrical and Computer Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Claudia L. Treviño
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Takuya Nishigaki
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico; (J.C.C.); (G.C.-M.)
| |
Collapse
|
5
|
Tang Y, Wu X, Li J, Li Y, Xu X, Li G, Zhang P, Qin C, Wu LJ, Tang Z, Tian DS. The Emerging Role of Microglial Hv1 as a Target for Immunomodulation in Myelin Repair. Aging Dis 2024; 15:1176-1203. [PMID: 38029392 PMCID: PMC11081154 DOI: 10.14336/ad.2023.1107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
In the central nervous system (CNS), the myelin sheath ensures efficient interconnection between neurons and contributes to the regulation of the proper function of neuronal networks. The maintenance of myelin and the well-organized subtle process of myelin plasticity requires cooperation among myelin-forming cells, glial cells, and neural networks. The process of cooperation is fragile, and the balance is highly susceptible to disruption by microenvironment influences. Reactive microglia play a critical and complicated role in the demyelination and remyelination process. Recent studies have shown that the voltage-gated proton channel Hv1 is selectively expressed in microglia in CNS, which regulates intracellular pH and is involved in the production of reactive oxygen species, underlying multifaceted roles in maintaining microglia function. This paper begins by examining the molecular mechanisms of demyelination and emphasizes the crucial role of the microenvironment in demyelination. It focuses specifically on the role of Hv1 in myelin repair and its therapeutic potential in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Ayuyan AG, Cherny VV, Chaves G, Musset B, Cohen FS, DeCoursey TE. Interaction with stomatin directs human proton channels into cholesterol-dependent membrane domains. Biophys J 2024:S0006-3495(24)00168-1. [PMID: 38444158 DOI: 10.1016/j.bpj.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/24/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024] Open
Abstract
Many membrane proteins are modulated by cholesterol. Here we report profound effects of cholesterol depletion and restoration on the human voltage-gated proton channel, hHV1, in excised patches but negligible effects in the whole-cell configuration. Despite the presence of a putative cholesterol-binding site, a CARC motif in hHV1, mutation of this motif did not affect cholesterol effects. The murine HV1 lacks a CARC sequence but displays similar cholesterol effects. These results argue against a direct effect of cholesterol on the HV1 protein. However, the data are fully explainable if HV1 preferentially associates with cholesterol-dependent lipid domains, or "rafts." The rafts would be expected to concentrate in the membrane/glass interface and to be depleted from the electrically accessible patch membrane. This idea is supported by evidence that HV1 channels can diffuse between seal and patch membranes when suction is applied. Simultaneous truncation of the large intracellular N and C termini of hHV1 greatly attenuated the cholesterol effect, but C truncation alone did not; this suggests that the N terminus is the region of attachment to lipid domains. Searching for abundant raft-associated proteins led to stomatin. Co-immunoprecipitation experiment results were consistent with hHV1 binding to stomatin. The stomatin-mediated association of HV1 with cholesterol-dependent lipid domains provides a mechanism for cells to direct HV1 to subcellular locations where it is needed, such as the phagosome in leukocytes.
Collapse
Affiliation(s)
- Artem G Ayuyan
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois.
| | - Vladimir V Cherny
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois
| | - Gustavo Chaves
- Institut für Physiologie, Pathophysiologie und Biophysik, CPPB, Paracelsus Medical University, Nürnberg, Germany
| | - Boris Musset
- Institut für Physiologie, Pathophysiologie und Biophysik, CPPB, Paracelsus Medical University, Nürnberg, Germany
| | - Fredric S Cohen
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois
| | - Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois.
| |
Collapse
|
7
|
DeCoursey TE. Transcendent Aspects of Proton Channels. Annu Rev Physiol 2024; 86:357-377. [PMID: 37931166 PMCID: PMC10938948 DOI: 10.1146/annurev-physiol-042222-023242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
A handful of biological proton-selective ion channels exist. Some open at positive or negative membrane potentials, others open at low or high pH, and some are light activated. This review focuses on common features that result from the unique properties of protons. Proton conduction through water or proteins differs qualitatively from that of all other ions. Extraordinary proton selectivity is needed to ensure that protons permeate and other ions do not. Proton selectivity arises from a proton pathway comprising a hydrogen-bonded chain that typically includes at least one titratable amino acid side chain. The enormously diverse functions of proton channels in disparate regions of the phylogenetic tree can be summarized by considering the chemical and electrical consequences of proton flux across membranes. This review discusses examples of cells in which proton efflux serves to increase pHi, decrease pHo, control the membrane potential, generate action potentials, or compensate transmembrane movement of electrical charge.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois, USA;
| |
Collapse
|
8
|
Grahn E, Kaufmann SV, Askarova M, Ninov M, Welp LM, Berger TK, Urlaub H, Kaupp UB. Control of intracellular pH and bicarbonate by CO 2 diffusion into human sperm. Nat Commun 2023; 14:5395. [PMID: 37669933 PMCID: PMC10480191 DOI: 10.1038/s41467-023-40855-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 08/14/2023] [Indexed: 09/07/2023] Open
Abstract
The reaction of CO2 with H2O to form bicarbonate (HCO3-) and H+ controls sperm motility and fertilization via HCO3--stimulated cAMP synthesis. A complex network of signaling proteins participates in this reaction. Here, we identify key players that regulate intracellular pH (pHi) and HCO3- in human sperm by quantitative mass spectrometry (MS) and kinetic patch-clamp fluorometry. The resting pHi is set by amiloride-sensitive Na+/H+ exchange. The sperm-specific putative Na+/H+ exchanger SLC9C1, unlike its sea urchin homologue, is not gated by voltage or cAMP. Transporters and channels implied in HCO3- transport are not detected, and may be present at copy numbers < 10 molecules/sperm cell. Instead, HCO3- is produced by diffusion of CO2 into cells and readjustment of the CO2/HCO3-/H+ equilibrium. The proton channel Hv1 may serve as a unidirectional valve that blunts the acidification ensuing from HCO3- synthesis. This work provides a new framework for the study of male infertility.
Collapse
Affiliation(s)
- Elena Grahn
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Svenja V Kaufmann
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Malika Askarova
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Momchil Ninov
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Luisa M Welp
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Thomas K Berger
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Deutschhausstrasse 1-2, 35037, Marburg, Germany.
| | - Henning Urlaub
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany.
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
- Cluster of Excellence, Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Göttingen, Germany.
| | - U Benjamin Kaupp
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
- Life & Medical Sciences Institute (LIMES), University Bonn, Carl-Troll-Strasse 31, 53115, Bonn, Germany.
| |
Collapse
|
9
|
Chaves G, Jardin C, Derst C, Musset B. Voltage-Gated Proton Channels in the Tree of Life. Biomolecules 2023; 13:1035. [PMID: 37509071 PMCID: PMC10377628 DOI: 10.3390/biom13071035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
With a single gene encoding HV1 channel, proton channel diversity is particularly low in mammals compared to other members of the superfamily of voltage-gated ion channels. Nonetheless, mammalian HV1 channels are expressed in many different tissues and cell types where they exert various functions. In the first part of this review, we regard novel aspects of the functional expression of HV1 channels in mammals by differentially comparing their involvement in (1) close conjunction with the NADPH oxidase complex responsible for the respiratory burst of phagocytes, and (2) in respiratory burst independent functions such as pH homeostasis or acid extrusion. In the second part, we dissect expression of HV channels within the eukaryotic tree of life, revealing the immense diversity of the channel in other phylae, such as mollusks or dinoflagellates, where several genes encoding HV channels can be found within a single species. In the last part, a comprehensive overview of the biophysical properties of a set of twenty different HV channels characterized electrophysiologically, from Mammalia to unicellular protists, is given.
Collapse
Affiliation(s)
- Gustavo Chaves
- Center of Physiology, Pathophysiology and Biophysics, The Nuremberg Location, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Christophe Jardin
- Center of Physiology, Pathophysiology and Biophysics, The Nuremberg Location, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Christian Derst
- Center of Physiology, Pathophysiology and Biophysics, The Nuremberg Location, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Boris Musset
- Center of Physiology, Pathophysiology and Biophysics, The Nuremberg Location, Paracelsus Medical University, 90419 Nuremberg, Germany
- Center of Physiology, Pathophysiology and Biophysics, The Salzburg Location, Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
10
|
Han S, Applewhite S, DeCata J, Jones S, Cummings J, Wang S. Arachidonic acid reverses cholesterol and zinc inhibition of human voltage-gated proton channels. J Biol Chem 2023:104918. [PMID: 37315791 PMCID: PMC10344949 DOI: 10.1016/j.jbc.2023.104918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/08/2023] [Accepted: 06/09/2023] [Indexed: 06/16/2023] Open
Abstract
Unlike other members of the voltage-gated ion channel superfamily, voltage-gated proton (Hv) channels are solely composed of voltage sensor domains without separate ion-conducting pores. Due to their unique dependence on both voltage and transmembrane pH gradients, Hv channels normally open to mediate proton efflux. Multiple cellular ligands were also found to regulate the function of Hv channels, including Zn2+, cholesterol, polyunsaturated arachidonic acid, and albumin. Our previous work showed that Zn2+ and cholesterol inhibit the human voltage-gated proton channel hHv1 by stabilizing its S4 segment at resting state conformations. Released from phospholipids by phospholipase A2 in cells upon infection or injury, arachidonic acid regulates the function of many ion channels, including hHv1. In the present work, we examined the effects of arachidonic acid on purified hHv1 channels using liposome flux assays and revealed underlying structural mechanisms using single-molecule Fluorescence Resonance Energy Transfer (smFRET). Our data indicated that arachidonic acid strongly activates hHv1 channels by promoting transitions of the S4 segment towards opening or 'pre-opening' conformations. Moreover, we found that arachidonic acid even activates hHv1 channels inhibited by Zn2+ and cholesterol, providing a biophysical mechanism to activate hHv1 channels in non-excitable cells upon infection or injury.
Collapse
Affiliation(s)
- Shuo Han
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Sarah Applewhite
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Jenna DeCata
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Samuel Jones
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - John Cummings
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Shizhen Wang
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA.
| |
Collapse
|
11
|
Cozzolino M, Gyöngyösi A, Korpos E, Gogolak P, Naseem MU, Kállai J, Lanyi A, Panyi G. The Voltage-Gated Hv1 H+ Channel Is Expressed in Tumor-Infiltrating Myeloid-Derived Suppressor Cells. Int J Mol Sci 2023; 24:ijms24076216. [PMID: 37047188 PMCID: PMC10094655 DOI: 10.3390/ijms24076216] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key determinants of the immunosuppressive microenvironment in tumors. As ion channels play key roles in the physiology/pathophysiology of immune cells, we aimed at studying the ion channel repertoire in tumor-derived polymorphonuclear (PMN-MDSC) and monocytic (Mo-MDSC) MDSCs. Subcutaneous tumors in mice were induced by the Lewis lung carcinoma cell line (LLC). The presence of PMN-MDSC (CD11b+/Ly6G+) and Mo-MDSCs (CD11b+/Ly6C+) in the tumor tissue was confirmed using immunofluorescence microscopy and cells were identified as CD11b+/Ly6G+ PMN-MDSCs and CD11b+/Ly6C+/F4/80−/MHCII− Mo-MDSCs using flow cytometry and sorting. The majority of the myeloid cells infiltrating the LLC tumors were PMN-MDSC (~60%) as compared to ~10% being Mo-MDSCs. We showed that PMN- and Mo-MDSCs express the Hv1 H+ channel both at the mRNA and at the protein level and that the biophysical and pharmacological properties of the whole-cell currents recapitulate the hallmarks of Hv1 currents: ~40 mV shift in the activation threshold of the current per unit change in the extracellular pH, high H+ selectivity, and sensitivity to the Hv1 inhibitor ClGBI. As MDSCs exert immunosuppression mainly by producing reactive oxygen species which is coupled to Hv1-mediated H+ currents, Hv1 might be an attractive target for inhibition of MDSCs in tumors.
Collapse
Affiliation(s)
- Marco Cozzolino
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
| | - Adrienn Gyöngyösi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Eva Korpos
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Peter Gogolak
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Muhammad Umair Naseem
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
| | - Judit Kállai
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Arpad Lanyi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
- Correspondence: ; Tel.: +36-52-352201
| |
Collapse
|
12
|
Demchenko AP. Proton transfer reactions: from photochemistry to biochemistry and bioenergetics. BBA ADVANCES 2023. [DOI: 10.1016/j.bbadva.2023.100085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023] Open
|
13
|
Chaves G, Ayuyan AG, Cherny VV, Morgan D, Franzen A, Fieber L, Nausch L, Derst C, Mahorivska I, Jardin C, DeCoursey TE, Musset B. Unexpected expansion of the voltage-gated proton channel family. FEBS J 2023; 290:1008-1026. [PMID: 36062330 PMCID: PMC10911540 DOI: 10.1111/febs.16617] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 11/27/2022]
Abstract
Voltage-gated ion channels, whose first identified function was to generate action potentials, are divided into subfamilies with numerous members. The family of voltage-gated proton channels (HV ) is tiny. To date, all species found to express HV have exclusively one gene that codes for this unique ion channel. Here we report the discovery and characterization of three proton channel genes in the classical model system of neural plasticity, Aplysia californica. The three channels (AcHV 1, AcHV 2, and AcHV 3) are distributed throughout the whole animal. Patch-clamp analysis confirmed proton selectivity of these channels but they all differed markedly in gating. AcHV 1 gating resembled HV in mammalian cells where it is responsible for proton extrusion and charge compensation. AcHV 2 activates more negatively and conducts extensive inward proton current, properties likely to acidify the cytosol. AcHV 3, which differs from AcHV 1 and AcHV 2 in lacking the first arginine in the S4 helix, exhibits proton selective leak currents and weak voltage dependence. We report the expansion of the proton channel family, demonstrating for the first time the expression of three functionally distinct proton channels in a single species.
Collapse
Affiliation(s)
- Gustavo Chaves
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
| | - Artem G Ayuyan
- Department of Physiology & Biophysics, Rush University, Chicago, IL, USA
| | - Vladimir V Cherny
- Department of Physiology & Biophysics, Rush University, Chicago, IL, USA
| | - Deri Morgan
- Department of Radiation Oncology, University of Kansas Medical Center, MO, USA
| | - Arne Franzen
- Institute of Biological Information Processing, Molecular and Cellular Physiology (IBI-1), Jülich, Germany
| | - Lynne Fieber
- Department of Marine Biology and Ecology - Rosenstiel School of Marine and Atmospheric Science, Miami, FL, USA
| | - Lydia Nausch
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
- Department of Agriculture, Food and Nutrition, Institute of Nutrition and Food Supply Management, University of Applied Sciences Weihenstephan-Triesdorf, Freising, Germany
| | - Christian Derst
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
| | - Iryna Mahorivska
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
| | - Christophe Jardin
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
| | - Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, IL, USA
| | - Boris Musset
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
14
|
Suárez-Delgado E, Orozco-Contreras M, Rangel-Yescas GE, Islas LD. Activation-pathway transitions in human voltage-gated proton channels revealed by a non-canonical fluorescent amino acid. eLife 2023; 12:85836. [PMID: 36695566 PMCID: PMC9925047 DOI: 10.7554/elife.85836] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Voltage-dependent gating of the voltage-gated proton channels (HV1) remains poorly understood, partly because of the difficulty of obtaining direct measurements of voltage sensor movement in the form of gating currents. To circumvent this problem, we have implemented patch-clamp fluorometry in combination with the incorporation of the fluorescent non-canonical amino acid Anap to monitor channel opening and movement of the S4 segment. Simultaneous recording of currents and fluorescence signals allows for direct correlation of these parameters and investigation of their dependence on voltage and the pH gradient (ΔpH). We present data that indicate that Anap incorporated in the S4 helix is quenched by an aromatic residue located in the S2 helix and that motion of the S4 relative to this quencher is responsible for fluorescence increases upon depolarization. The kinetics of the fluorescence signal reveal the existence of a very slow transition in the deactivation pathway, which seems to be singularly regulated by ΔpH. Our experiments also suggest that the voltage sensor can move after channel opening and that the absolute value of the pH can influence the channel opening step. These results shed light on the complexities of voltage-dependent opening of human HV1 channels.
Collapse
Affiliation(s)
- Esteban Suárez-Delgado
- Department of Physiology, School of Medicine, Universidad Nacional Autónoma de México, México City, Mexico
| | - Maru Orozco-Contreras
- Department of Physiology, School of Medicine, Universidad Nacional Autónoma de México, México City, Mexico
| | - Gisela E Rangel-Yescas
- Department of Physiology, School of Medicine, Universidad Nacional Autónoma de México, México City, Mexico
| | - Leon D Islas
- Department of Physiology, School of Medicine, Universidad Nacional Autónoma de México, México City, Mexico
| |
Collapse
|
15
|
Alvear-Arias JJ, Pena-Pichicoi A, Carrillo C, Fernandez M, Gonzalez T, Garate JA, Gonzalez C. Role of voltage-gated proton channel (Hv1) in cancer biology. Front Pharmacol 2023; 14:1175702. [PMID: 37153807 PMCID: PMC10157179 DOI: 10.3389/fphar.2023.1175702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/11/2023] [Indexed: 05/10/2023] Open
Abstract
The acid-base characteristics of tumor cells and the other elements that compose the tumor microenvironment have been topics of scientific interest in oncological research. There is much evidence confirming that pH conditions are maintained by changes in the patterns of expression of certain proton transporters. In the past decade, the voltage-gated proton channel (Hv1) has been added to this list and is increasingly being recognized as a target with onco-therapeutic potential. The Hv1 channel is key to proton extrusion for maintaining a balanced cytosolic pH. This protein-channel is expressed in a myriad of tissues and cell lineages whose functions vary from producing bioluminescence in dinoflagellates to alkalizing spermatozoa cytoplasm for reproduction, and regulating the respiratory burst for immune system response. It is no wonder that in acidic environments such as the tumor microenvironment, an exacerbated expression and function of this channel has been reported. Indeed, multiple studies have revealed a strong relationship between pH balance, cancer development, and the overexpression of the Hv1 channel, being proposed as a marker for malignancy in cancer. In this review, we present data that supports the idea that the Hv1 channel plays a significant role in cancer by maintaining pH conditions that favor the development of malignancy features in solid tumor models. With the antecedents presented in this bibliographic report, we want to strengthen the idea that the Hv1 proton channel is an excellent therapeutic strategy to counter the development of solid tumors.
Collapse
Affiliation(s)
- Juan J. Alvear-Arias
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Universidad de Valparaíso, Valparaíso, Chile
| | - Antonio Pena-Pichicoi
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Universidad de Valparaíso, Valparaíso, Chile
| | - Christian Carrillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Universidad de Valparaíso, Valparaíso, Chile
| | - Miguel Fernandez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Universidad de Valparaíso, Valparaíso, Chile
| | - Tania Gonzalez
- National Center for Minimally Invasive Surgery, La Habana, Cuba
| | - Jose A. Garate
- Millennium Nucleus in NanoBioPhysics, Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago, Chile
| | - Carlos Gonzalez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Universidad de Valparaíso, Valparaíso, Chile
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, United States
- *Correspondence: Carlos Gonzalez,
| |
Collapse
|
16
|
Wu X, Zhang L, Hong L. The role of Phe150 in human voltage-gated proton channel. iScience 2022; 25:105420. [PMID: 36388967 PMCID: PMC9646954 DOI: 10.1016/j.isci.2022.105420] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/15/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
The voltage-gated proton channel Hv1 is a member of voltage-gated ion channels containing voltage-sensing domains (VSDs). The VSDs are made of four membrane-spanning segments (S1 through S4), and their function is to detect changes in membrane potential in the cells. A highly conserved phenylalanine 150 (F150) is located in the S2 segment of human voltage-gated proton channels. We previously discovered that the F150 is a binding site for the open channel blocker 2GBI. Here, we show that the Hv1 VSD voltage-dependent activation requires a hydrophobic group at position F150. We perform double-mutant cycle analysis to probe interactions between F150 and positively charged arginines in the S4 segment of the channel. Our results indicate that F150 interacts with two arginines (R2 and R3) in the S4 segment and catalyzes the transfer of the S4 arginines in the process of voltage-dependent activation. Hydrophobicity of F150 is crucial for human Hv1 channel voltage-dependent activation F150 interacts with R2 to stabilize the closed state of the Hv1 channel When depolarized, R3 moves upward to interact with F150 stabilizing the open state of Hv1 F150 is essential for the transfer of the Hv1 arginines in the process of voltage sensing
Collapse
Affiliation(s)
- Xin Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lu Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
- Corresponding author
| |
Collapse
|
17
|
Abstract
Although human sperm is morphologically mature in the epididymis, it cannot fertilize eggs before capacitation. Cholesterol efflux from the sperm plasma membrane is a key molecular event essential for cytoplasmic alkalinization and hyperactivation, but the underlying mechanism remains unclear. The human voltage-gated proton (hHv1) channel functions as an acid extruder to regulate intracellular pHs of many cell types, including sperm. Aside from voltage and pH, Hv channels are also regulated by distinct ligands, such as Zn2+ and albumin. In the present work, we identified cholesterol as an inhibitory ligand of the hHv1 channel and further investigated the underlying mechanism using the single-molecule fluorescence resonance energy transfer (smFRET) approach. Our results indicated that cholesterol inhibits the hHv1 channel by stabilizing the voltage-sensing S4 segment at resting conformations, a similar mechanism also utilized by Zn2+. Our results suggested that the S4 segment is the central gating machinery in the hHv1 channel, on which voltage and distinct ligands are converged to regulate channel function. Identification of membrane cholesterol as an inhibitory ligand provides a mechanism by which the hHv1 channel regulates fertilization by linking the cholesterol efflux with cytoplasmic alkalinization, a change that triggers calcium influx through the CatSper channel. These events finally lead to hyperactivation, a remarkable change in the mobility pattern indicating fertilization competence of human sperm.
Collapse
|
18
|
Delgado-Bermúdez A, Yeste M, Bonet S, Pinart E. A Review on the Role of Bicarbonate and Proton Transporters during Sperm Capacitation in Mammals. Int J Mol Sci 2022; 23:ijms23116333. [PMID: 35683013 PMCID: PMC9180951 DOI: 10.3390/ijms23116333] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/01/2022] [Accepted: 06/05/2022] [Indexed: 12/14/2022] Open
Abstract
Alkalinization of sperm cytosol is essential for plasma membrane hyperpolarization, hyperactivation of motility, and acrosomal exocytosis during sperm capacitation in mammals. The plasma membrane of sperm cells contains different ion channels implicated in the increase of internal pH (pHi) by favoring either bicarbonate entrance or proton efflux. Bicarbonate transporters belong to the solute carrier families 4 (SLC4) and 26 (SLC26) and are currently grouped into Na+/HCO3− transporters and Cl−/HCO3− exchangers. Na+/HCO3− transporters are reported to be essential for the initial and fast entrance of HCO3− that triggers sperm capacitation, whereas Cl−/HCO3− exchangers are responsible for the sustained HCO3− entrance which orchestrates the sequence of changes associated with sperm capacitation. Proton efflux is required for the fast alkalinization of capacitated sperm cells and the activation of pH-dependent proteins; according to the species, this transport can be mediated by Na+/H+ exchangers (NHE) belonging to the SLC9 family and/or voltage-gated proton channels (HVCN1). Herein, we discuss the involvement of each of these channels in sperm capacitation and the acrosome reaction.
Collapse
Affiliation(s)
- Ariadna Delgado-Bermúdez
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; (A.D.-B.); (M.Y.); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; (A.D.-B.); (M.Y.); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), ES-08010 Barcelona, Spain
| | - Sergi Bonet
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; (A.D.-B.); (M.Y.); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
| | - Elisabeth Pinart
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, ES-17003 Girona, Spain; (A.D.-B.); (M.Y.); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, ES-17003 Girona, Spain
- Correspondence: ; Tel.: +34-972-419-514
| |
Collapse
|
19
|
Jardin C, Ohlwein N, Franzen A, Chaves G, Musset B. The pH-dependent gating of the human voltage-gated proton channel from computational simulations. Phys Chem Chem Phys 2022; 24:9964-9977. [PMID: 35445675 DOI: 10.1039/d1cp05609c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Gating of the voltage-gated proton channel HV1 is strongly controlled by pH. There is evidence that this involves the sidechains of titratable amino acids that change their protonation state with changes of the pH. Despite experimental investigations to identify the amino acids involved in pH sensing only few progress has been made, including one histidine at the cytoplasmic side of the channel that is involved in sensing cellular pH. We have used constant pH molecular dynamics simulations in symmetrical and asymmetrical pH conditions across the membrane to investigate the pH- and ΔpH-dependent gating of the human HV1 channel. Therefore, the pKa of every titratable amino acids has been assessed in single simulations. Our simulations captured initial conformational changes between a deactivated and an activated state of the channel induced solely by changes of the pH. The pH-dependent gating is accompanied by an outward displacement of the three S4 voltage sensing arginines that moves the second arginine past the hydrophobic gasket (HG) which separates the inner and outer pores of the channel. HV1 activation, when outer pH increases, involves amino acids at the extracellular entrance of the channel that extend the network of interactions from the external solution down to the HG. Whereas, amino acids at the cytoplasmic entrance of the channel are involved in activation, when inner pH decreases, and in a network of interactions that extend from the cytoplasm up to the HG.
Collapse
Affiliation(s)
- Christophe Jardin
- Klinikum Nürnberg Medical School, CPPB, Institute of Physiology, Pathophysiology and Biophysics, Nuremberg, Germany.
| | - Niklas Ohlwein
- Klinikum Nürnberg Medical School, CPPB, Institute of Physiology, Pathophysiology and Biophysics, Nuremberg, Germany. .,Klinik für Anästhesiologie und operative Intensivmedizin, Universitätklinik der Paracelsus Medizinischen Privatuniversität, Nuremberg, Germany
| | - Arne Franzen
- Institute of Biological Information Processing, Molecular and Cellular Physiology (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | - Gustavo Chaves
- Klinikum Nürnberg Medical School, CPPB, Institute of Physiology, Pathophysiology and Biophysics, Nuremberg, Germany.
| | - Boris Musset
- Klinikum Nürnberg Medical School, CPPB, Institute of Physiology, Pathophysiology and Biophysics, Nuremberg, Germany.
| |
Collapse
|
20
|
Alvear-Arias JJ, Carrillo C, Villar JP, Garcia-Betancourt R, Peña-Pichicoi A, Fernandez A, Fernandez M, Carmona EM, Pupo A, Neely A, Alvarez O, Garate J, Barajas-Martinez H, Larsson HP, Lopez-Rodriguez A, Latorre R, Gonzalez C. Expression of H v1 proton channels in myeloid-derived suppressor cells (MDSC) and its potential role in T cell regulation. Proc Natl Acad Sci U S A 2022; 119:e2104453119. [PMID: 35377790 PMCID: PMC9169626 DOI: 10.1073/pnas.2104453119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 02/14/2022] [Indexed: 12/07/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous cell population with high immunosuppressive activity that proliferates in infections, inflammation, and tumor microenvironments. In tumors, MDSC exert immunosuppression mainly by producing reactive oxygen species (ROS), a process triggered by the NADPH oxidase 2 (NOX2) activity. NOX2 is functionally coupled with the Hv1 proton channel in certain immune cells to support sustained free-radical production. However, a functional expression of the Hv1 channel in MDSC has not yet been reported. Here, we demonstrate that mouse MDSC express functional Hv1 proton channel by immunofluorescence microscopy, flow cytometry, and Western blot, besides performing a biophysical characterization of its macroscopic currents via patch-clamp technique. Our results show that the immunosuppression by MDSC is conditional to their ability to decrease the proton concentration elevated by the NOX2 activity, rendering Hv1 a potential drug target for cancer treatment.
Collapse
Affiliation(s)
- Juan J. Alvear-Arias
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Christian Carrillo
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Javiera Paz Villar
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Richard Garcia-Betancourt
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Antonio Peña-Pichicoi
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Audry Fernandez
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Miguel Fernandez
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Emerson M. Carmona
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Amaury Pupo
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Alan Neely
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Osvaldo Alvarez
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
| | - Jose Garate
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | | | - H. Peter Larsson
- Department of Physiology & Biophysics, University of Miami, Coral Gables, FL 33101
| | | | - Ramon Latorre
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Carlos Gonzalez
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
| |
Collapse
|
21
|
Hermann C, Treder A, Näher M, Geiseler R, Gudermann T, Mederos Y Schnitzler M, Storch U. The normalized slope conductance as a tool for quantitative analysis of current-voltage relations. Biophys J 2022; 121:1435-1448. [PMID: 35300969 PMCID: PMC9072577 DOI: 10.1016/j.bpj.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/02/2021] [Accepted: 03/11/2022] [Indexed: 11/29/2022] Open
Abstract
The patch-clamp method which was rewarded with the Nobel Prize in 1991 is a well-established and indispensable method to study ion channels in living cells and to biophysically characterize non-voltage-gated ion channels that comprise about 70% of all ion channels in the human genome. To investigate the biophysical properties of non-voltage-gated ion channels, whole-cell measurements with application of continuous voltage-ramps are routinely conducted to obtain current-voltage (IV) relationships. However, adequate tools for detailed and quantitative analysis of IV curves are still missing. We use the example of the transient receptor potential classical (TRPC) channel family to elucidate whether the normalized slope conductance (NSC) is an appropriate tool for a reliable discrimination of the IV curves of diverse TRPC channels that differ in their individual curve progression. We provide a robust calculation method for the NSC, and by applying this method we find that TRPC channel activators and modulators can evoke different NSC progressions independent from their expression levels which is pointing to distinguishable active channel states. TRPC6 mutations of patients suffering from focal segmental glomerulosclerosis (FSGS) resulted in distinct NSC progressions suggesting that the NSC is suitable to investigate structure-function relations and might help unravel the unknown pathomechanisms leading to FSGS. Altogether, the NCS represents an effective algorithm for extended biophysical characterization of non-voltage-gated ion channels.
Collapse
Affiliation(s)
- Christian Hermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Aaron Treder
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Marius Näher
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Roman Geiseler
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany;; Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research, Munich, Germany
| | - Michael Mederos Y Schnitzler
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany;; DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany.
| | - Ursula Storch
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany;; Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, 80336 Munich, Germany.
| |
Collapse
|
22
|
Han S, Peng S, Vance J, Tran K, Do N, Bui N, Gui Z, Wang S. Structural dynamics determine voltage and pH gating in human voltage-gated proton channel. eLife 2022; 11:73093. [PMID: 35244539 PMCID: PMC8926398 DOI: 10.7554/elife.73093] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 02/24/2022] [Indexed: 11/25/2022] Open
Abstract
Voltage-gated proton (Hv) channels are standalone voltage sensors without separate ion conductive pores. They are gated by both voltage and transmembrane proton gradient (i.e., ∆pH), serving as acid extruders in most cells. Like the canonical voltage sensors, Hv channels are a bundle of four helices (named S1 –S4), with the S4 segment carrying three positively charged Arg residues. Extensive structural and electrophysiological studies on voltage-gated ion channels, in general, agree on an outwards movement of the S4 segment upon activating voltage, but the real-time conformational transitions are still unattainable. With purified human voltage-gated proton (hHv1) channels reconstituted in liposomes, we have examined its conformational dynamics, including the S4 segment at different voltage and pHs using single-molecule fluorescence resonance energy transfer (smFRET). Here, we provide the first glimpse of real-time conformational trajectories of the hHv1 voltage sensor and show that both voltage and pH gradient shift the conformational dynamics of the S4 segment to control channel gating. Our results indicate that the S4 segment transits among three major conformational states and only the transitions between the inward and outward conformations are highly dependent on voltage and pH. Altogether, we propose a kinetic model that explains the mechanisms underlying voltage and pH gating in Hv channels, which may also serve as a general framework for understanding the voltage sensing and gating in other voltage-gated ion channels.
Collapse
Affiliation(s)
- Shuo Han
- Department of Cell Biology and Biophysics, University of Missouri-Kansas City, Kansas City, United States
| | - Sophia Peng
- Department of Cell Biology and Biophysics, University of Missouri-Kansas City, Kansas City, United States
| | - Joshua Vance
- Department of Cell Biology and Biophysics, University of Missouri-Kansas City, Kansas City, United States
| | - Kimberly Tran
- Department of Cell Biology and Biophysics, University of Missouri-Kansas City, Kansas City, United States
| | - Nhu Do
- Department of Cell Biology and Biophysics, University of Missouri-Kansas City, Kansas City, United States
| | - Nauy Bui
- Department of Cell Biology and Biophysics, University of Missouri-Kansas City, Kansas City, United States
| | - Zhenhua Gui
- Department of Cell Biology and Biophysics, University of Missouri-Kansas City, Kansas City, United States
| | - Shizhen Wang
- Department of Cell Biology and Biophysics, University of Missouri-Kansas City, Kansas City, United States
| |
Collapse
|
23
|
Ma J, Gao X, Li Y, DeCoursey TE, Shull GE, Wang HS. The HVCN1 voltage-gated proton channel contributes to pH regulation in canine ventricular myocytes. J Physiol 2022; 600:2089-2103. [PMID: 35244217 PMCID: PMC9058222 DOI: 10.1113/jp282126] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/24/2022] [Indexed: 11/09/2022] Open
Abstract
KEY POINTS Intracellular pH (pHi ) regulation is crucial for cardiac function, as acidification depresses contractility and causes arrhythmias. H+ ions are generated in cardiomyocytes from metabolic processes and particularly from CO2 hydration, which has been shown to facilitate CO2 -venting from mitochondria. Currently, the NHE1 Na+ /H+ exchanger is viewed as the dominant H+ -extrusion mechanism in cardiac muscle. We show that the HVCN1 voltage-gated proton channel is present and functional in canine ventricular myocytes, and that HVCN1 and NHE1 both contribute to pHi regulation. HVCN1 provides an energetically-efficient mechanism of H+ -extrusion that would not cause Na+ -loading, which can cause pathology, and that could contribute to transport-mediated CO2 disposal. These results provide a major advance in our understanding of pHi regulation in cardiac muscle. ABSTRACT Regulation of intracellular pH (pHi ) in cardiomyocytes is crucial for cardiac function; however, currently known mechanisms for direct or indirect extrusion of acid from cardiomyocytes seem insufficient for energetically-efficient extrusion of the massive H+ loads generated under in vivo conditions. In cardiomyocytes, voltage-sensitive H+ channel activity mediated by the HVCN1 proton channel would be a highly efficient means of disposing of H+ , while avoiding Na+ -loading, as occurs during direct acid extrusion via Na+ /H+ exchange or indirect acid extrusion via Na+ -HCO3 - cotransport. PCR and immunoblotting demonstrated expression of HVCN1 mRNA and protein in canine heart. Patch clamp analysis of canine ventricular myocytes revealed a voltage-gated H+ current that was highly H+ -selective. The current was blocked by external Zn2+ and the HVCN1 blocker 5-chloro-2-guanidinobenzimidazole (ClGBI). Both the gating and Zn2+ blockade of the current were strongly influenced by the pH gradient across the membrane. All characteristics of the observed current were consistent with the known hallmarks of HVCN1-mediated H+ current. Inhibition of HVCN1 and the NHE1 Na+ /H+ exchanger, singly and in combination, showed that either mechanism is largely sufficient to maintain pHi in beating cardiomyocytes, but that inhibition of both activities causes rapid acidification. These results show that HVCN1 is expressed in canine ventricular myocytes and provides a major H+ -extrusion activity, with a capacity similar to that of NHE1. In the beating heart in vivo, this activity would allow Na+ -independent extrusion of H+ during each action potential and, when functionally coupled with anion transport mechanisms, could facilitate transport-mediated CO2 disposal. Abstract figure legend The HVCN1 proton channel is expressed in canine ventricular myocytes and contributes to H+ extrusion. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Xiaoqian Gao
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Yutian Li
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois, 60612, USA
| | - Gary E Shull
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| |
Collapse
|
24
|
Teng B, Kaplan JP, Liang Z, Krieger Z, Tu YH, Burendei B, Ward AB, Liman ER. Structural motifs for subtype-specific pH-sensitive gating of vertebrate otopetrin proton channels. eLife 2022; 11:77946. [PMID: 35920807 PMCID: PMC9348849 DOI: 10.7554/elife.77946] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/19/2022] [Indexed: 01/05/2023] Open
Abstract
Otopetrin (OTOP) channels are proton-selective ion channels conserved among vertebrates and invertebrates, with no structural similarity to other ion channels. There are three vertebrate OTOP channels (OTOP1, OTOP2, and OTOP3), of which one (OTOP1) functions as a sour taste receptor. Whether extracellular protons gate OTOP channels, in addition to permeating them, was not known. Here, we compare the functional properties of the three murine OTOP channels using patch-clamp recording and cytosolic pH microfluorimetry. We find that OTOP1 and OTOP3 are both steeply activated by extracellular protons, with thresholds of pHo <6.0 and 5.5, respectively, and kinetics that are pH-dependent. In contrast, OTOP2 channels are broadly active over a large pH range (pH 5 pH 10) and carry outward currents in response to extracellular alkalinization (>pH 9.0). Strikingly, we could change the pH-sensitive gating of OTOP2 and OTOP3 channels by swapping extracellular linkers that connect transmembrane domains. Swaps of extracellular linkers in the N domain, comprising transmembrane domains 1-6, tended to change the relative conductance at alkaline pH of chimeric channels, while swaps within the C domain, containing transmembrane domains 7-12, tended to change the rates of OTOP3 current activation. We conclude that members of the OTOP channel family are proton-gated (acid-sensitive) proton channels and that the gating apparatus is distributed across multiple extracellular regions within both the N and C domains of the channels. In addition to the taste system, OTOP channels are expressed in the vertebrate vestibular and digestive systems. The distinct gating properties we describe may allow them to subserve varying cell-type specific functions in these and other biological systems.
Collapse
Affiliation(s)
- Bochuan Teng
- Section of Neurobiology, Department of Biological Sciences, University of Southern CaliforniaLos AngelesUnited States,Program in Neuroscience, University of Southern CaliforniaLos AngelesUnited States
| | - Joshua P Kaplan
- Section of Neurobiology, Department of Biological Sciences, University of Southern CaliforniaLos AngelesUnited States,Program in Neuroscience, University of Southern CaliforniaLos AngelesUnited States
| | - Ziyu Liang
- Section of Neurobiology, Department of Biological Sciences, University of Southern CaliforniaLos AngelesUnited States,Program in Neuroscience, University of Southern CaliforniaLos AngelesUnited States
| | - Zachary Krieger
- Section of Neurobiology, Department of Biological Sciences, University of Southern CaliforniaLos AngelesUnited States
| | - Yu-Hsiang Tu
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaUnited States
| | - Batuujin Burendei
- Department of Integrative Structural and Computational Biology, The Scripps Research InstituteLa JollaUnited States
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research InstituteLa JollaUnited States
| | - Emily R Liman
- Section of Neurobiology, Department of Biological Sciences, University of Southern CaliforniaLos AngelesUnited States,Program in Neuroscience, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
25
|
Dietl P, Frick M. Channels and Transporters of the Pulmonary Lamellar Body in Health and Disease. Cells 2021; 11:45. [PMID: 35011607 PMCID: PMC8750383 DOI: 10.3390/cells11010045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023] Open
Abstract
The lamellar body (LB) of the alveolar type II (ATII) cell is a lysosome-related organelle (LRO) that contains surfactant, a complex mix of mainly lipids and specific surfactant proteins. The major function of surfactant in the lung is the reduction of surface tension and stabilization of alveoli during respiration. Its lack or deficiency may cause various forms of respiratory distress syndrome (RDS). Surfactant is also part of the innate immune system in the lung, defending the organism against air-borne pathogens. The limiting (organelle) membrane that encloses the LB contains various transporters that are in part responsible for translocating lipids and other organic material into the LB. On the other hand, this membrane contains ion transporters and channels that maintain a specific internal ion composition including the acidic pH of about 5. Furthermore, P2X4 receptors, ligand gated ion channels of the danger signal ATP, are expressed in the limiting LB membrane. They play a role in boosting surfactant secretion and fluid clearance. In this review, we discuss the functions of these transporting pathways of the LB, including possible roles in disease and as therapeutic targets, including viral infections such as SARS-CoV-2.
Collapse
Affiliation(s)
- Paul Dietl
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
26
|
Orts DJB, Arcisio-Miranda M. Cell glycosaminoglycans content modulates human voltage-gated proton channel (H V 1) gating. FEBS J 2021; 289:2593-2612. [PMID: 34800064 DOI: 10.1111/febs.16290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/01/2021] [Accepted: 11/19/2021] [Indexed: 11/28/2022]
Abstract
Voltage-gated proton channels (HV 1) have been found in many mammalian cells and play a crucial role in the immune system, male fertility, and cancer progression. Glycosaminoglycans play a significant role in various aspects of cell physiology, including the modulation of membrane receptors and ion channel function. We present here evidence that mechanosensitivity of the dimeric HV 1 channel transduce changes on cell membrane fluidity related to the defective biosynthesis of chondroitin sulfate and heparan sulfate in Chinese Hamster Ovary (CHO-745) cells into a leftward shift in the activation voltage dependence. This effect was accompanied by an increase in the H+ current, and an acceleration of the activation kinetics, under symmetrical or asymmetrical pH gradient (ΔpH) conditions. Similar gating alterations were evoked by two naturally occurring HV 1 N-terminal truncated isoforms expressed in wild-type CHO-K1 and CHO-745 cells. On three different monomeric HV 1 constructs, no alterations in the biophysical parameters were observed. Moreover, we have shown that HV 1 gating can be modulated by manipulating CHO-K1 cell membrane fluidity. Our results suggest that the defective biosynthesis of chondroitin sulfate and heparan sulfate on CHO-745 cell increases membrane fluidity and allosterically modulates the coupling between voltage- and ΔpH-sensing through the dimeric HV 1 channel.
Collapse
Affiliation(s)
- Diego J B Orts
- Departamento de Biofísica, Laboratório de Neurobiologia Estrutural e Funcional (LaNEF), Universidade Federal de São Paulo - UNIFESP, Brasil
| | - Manoel Arcisio-Miranda
- Departamento de Biofísica, Laboratório de Neurobiologia Estrutural e Funcional (LaNEF), Universidade Federal de São Paulo - UNIFESP, Brasil
| |
Collapse
|
27
|
Sokolov VS, Cherny VV, Ayuyan AG, DeCoursey TE. Analysis of an electrostatic mechanism for ΔpH dependent gating of the voltage-gated proton channel, H V1, supports a contribution of protons to gating charge. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2021; 1862:148480. [PMID: 34363792 PMCID: PMC8432343 DOI: 10.1016/j.bbabio.2021.148480] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/23/2021] [Accepted: 08/02/2021] [Indexed: 11/23/2022]
Abstract
Voltage-gated proton channels (HV1) resemble the voltage-sensing domain of other voltage-gated ion channels, but differ in containing the conduction pathway. Essential to the functions of HV1 channels in many cells and species is a unique feature called ΔpH dependent gating. The pH on both sides of the membrane strictly regulates the voltage range of channel opening, generally resulting in exclusively outward proton current. Two types of mechanisms could produce ΔpH dependent gating. The "countercharge" mechanism proposes that protons destabilize salt bridges between amino acids in the protein that stabilize specific gating configurations (closed or open). An "electrostatic" mechanism proposes that protons bound to the channel alter the electrical field sensed by the protein. Obligatory proton binding within the membrane electrical field would contribute to measured gating charge. Estimations on the basis of the electrostatic model explain ΔpH dependent gating, but quantitative modeling requires calculations of the electric field inside the protein which, in turn, requires knowledge of its structure. We conclude that both mechanisms operate and contribute to ΔpH dependent gating of HV1.
Collapse
Affiliation(s)
- Valerij S Sokolov
- Frumkin Institute of Physical Chemistry and Electrochemistry of Russian Academy of Sciences, Moscow 119071, Russia
| | - Vladimir V Cherny
- Department of Physiology & Biophysics, Rush University, Chicago, IL 60612, USA
| | - Artem G Ayuyan
- Department of Physiology & Biophysics, Rush University, Chicago, IL 60612, USA
| | - Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, IL 60612, USA.
| |
Collapse
|
28
|
Droste A, Chaves G, Stein S, Trzmiel A, Schweizer M, Karl H, Musset B. Zinc accelerates respiratory burst termination in human PMN. Redox Biol 2021; 47:102133. [PMID: 34562872 PMCID: PMC8476447 DOI: 10.1016/j.redox.2021.102133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 11/25/2022] Open
Abstract
The respiratory burst of phagocytes is essential for human survival. Innate immune defence against pathogens relies strongly on reactive oxygen species (ROS) production by the NADPH oxidase (NOX2). ROS kill pathogens while the translocation of electrons across the plasma membrane via NOX2 depolarizes the cell. Simultaneously, protons are released into the cytosol. Here, we compare freshly isolated human polymorphonuclear leukocytes (PMN) to the granulocytes-like cell line PLB 985. We are recording ROS production while inhibiting the charge compensating and pH regulating voltage-gated proton channel (HV1). The data suggests that human PMN and the PLB 985 generate ROS via a general mechanism, consistent of NOX2 and HV1. Additionally, we advanced a mathematical model based on the biophysical properties of NOX2 and HV1. Our results strongly suggest the essential interconnection of HV1 and NOX2 during the respiratory burst of phagocytes. Zinc chelation during the time course of the experiments postulates that zinc leads to an irreversible termination of the respiratory burst over time. Flow cytometry shows cell death triggered by high zinc concentrations and PMA. Our data might help to elucidate the complex interaction of proteins during the respiratory burst and contribute to decipher its termination.
Collapse
Affiliation(s)
- Annika Droste
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany; Department of Gynecology and Obstetrics, Johannes Gutenberg University, Mainz, Germany
| | - Gustavo Chaves
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
| | - Stefan Stein
- Flow Cytometry Unit, Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Annette Trzmiel
- Flow Cytometry Unit, Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Matthias Schweizer
- Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| | - Hubert Karl
- Department efi, Technische Hochschule Nürnberg Georg Simon Ohm, Nuremberg, Germany
| | - Boris Musset
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany; Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
29
|
Cherny VV, Musset B, Morgan D, Thomas S, Smith SME, DeCoursey TE. Engineered high-affinity zinc binding site reveals gating configurations of a human proton channel. J Gen Physiol 2021; 152:152076. [PMID: 32902579 PMCID: PMC7537347 DOI: 10.1085/jgp.202012664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/10/2020] [Indexed: 11/23/2022] Open
Abstract
The voltage-gated proton channel (HV1) is a voltage sensor that also conducts protons. The singular ability of protons to penetrate proteins complicates distinguishing closed and open channels. When we replaced valine with histidine at position 116 in the external vestibule of hHV1, current was potently inhibited by externally applied Zn2+ in a construct lacking the two His that bind Zn2+ in WT channels. High-affinity binding with profound effects at 10 nM Zn2+ at pHo 7 suggests additional groups contribute. We hypothesized that Asp185, which faces position 116 in our closed-state model, contributes to Zn2+ chelation. Confirming this prediction, V116H/D185N abolished Zn2+ binding. Studied in a C-terminal truncated monomeric construct, V116H channels activated rapidly. Anomalously, Zn2+ slowed activation, producing a time constant independent of both voltage and Zn2+ concentration. We hypothesized that slow turn-on of H+ current in the presence of Zn2+ reflects the rate of Zn2+ unbinding from the channel, analogous to drug-receptor dissociation reactions. This behavior in turn suggests that the affinity for Zn2+ is greater in the closed state of hHV1. Supporting this hypothesis, pulse pairs revealed a rapid component of activation whose amplitude decreased after longer intervals at negative voltages as closed channels bound Zn2+. The lower affinity of Zn2+ in open channels is consistent with the idea that structural rearrangements within the transmembrane region bring Arg205 near position 116, electrostatically expelling Zn2+. This phenomenon provides direct evidence that Asp185 opposes position 116 in closed channels and that Arg205 moves between them when the channel opens.
Collapse
Affiliation(s)
| | - Boris Musset
- Institut für Physiologie und Pathophysiologie, Paracelsus Medizinische Privatuniversität, Nürnberg, Germany
| | - Deri Morgan
- Department of Physiology & Biophysics, Rush University, Chicago IL
| | - Sarah Thomas
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - Susan M E Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | | |
Collapse
|
30
|
Rangel-Yescas G, Cervantes C, Cervantes-Rocha MA, Suárez-Delgado E, Banaszak AT, Maldonado E, Ramsey IS, Rosenbaum T, Islas LD. Discovery and characterization of H v1-type proton channels in reef-building corals. eLife 2021; 10:e69248. [PMID: 34355697 PMCID: PMC8346283 DOI: 10.7554/elife.69248] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
Voltage-dependent proton-permeable channels are membrane proteins mediating a number of important physiological functions. Here we report the presence of a gene encoding Hv1 voltage-dependent, proton-permeable channels in two species of reef-building corals. We performed a characterization of their biophysical properties and found that these channels are fast-activating and modulated by the pH gradient in a distinct manner. The biophysical properties of these novel channels make them interesting model systems. We have also developed an allosteric gating model that provides mechanistic insight into the modulation of voltage-dependence by protons. This work also represents the first functional characterization of any ion channel in scleractinian corals. We discuss the implications of the presence of these channels in the membranes of coral cells in the calcification and pH-regulation processes and possible consequences of ocean acidification related to the function of these channels.
Collapse
Affiliation(s)
- Gisela Rangel-Yescas
- Departmento de Fisiología, Facultad of Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Cecilia Cervantes
- Departmento de Fisiología, Facultad of Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel A Cervantes-Rocha
- Departmento de Fisiología, Facultad of Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Esteban Suárez-Delgado
- Departmento de Fisiología, Facultad of Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Anastazia T Banaszak
- Unidad Académica de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Puerto Morelos, Mexico
| | - Ernesto Maldonado
- EvoDevo Research Group, Unidad Académica de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Puerto Morelos, Mexico
| | - Ian Scott Ramsey
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, United States
| | - Tamara Rosenbaum
- Departmento of Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Leon D Islas
- Departmento de Fisiología, Facultad of Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
31
|
Carmona EM, Fernandez M, Alvear-Arias JJ, Neely A, Larsson HP, Alvarez O, Garate JA, Latorre R, Gonzalez C. The voltage sensor is responsible for ΔpH dependence in H v1 channels. Proc Natl Acad Sci U S A 2021; 118:e2025556118. [PMID: 33941706 PMCID: PMC8126849 DOI: 10.1073/pnas.2025556118] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The dissipation of acute acid loads by the voltage-gated proton channel (Hv1) relies on regulating the channel's open probability by the voltage and the ΔpH across the membrane (ΔpH = pHex - pHin). Using monomeric Ciona-Hv1, we asked whether ΔpH-dependent gating is produced during the voltage sensor activation or permeation pathway opening. A leftward shift of the conductance-voltage (G-V) curve was produced at higher ΔpH values in the monomeric channel. Next, we measured the voltage sensor pH dependence in the absence of a functional permeation pathway by recording gating currents in the monomeric nonconducting D160N mutant. Increasing the ΔpH leftward shifted the gating charge-voltage (Q-V) curve, demonstrating that the ΔpH-dependent gating in Hv1 arises by modulating its voltage sensor. We fitted our data to a model that explicitly supposes the Hv1 voltage sensor free energy is a function of both the proton chemical and the electrical potential. The parameters obtained showed that around 60% of the free energy stored in the ΔpH is coupled to the Hv1 voltage sensor activation. Our results suggest that the molecular mechanism underlying the Hv1 ΔpH dependence is produced by protons, which alter the free-energy landscape around the voltage sensor domain. We propose that this alteration is produced by accessibility changes of the protons in the Hv1 voltage sensor during activation.
Collapse
Affiliation(s)
- Emerson M Carmona
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
| | - Miguel Fernandez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
| | - Juan J Alvear-Arias
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
| | - Alan Neely
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
| | - H Peter Larsson
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Osvaldo Alvarez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, 7800003 Santiago, Chile
| | - Jose Antonio Garate
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
| | - Carlos Gonzalez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile;
- Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX 79430
- Facultad de Ciencias Químicas, Universidad Juarez del Estado de Durango, Durango 34000, México
| |
Collapse
|
32
|
Zajac M, Dreano E, Edwards A, Planelles G, Sermet-Gaudelus I. Airway Surface Liquid pH Regulation in Airway Epithelium Current Understandings and Gaps in Knowledge. Int J Mol Sci 2021; 22:3384. [PMID: 33806154 PMCID: PMC8037888 DOI: 10.3390/ijms22073384] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022] Open
Abstract
Knowledge on the mechanisms of acid and base secretion in airways has progressed recently. The aim of this review is to summarize the known mechanisms of airway surface liquid (ASL) pH regulation and their implication in lung diseases. Normal ASL is slightly acidic relative to the interstitium, and defects in ASL pH regulation are associated with various respiratory diseases, such as cystic fibrosis. Basolateral bicarbonate (HCO3-) entry occurs via the electrogenic, coupled transport of sodium (Na+) and HCO3-, and, together with carbonic anhydrase enzymatic activity, provides HCO3- for apical secretion. The latter mainly involves CFTR, the apical chloride/bicarbonate exchanger pendrin and paracellular transport. Proton (H+) secretion into ASL is crucial to maintain its relative acidity compared to the blood. This is enabled by H+ apical secretion, mainly involving H+/K+ ATPase and vacuolar H+-ATPase that carry H+ against the electrochemical potential gradient. Paracellular HCO3- transport, the direction of which depends on the ASL pH value, acts as an ASL protective buffering mechanism. How the transepithelial transport of H+ and HCO3- is coordinated to tightly regulate ASL pH remains poorly understood, and should be the focus of new studies.
Collapse
Affiliation(s)
- Miroslaw Zajac
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| | - Elise Dreano
- Institut Necker Enfants Malades, INSERM U1151, 75015 Paris, France;
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
| | - Aurelie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA;
| | - Gabrielle Planelles
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
- Laboratoire de Physiologie rénale et Tubulopathies, CNRS ERL 8228, 75006 Paris, France
| | - Isabelle Sermet-Gaudelus
- Institut Necker Enfants Malades, INSERM U1151, 75015 Paris, France;
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
- Centre de Référence Maladies Rares, Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, 75015 Paris, France
- Clinical Trial Network, European Cystic Fibrosis Society, BT2 Belfast, Ireland
- European Respiratory Network Lung, 75006 Paris, France
| |
Collapse
|
33
|
Voltage and pH difference across the membrane control the S4 voltage-sensor motion of the Hv1 proton channel. Sci Rep 2020; 10:21293. [PMID: 33277511 PMCID: PMC7718894 DOI: 10.1038/s41598-020-77986-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022] Open
Abstract
The voltage-gated proton channel Hv1 is expressed in a variety of cells, including macrophages, sperm, and lung epithelial cells. Hv1 is gated by both the membrane potential and the difference between the intra- and extracellular pH (ΔpH). The coupling of voltage- and ∆pH-sensing is such that Hv1 opens only when the electrochemical proton gradient is outwardly directed. However, the molecular mechanism of this coupling is not known. Here, we investigate the coupling between voltage- and ΔpH-sensing of Ciona intestinalis proton channel (ciHv1) using patch-clamp fluorometry (PCF) and proton uncaging. We show that changes in ΔpH can induce conformational changes of the S4 voltage sensor. Our results are consistent with the idea that S4 can detect both voltage and ΔpH.
Collapse
|
34
|
Abstract
Cherny and coworkers use zinc ion as a probe to identify different conformational states of voltage-gated proton (Hv1) channels.
Collapse
Affiliation(s)
- H Peter Larsson
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
35
|
Smith RY, Morgan D, Sharma L, Cherny VV, Tidswell N, Molo MW, DeCoursey TE. Voltage-gated proton channels exist in the plasma membrane of human oocytes. Hum Reprod 2020; 34:1974-1983. [PMID: 31633762 DOI: 10.1093/humrep/dez178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/10/2019] [Indexed: 12/25/2022] Open
Abstract
STUDY QUESTION Do human oocytes express voltage-gated proton channels? SUMMARY ANSWER Human oocytes exhibit voltage-gated proton currents. WHAT IS KNOWN ALREADY Voltage-gated proton currents have been reported in human sperm, where they contribute to capacitation and motility. No such studies of human oocytes exist. STUDY DESIGN, SIZE, DURATION Voltage-clamp studies were undertaken using entire oocytes and vesicles derived from oocytes and in excised patches of membrane from oocytes. PARTICIPANTS/MATERIALS, SETTING, METHODS Frozen, thawed human metaphase II oocytes were obtained from material donated to the gamete repository at the Rush Center for Advanced Reproductive Care. Prior to patch clamping, oocytes were warmed and equilibrated. Formation of an electrically tight seal requires exposing bare oolemma. Sections of the zona pellucida (ZP) were removed using a laser, followed by repeated pipetting, to further separate the oocyte from the ZP. Patch-clamp studies were performed using the whole-cell configuration on oocytes or vesicles derived from oocytes, and using inside-out patches of membrane, under conditions optimized to detect voltage-gated proton currents. MAIN RESULTS AND THE ROLE OF CHANCE Proton currents are present at significant levels in human oocytes where they exhibit properties similar to those reported in other human cells, as well as those in heterologous expression systems transfected with the HVCN1 gene that codes for the voltage-gated proton channel. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Human oocytes are large cells, which limits our ability to control the intracellular solution. Subtle effects of cryopreservation by vitrification and subsequent warming on properties of HVCN1, the HVCN1 gene product, cannot be ruled out. WIDER IMPLICATIONS OF THE FINDINGS Possible functions for voltage-gated proton channels in human oocytes may now be contemplated. STUDY FUNDING/COMPETING INTEREST(S) NIH R35GM126902 (TED), Bears Care (DM). No competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- R Ya Smith
- Rush Center for Advanced Reproductive Care, Department of Obstetrics and Gynecology, Rush University Medical Center, Chicago, IL 60612, USA
| | - D Morgan
- Department of Physiology & Biophysics, Rush University, Chicago, IL 60612, USA
| | - L Sharma
- Rush Center for Advanced Reproductive Care, Department of Obstetrics and Gynecology, Rush University Medical Center, Chicago, IL 60612, USA
| | - V V Cherny
- Department of Physiology & Biophysics, Rush University, Chicago, IL 60612, USA
| | - N Tidswell
- Rush Center for Advanced Reproductive Care, Department of Obstetrics and Gynecology, Rush University Medical Center, Chicago, IL 60612, USA
| | - M W Molo
- Rush Center for Advanced Reproductive Care, Department of Obstetrics and Gynecology, Rush University Medical Center, Chicago, IL 60612, USA
| | - T E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, IL 60612, USA
| |
Collapse
|
36
|
Bare DJ, Cherny VV, DeCoursey TE, Abukhdeir AM, Morgan D. Expression and function of voltage gated proton channels (Hv1) in MDA-MB-231 cells. PLoS One 2020; 15:e0227522. [PMID: 32374759 PMCID: PMC7202653 DOI: 10.1371/journal.pone.0227522] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/09/2020] [Indexed: 02/07/2023] Open
Abstract
Expression of the voltage gated proton channel (Hv1) as identified by immunocytochemistry has been reported previously in breast cancer tissue. Increased expression of HV1 was correlated with poor prognosis and decreased overall and disease-free survival but the mechanism of its involvement in the disease is unknown. Here we present electrophysiological recordings of HV1 channel activity, confirming its presence and function in the plasma membrane of a breast cancer cell line, MDA-MB-231. With western blotting we identify significant levels of HV1 expression in 3 out of 8 “triple negative” breast cancer cell lines (estrogen, progesterone, and HER2 receptor expression negative). We examine the function of HV1 in breast cancer using MDA-MB-231 cells as a model by suppressing the expression of HV1 using shRNA (knock-down; KD) and by eliminating HV1 using CRISPR/Cas9 gene editing (knock-out; KO). Surprisingly, these two approaches produced incongruous effects. Knock-down of HV1 using shRNA resulted in slower cell migration in a scratch assay and a significant reduction in H2O2 release. In contrast, HV1 Knock-out cells did not show reduced migration or H2O2 release. HV1 KO but not KD cells showed an increased glycolytic rate accompanied by an increase in p-AKT (phospho-AKT, Ser473) activity. The expression of CD171/LCAM-1, an adhesion molecule and prognostic indicator for breast cancer, was reduced in HV1 KO cells. When we compared MDA-MB-231 xenograft growth rates in immunocompromised mice, tumors from HV1 KO cells grew less than WT in mass, with lower staining for the Ki-67 marker for cell proliferation rate. Therefore, deletion of HV1 expression in MDA-MB-231 cells limits tumor growth rate. The limited growth thus appears to be independent of oxidant production by NADPH oxidase molecules and to be mediated by cell adhesion molecules. Although HV1 KO and KD affect certain cellular mechanisms differently, both implicate HV1-mediated pathways for control of tumor growth in the MDA-MB-231 cell line.
Collapse
Affiliation(s)
- Dan J Bare
- Department of Physiology & Biophysics, Rush University, Chicago, IL, United States of America
| | - Vladimir V Cherny
- Department of Physiology & Biophysics, Rush University, Chicago, IL, United States of America
| | - Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, IL, United States of America
| | - Abde M Abukhdeir
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States of America
| | - Deri Morgan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, MO, United States of America
| |
Collapse
|
37
|
Yeste M, Llavanera M, Mateo-Otero Y, Catalán J, Bonet S, Pinart E. HVCN1 Channels Are Relevant for the Maintenance of Sperm Motility During In Vitro Capacitation of Pig Spermatozoa. Int J Mol Sci 2020; 21:ijms21093255. [PMID: 32375375 PMCID: PMC7246839 DOI: 10.3390/ijms21093255] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022] Open
Abstract
The objective of the present study was to determine the physiological role of voltage-gated hydrogen channels 1 (HVCN1 channels) during in vitro capacitation of pig spermatozoa. Sperm samples from 20 boars were incubated in capacitating medium for 300 minutes (min) in the presence of 2-guanidino benzimidazole (2-GBI), a specific HVCN1-channel blocker, added either at 0 min or after 240 min of incubation. Control samples were incubated in capacitating medium without the inhibitor. In all samples, acrosomal exocytosis was triggered with progesterone after 240 min of incubation. Sperm viability, sperm motility and kinematics, acrosomal exocytosis, membrane lipid disorder, intracellular calcium levels and mitochondrial membrane potential were evaluated after 0, 60, 120, 180, 240, 250, 270 and 300 min of incubation. While HVCN1-blockage resulted in altered sperm viability, sperm motility and kinematics and reduced mitochondrial membrane potential as compared to control samples, at any blocker concentration and incubation time, it had a non-significant effect on intracellular Ca2+ levels determined through Fluo3-staining. The effects on acrosomal exocytosis were only significant in blocked samples at 0 min, and were associated with increased membrane lipid disorder and Ca2+ levels of the sperm head determined through Rhod5-staining. In conclusion, HVCN1 channels play a crucial role in the modulation of sperm motility and kinematics, and in Ca2+ entrance to the sperm head.
Collapse
Affiliation(s)
- Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (Y.M.-O); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, E-17003 Girona, Spain
| | - Marc Llavanera
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (Y.M.-O); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, E-17003 Girona, Spain
| | - Yentel Mateo-Otero
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (Y.M.-O); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, E-17003 Girona, Spain
| | - Jaime Catalán
- Unit of Animal Reproduction, Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, Autonomous University of Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain;
| | - Sergi Bonet
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (Y.M.-O); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, E-17003 Girona, Spain
| | - Elisabeth Pinart
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain; (M.Y.); (M.L.); (Y.M.-O); (S.B.)
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, E-17003 Girona, Spain
- Correspondence: ; Tel.: +34-972-419-514
| |
Collapse
|
38
|
The voltage-gated proton channel hHv1 is functionally expressed in human chorion-derived mesenchymal stem cells. Sci Rep 2020; 10:7100. [PMID: 32346069 PMCID: PMC7188850 DOI: 10.1038/s41598-020-63517-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/01/2020] [Indexed: 01/08/2023] Open
Abstract
The voltage-gated proton channel Hv1 is widely expressed, among others, in immune and cancer cells, it provides an efficient cytosolic H+extrusion mechanism and regulates vital functions such as oxidative burst, migration and proliferation. Here we demonstrate the presence of human Hv1 (hHv1) in the placenta/chorion-derived mesenchymal stem cells (cMSCs) using RT-PCR. The voltage- and pH-dependent gating of the current is similar to that of hHv1 expressed in cell lines and that the current is blocked by 5-chloro-2-guanidinobenzimidazole (ClGBI) and activated by arachidonic acid (AA). Inhibition of hHv1 by ClGBI significantly decreases mineral matrix production of cMSCs induced by conditions mimicking physiological or pathological (inorganic phosphate, Pi) induction of osteogenesis. Wound healing assay and single cell motility analysis show that ClGBI significantly inhibits the migration of cMSCs. Thus, seminal functions of cMSCs are modulated by hHv1 which makes this channel as an attractive target for controlling advantages/disadvantages of MSCs therapy.
Collapse
|
39
|
Guidelli R, Becucci L, Aloisi G. Role of the time dependence of Boltzmann open probability in voltage-gated proton channels. Bioelectrochemistry 2020; 134:107520. [PMID: 32279034 DOI: 10.1016/j.bioelechem.2020.107520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 11/28/2022]
Abstract
The modeling and simulation of experimental families of current-time (I-t) curves of dimeric voltage-gated proton channels and of proton-conducting voltage sensing domains (VSDs) with a minimum of free parameters requires the movement of protons to be controlled by the rate of increase of the Boltzmann open probability p over time in passing from the holding to the depolarizing potential. Families of I-t curves of protomers and proton-conducting VSDs can be satisfactorily fitted by the use of a single free parameter expressing the rate constant kp for the increase of p over time. Families of I-t curves of dimeric Hv1 channels can be fitted by a model that assumes an initial proton current I1 flowing along the two monomeric units, while they are still operating separately; I1 is gradually replaced by a slower and more potential-dependent current I2 flowing when the two monomers start operating jointly under the control of the coiled-coil domain. Here too, p is assumed to increase over time with a rate constant kp that doubles in passing from I1 to I2, with fit requiring three free parameters. Chord conductance yields erroneously high gating charges when fitted by the Boltzmann function, differently from slope conductance.
Collapse
Affiliation(s)
- Rolando Guidelli
- Department of Chemistry "Ugo Schiff", Florence University, Via della Lastruccia 3, 50019 Sesto Fiorentino (Firenze), Italy.
| | - Lucia Becucci
- Department of Chemistry "Ugo Schiff", Florence University, Via della Lastruccia 3, 50019 Sesto Fiorentino (Firenze), Italy
| | - Giovanni Aloisi
- Department of Chemistry "Ugo Schiff", Florence University, Via della Lastruccia 3, 50019 Sesto Fiorentino (Firenze), Italy
| |
Collapse
|
40
|
Chaves G, Bungert-Plümke S, Franzen A, Mahorivska I, Musset B. Zinc modulation of proton currents in a new voltage-gated proton channel suggests a mechanism of inhibition. FEBS J 2020; 287:4996-5018. [PMID: 32160407 PMCID: PMC7754295 DOI: 10.1111/febs.15291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/10/2020] [Accepted: 03/10/2020] [Indexed: 02/03/2023]
Abstract
The HV1 voltage‐gated proton (HV1) channel is a key component of the cellular proton extrusion machinery and is pivotal for charge compensation during the respiratory burst of phagocytes. The best‐described physiological inhibitor of HV1 is Zn2+. Externally applied ZnCl2 drastically reduces proton currents reportedly recorded in Homo sapiens, Rattus norvegicus, Mus musculus, Oryctolagus cuniculus, Rana esculenta, Helix aspersa, Ciona intestinalis, Coccolithus pelagicus, Emiliania huxleyi, Danio rerio, Helisoma trivolvis, and Lingulodinium polyedrum, but with considerable species variability. Here, we report the effects of Zn2+ and Cd2+ on HV1 from Nicoletia phytophila, NpHV1. We introduced mutations at potential Zn2+ coordination sites and measured Zn2+ inhibition in different extracellular pH, with Zn2+ concentrations up to 1000 μm. Zn2+ inhibition in NpHV1 was quantified by the slowing of the activation time constant and a positive shift of the conductance–voltage curve. Replacing aspartate in the S3‐S4 loop with histidine (D145H) enhanced both the slowing of activation kinetics and the shift in the voltage–conductance curve, such that Zn2+ inhibition closely resembled that of the human channel. Histidine is much more effective than aspartate in coordinating Zn2+ in the S3‐S4 linker. A simple Hodgkin Huxley model of NpHV1 suggests a decrease in the opening rate if it is inhibited by zinc or cadmium. Limiting slope measurements and high‐resolution clear native gel electrophoresis (hrCNE) confirmed that NpHV1 functions as a dimer. The data support the hypothesis that zinc is coordinated in between the dimer instead of the monomer. Zinc coordination sites may be potential targets for drug development.
Collapse
Affiliation(s)
- Gustavo Chaves
- Institut für Physiologie und Pathophysiologie, Paracelsus Universität Salzburg Standort Nürnberg, Nuremberg, Germany
| | - Stefanie Bungert-Plümke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4) Forschungszentrum Jülich, Jülich, Germany
| | - Arne Franzen
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4) Forschungszentrum Jülich, Jülich, Germany
| | - Iryna Mahorivska
- Institut für Physiologie und Pathophysiologie, Paracelsus Universität Salzburg Standort Nürnberg, Nuremberg, Germany
| | - Boris Musset
- Institut für Physiologie und Pathophysiologie, Paracelsus Universität Salzburg Standort Nürnberg, Nuremberg, Germany
| |
Collapse
|
41
|
Tang D, Yang Y, Xiao Z, Xu J, Yang Q, Dai H, Liang S, Tang C, Dong H, Liu Z. Scorpion toxin inhibits the voltage-gated proton channel using a Zn 2+ -like long-range conformational coupling mechanism. Br J Pharmacol 2020; 177:2351-2364. [PMID: 31975366 DOI: 10.1111/bph.14984] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Blocking the voltage-gated proton channel HV 1 is a promising strategy for the treatment of diseases like ischaemia stroke and cancer. However, few HV 1 channel antagonists have been reported. Here, we have identified a novel HV 1 channel antagonist from scorpion venom and have elucidated its action mechanism. EXPERIMENTAL APPROACH HV 1 and NaV channels were heterologously expressed in mammalian cell lines and their currents recorded using whole-cell patch clamp. Site-directed mutagenesis was used to generate mutants. Toxins were recombinantly produced in Escherichia coli. AGAP/W38F-HV 1 interaction was modelled by molecular dynamics simulations. KEY RESULTS The scorpion toxin AGAP (anti-tumour analgesic peptide) potently inhibited HV 1 currents. One AGAP mutant has reduced NaV channel activity but intact HV 1 activity (AGAP/W38F). AGAP/W38F inhibited HV 1 channel activation by trapping its S4 voltage sensor in a deactivated state and inhibited HV 1 currents with less pH dependence than Zn2+ . Mutation analysis showed that the binding pockets of AGAP/W38F and Zn2+ in HV 1 channel partly overlapped (common sites are His140 and His193). The E153A mutation at the intracellular Coulombic network (ICN) in HV 1 channel markedly reduced AGAP/W38F inhibition, as observed for Zn2+ . Experimental data and MD simulations suggested that AGAP/W38F inhibited HV 1 channel using a Zn2+ -like long-range conformational coupling mechanism. CONCLUSION AND IMPLICATIONS Our results suggest that the Zn2+ binding pocket in HV 1 channel might be a hotspot for modulators and valuable for designing HV 1 channel ligands. Moreover, AGAP/W38F is a useful molecular probe to study HV 1 channel and a lead compound for drug development.
Collapse
Affiliation(s)
- Dongfang Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yuqin Yang
- Kuang Yaming Honors School, Nanjing University, Nanjing, China.,Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Zhen Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jiahui Xu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Qiuchu Yang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Han Dai
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Songping Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Hao Dong
- Kuang Yaming Honors School, Nanjing University, Nanjing, China.,Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
42
|
Proton channel blockers inhibit Duox activity independent of Hv1 effects. Redox Biol 2019; 28:101346. [PMID: 31678720 PMCID: PMC6920136 DOI: 10.1016/j.redox.2019.101346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/01/2019] [Accepted: 10/16/2019] [Indexed: 12/29/2022] Open
Abstract
The NADPH oxidase reaction produces protons. In the case of the NADPH oxidase, NOX2, activity depends on secretion of these protons and is inhibited by blockade of the voltage-gated proton channel (Hv1). Duox1 and Duox2 activities similarly produce intracellular protons but synthesize hydrogen peroxide directly instead of superoxide. Hv1 contributes to acid secretion in some epithelia that express Duox. To test the hypothesis that Duox activity is also sensitive to Hv1 channel blockers, Duox was assayed in the presence of either Zn2+ or 5-chloro-2-guanidinobenzimidazole (ClGBI). Both compounds inhibited Duox activity in normal human bronchial epithelial cells but with an IC50 over 10-fold higher than that reported for Hv1 (IC50 Zn2+ = 0.68 mM; IC50 ClGBI = 0.07–0.14 mM). Homogenized HEK293T cells expressing either Duox1 or Duox2 showed similar IC50 values for ClGBI suggesting these compounds inhibit the enzymes through alternate mechanisms independent of Hv1 proton secretion. Inclusion of superoxide dismutase did not restore Duox hydrogen peroxide synthesis. Addition of nigericin to eliminate any possible transmembrane pH gradients in intracellular membrane-localized Duox did not alter activity in HEK293T homogenates. Extracellular Zn2+ blocked intracellular Ca2+ increases needed for Duox activity. Together the data suggest that Duox enzyme activities in epithelia are inhibited by compounds that block Hv1 but inhibition occurs through Hv1-independent mechanisms and support the idea that Hv1 is not required for Duox activity. Hv1 proton channel inhibitors block Duox in differentiated bronchial epithelial cells. Zinc blocks Duox activity concurrently with reduction of calcium transients. ClGBI, an inhibitor of Hv1, blocks Duox activity in homogenates of cells lacking Hv1. In differentiated bronchial epithelia, Hv1 blockers did not alter intracellular pH. H+/K+ ATPase inhibition acidified cytoplasm but did not block Duox activity.
Collapse
|
43
|
De La Rosa V, Ramsey IS. Gating Currents in the Hv1 Proton Channel. Biophys J 2019; 114:2844-2854. [PMID: 29925021 DOI: 10.1016/j.bpj.2018.04.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/19/2018] [Accepted: 04/30/2018] [Indexed: 11/25/2022] Open
Abstract
The Hv1 proton channel shares striking structural homology with fourth transmembrane helical segment-type voltage-sensor (VS) domains but manifests distinctive functional properties, including a proton-selective "aqueous" conductance and allosteric control of voltage-dependent gating by changes in the transmembrane pH gradient. The mechanisms responsible for Hv1's functional properties remain poorly understood, in part because methods for measuring gating currents that directly report VS activation have not yet been described. Here, we describe an approach that allows robust and reproducible measurement of gating-associated charge movements in Hv1. Gating currents reveal that VS activation and proton-selective aqueous conductance opening are thermodynamically distinct steps in the Hv1 activation pathway and show that pH changes directly alter VS activation. The availability of an assay for gating currents in Hv1 may aid future efforts to elucidate the molecular mechanisms of gating cooperativity, pH-dependent modulation, and H+ selectivity in a model VS domain protein.
Collapse
Affiliation(s)
- Victor De La Rosa
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Ian Scott Ramsey
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
44
|
Hydrophobic gasket mutation produces gating pore currents in closed human voltage-gated proton channels. Proc Natl Acad Sci U S A 2019; 116:18951-18961. [PMID: 31462498 PMCID: PMC6754559 DOI: 10.1073/pnas.1905462116] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A large family of membrane proteins, voltage-gated ion channels, regulate a vast array of physiological functions in essentially all life forms. How these molecules sense membrane potential and respond by creating ionic conduction is incompletely understood. The voltage sensors of these channels contain a “hydrophobic gasket,” a ring of hydrophobic amino acids near the center of the membrane, separating internal and external aqueous solutions. Although voltage-gated proton channels, HV1, resemble voltage-sensing domains of other channels, they differ fundamentally. On depolarization, HV1 conducts protons, whereas other voltage sensors open a physically distinct pore. We identify Val109, Phe150, Val177, and Val178 as the hHV1 hydrophobic gasket. Replacement with less hydrophobic amino acids accelerated channel opening and caused proton-selective leak through closed channels. The hydrophobic gasket (HG), a ring of hydrophobic amino acids in the voltage-sensing domain of most voltage-gated ion channels, forms a constriction between internal and external aqueous vestibules. Cationic Arg or Lys side chains lining the S4 helix move through this “gating pore” when the channel opens. S4 movement may occur during gating of the human voltage-gated proton channel, hHV1, but proton current flows through the same pore in open channels. Here, we replaced putative HG residues with less hydrophobic residues or acidic Asp. Substitution of individuals, pairs, or all 3 HG positions did not impair proton selectivity. Evidently, the HG does not act as a secondary selectivity filter. However, 2 unexpected functions of the HG in HV1 were discovered. Mutating HG residues independently accelerated channel opening and compromised the closed state. Mutants exhibited open–closed gating, but strikingly, at negative voltages where “normal” gating produces a nonconducting closed state, the channel leaked protons. Closed-channel proton current was smaller than open-channel current and was inhibited by 10 μM Zn2+. Extreme hyperpolarization produced a deeper closed state through a weakly voltage-dependent transition. We functionally identify the HG as Val109, Phe150, Val177, and Val178, which play a critical and exclusive role in preventing H+ influx through closed channels. Molecular dynamics simulations revealed enhanced mobility of Arg208 in mutants exhibiting H+ leak. Mutation of HG residues produces gating pore currents reminiscent of several channelopathies.
Collapse
|
45
|
Boonamnaj P, Sompornpisut P. Effect of Ionization State on Voltage-Sensor Structure in Resting State of the Hv1 Channel. J Phys Chem B 2019; 123:2864-2873. [DOI: 10.1021/acs.jpcb.9b00634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Panisak Boonamnaj
- Center of Excellence in Computational Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pornthep Sompornpisut
- Center of Excellence in Computational Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
46
|
Carmona EM, Larsson HP, Neely A, Alvarez O, Latorre R, Gonzalez C. Gating charge displacement in a monomeric voltage-gated proton (H v1) channel. Proc Natl Acad Sci U S A 2018; 115:9240-9245. [PMID: 30127012 PMCID: PMC6140481 DOI: 10.1073/pnas.1809705115] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The voltage-gated proton (Hv1) channel, a voltage sensor and a conductive pore contained in one structural module, plays important roles in many physiological processes. Voltage sensor movements can be directly detected by measuring gating currents, and a detailed characterization of Hv1 charge displacements during channel activation can help to understand the function of this channel. We succeeded in detecting gating currents in the monomeric form of the Ciona-Hv1 channel. To decrease proton currents and better separate gating currents from ion currents, we used the low-conducting Hv1 mutant N264R. Isolated ON-gating currents decayed at increasing rates with increasing membrane depolarization, and the amount of gating charges displaced saturates at high voltages. These are two hallmarks of currents arising from the movement of charged elements within the boundaries of the cell membrane. The kinetic analysis of gating currents revealed a complex time course of the ON-gating current characterized by two peaks and a marked Cole-Moore effect. Both features argue that the voltage sensor undergoes several voltage-dependent conformational changes during activation. However, most of the charge is displaced in a single central transition. Upon voltage sensor activation, the charge is trapped, and only a fast component that carries a small percentage of the total charge is observed in the OFF. We hypothesize that trapping is due to the presence of the arginine side chain in position 264, which acts as a blocking ion. We conclude that the movement of the voltage sensor must proceed through at least five states to account for our experimental data satisfactorily.
Collapse
Affiliation(s)
- Emerson M Carmona
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
| | - H Peter Larsson
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136
| | - Alan Neely
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
| | - Osvaldo Alvarez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, 7800003 Santiago, Chile
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile;
| | - Carlos Gonzalez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, 2351319 Valparaíso, Chile;
| |
Collapse
|
47
|
Gating currents indicate complex gating of voltage-gated proton channels. Proc Natl Acad Sci U S A 2018; 115:9057-9059. [PMID: 30135099 DOI: 10.1073/pnas.1813013115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
48
|
Thomas S, Cherny VV, Morgan D, Artinian LR, Rehder V, Smith SME, DeCoursey TE. Exotic properties of a voltage-gated proton channel from the snail Helisoma trivolvis. J Gen Physiol 2018; 150:835-850. [PMID: 29743301 PMCID: PMC5987876 DOI: 10.1085/jgp.201711967] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/27/2018] [Indexed: 12/24/2022] Open
Abstract
Voltage-gated proton channels, HV1, were first reported in Helix aspersa snail neurons. These H+ channels open very rapidly, two to three orders of magnitude faster than mammalian HV1. Here we identify an HV1 gene in the snail Helisoma trivolvis and verify protein level expression by Western blotting of H. trivolvis brain lysate. Expressed in mammalian cells, HtHV1 currents in most respects resemble those described in other snails, including rapid activation, 476 times faster than hHV1 (human) at pHo 7, between 50 and 90 mV. In contrast to most HV1, activation of HtHV1 is exponential, suggesting first-order kinetics. However, the large gating charge of ∼5.5 e0 suggests that HtHV1 functions as a dimer, evidently with highly cooperative gating. HtHV1 opening is exquisitely sensitive to pHo, whereas closing is nearly independent of pHo Zn2+ and Cd2+ inhibit HtHV1 currents in the micromolar range, slowing activation, shifting the proton conductance-voltage (gH-V) relationship to more positive potentials, and lowering the maximum conductance. This is consistent with HtHV1 possessing three of the four amino acids that coordinate Zn2+ in mammalian HV1. All known HV1 exhibit ΔpH-dependent gating that results in a 40-mV shift of the gH-V relationship for a unit change in either pHo or pHi This property is crucial for all the functions of HV1 in many species and numerous human cells. The HtHV1 channel exhibits normal or supernormal pHo dependence, but weak pHi dependence. Under favorable conditions, this might result in the HtHV1 channel conducting inward currents and perhaps mediating a proton action potential. The anomalous ΔpH-dependent gating of HtHV1 channels suggests a structural basis for this important property, which is further explored in this issue (Cherny et al. 2018. J. Gen. Physiol. https://doi.org/10.1085/jgp.201711968).
Collapse
Affiliation(s)
- Sarah Thomas
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | | | - Deri Morgan
- Department of Physiology & Biophysics, Rush University, Chicago, IL
| | | | - Vincent Rehder
- Department of Biology, Georgia State University, Atlanta, GA
| | - Susan M E Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | | |
Collapse
|
49
|
Ratanayotha A, Kawai T, Higashijima SI, Okamura Y. Molecular and functional characterization of the voltage-gated proton channel in zebrafish neutrophils. Physiol Rep 2018; 5:5/15/e13345. [PMID: 28774948 PMCID: PMC5555884 DOI: 10.14814/phy2.13345] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 11/24/2022] Open
Abstract
Voltage‐gated proton channels (Hv1/VSOP) are expressed in various cells types, including phagocytes, and are involved in diverse physiological processes. Although hvcn1, the gene encoding Hv1, has been identified across a wide range of species, most of the knowledge about its physiological function and expression profile is limited to mammals. In this study, we investigated the basic properties of DrHv1, the Hv1 ortholog in zebrafish (Danio rerio) which is an excellent animal model owing to the transparency, as well as its functional expression in native cells. Electrophysiological analysis using a heterologous expression system confirmed the properties of a voltage‐gated proton channel are conserved in DrHv1 with differences in threshold and activation kinetics as compared to mouse (Mus musculus) Hv1 (mHv1). RT‐PCR analysis revealed that hvcn1 is expressed in zebrafish neutrophils, as is the case in mammals. Subsequent electrophysiological analysis confirmed the functional expression of DrHv1 in zebrafish neutrophils, which suggests Hv1 function in phagocytes is conserved among vertebrates. We also found that DrHv1 is comparatively resistant to extracellular Zn2+, which is a potent inhibitor of mammalian Hv1, and this phenomenon appears to reflect variation in the Zn2+‐coordinating residue (histidine) within the extracellular linker region in mammalian Hv1. Notably, the serum Zn2+ concentration is much higher in zebrafish than in mouse, raising the possibility that Zn2+ sensitivity was acquired in accordance with a change in the serum Zn2+ concentration. This study highlights the biological variation and importance of Hv1 in different animal species.
Collapse
Affiliation(s)
- Adisorn Ratanayotha
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takafumi Kawai
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shin-Ichi Higashijima
- Laboratory of Behavioral Neurobiology, Department of Biodesign Research, Okazaki Institute for Integrative Bioscience, Okazaki, Aichi, Japan
| | - Yasushi Okamura
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
50
|
Kigundu G, Cooper JL, Smith SME. H v 1 Proton Channels in Dinoflagellates: Not Just for Bioluminescence? J Eukaryot Microbiol 2018; 65:928-933. [PMID: 29698585 PMCID: PMC7167071 DOI: 10.1111/jeu.12627] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/11/2018] [Accepted: 04/15/2018] [Indexed: 01/08/2023]
Abstract
Bioluminescence in dinoflagellates is controlled by HV1 proton channels. Database searches of dinoflagellate transcriptomes and genomes yielded hits with sequence features diagnostic of all confirmed HV1, and show that HV1 is widely distributed in the dinoflagellate phylogeny including the basal species Oxyrrhis marina. Multiple sequence alignments followed by phylogenetic analysis revealed three major subfamilies of HV1 that do not correlate with presence of theca, autotrophy, geographic location, or bioluminescence. These data suggest that most dinoflagellates express a HV1 which has a function separate from bioluminescence. Sequence evidence also suggests that dinoflagellates can contain more than one HV1 gene.
Collapse
Affiliation(s)
- Gabriel Kigundu
- Department of Molecular and Cellular Biology, Kennesaw State University, 370 Paulding Avenue MD 1202, Kennesaw, Georgia, 30144
| | - Jennifer L Cooper
- Department of Molecular and Cellular Biology, Kennesaw State University, 370 Paulding Avenue MD 1202, Kennesaw, Georgia, 30144
| | - Susan M E Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, 370 Paulding Avenue MD 1202, Kennesaw, Georgia, 30144
| |
Collapse
|