1
|
Huang J, Pan X, Yan N. Structural biology and molecular pharmacology of voltage-gated ion channels. Nat Rev Mol Cell Biol 2024; 25:904-925. [PMID: 39103479 DOI: 10.1038/s41580-024-00763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 08/07/2024]
Abstract
Voltage-gated ion channels (VGICs), including those for Na+, Ca2+ and K+, selectively permeate ions across the cell membrane in response to changes in membrane potential, thus participating in physiological processes involving electrical signalling, such as neurotransmission, muscle contraction and hormone secretion. Aberrant function or dysregulation of VGICs is associated with a diversity of neurological, psychiatric, cardiovascular and muscular disorders, and approximately 10% of FDA-approved drugs directly target VGICs. Understanding the structure-function relationship of VGICs is crucial for our comprehension of their working mechanisms and role in diseases. In this Review, we discuss how advances in single-particle cryo-electron microscopy have afforded unprecedented structural insights into VGICs, especially on their interactions with clinical and investigational drugs. We present a comprehensive overview of the recent advances in the structural biology of VGICs, with a focus on how prototypical drugs and toxins modulate VGIC activities. We explore how these structures elucidate the molecular basis for drug actions, reveal novel pharmacological sites, and provide critical clues to future drug discovery.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Xiaojing Pan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
| | - Nieng Yan
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
2
|
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
|
3
|
Sağsöz ME, Sağlam B, Arslan K, Baştuğ T, Çavuş M, Puralı N. Structural, Functional and Molecular Dynamics Examination of a de novo cloned Otopetrin-like Proton Channel in crayfish. Cell Biochem Biophys 2024; 82:2029-2036. [PMID: 38811473 DOI: 10.1007/s12013-024-01310-z] [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] [Accepted: 05/11/2024] [Indexed: 05/31/2024]
Abstract
Proton channels play a crucial role in many biological functions, as they are responsible for the selective transport of protons across cell membranes. Recently, Otopetrins, a family of eukaryotic proton-selective ion channels, have attracted significant attention due to their diverse physiological roles. Despite the importance of Otopetrins, their structural and functional properties remain relatively unexplored. As a model organism, crayfish have been extensively studied to gain insights into the functioning of the nervous system. These studies cover a wide range of aspects, including the properties of individual neurons and behavioral science. However, studying the physiological systems of crayfish poses challenges for molecular research due to limited molecular sequence information available for these organisms. In the present work was identified an originally cloned mRNA, coding an Otopetrin like proton channel in the crayfish. The coded protein was modeled in silico and possible conduction mechanisms and pathways were revealed. A plasmid of the cloned mRNA was heterologously expressed in HEK293T cells. Functional experiments on transfected cells indicated that the expressed mRNA was coupled to proton conduction across the cell membrane.
Collapse
Affiliation(s)
- Mustafa Erdem Sağsöz
- Biophysics Department, Hacettepe University, Faculty of Medicine, Ankara, Türkiye
- Biophysics Department, Atatürk University, Faculty of Medicine, Erzurum, Türkiye
| | - Berk Sağlam
- Biophysics Department, Hacettepe University, Faculty of Medicine, Ankara, Türkiye
| | - Kaan Arslan
- Biophysics Department, Hacettepe University, Faculty of Medicine, Ankara, Türkiye
| | - Turgut Baştuğ
- Biophysics Department, Hacettepe University, Faculty of Medicine, Ankara, Türkiye
| | - Murat Çavuş
- Bozok University, Faculty of Education, Mathematics and Science Education, Yozgat, Türkiye
| | - Nuhan Puralı
- Biophysics Department, Hacettepe University, Faculty of Medicine, Ankara, Türkiye.
| |
Collapse
|
4
|
Piga M, Varga Z, Feher A, Papp F, Korpos E, Bangera KC, Frlan R, Ilaš J, Dernovšek J, Tomašič T, Zidar N. Identification of a Novel Structural Class of H V1 Inhibitors by Structure-Based Virtual Screening. J Chem Inf Model 2024; 64:4850-4862. [PMID: 38850237 PMCID: PMC11200261 DOI: 10.1021/acs.jcim.4c00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
The human voltage-gated proton channel, hHV1, is highly expressed in various cell types including macrophages, B lymphocytes, microglia, sperm cells and also in various cancer cells. Overexpression of HV1 has been shown to promote tumor formation by highly metastatic cancer cells, and has been associated with neuroinflammatory diseases, immune response disorders and infertility, suggesting a potential use of hHV1 inhibitors in numerous therapeutic areas. To identify compounds targeting this channel, we performed a structure-based virtual screening on an open structure of the human HV1 channel. Twenty selected virtual screening hits were tested on Chinese hamster ovary (CHO) cells transiently expressing hHV1, with compound 13 showing strong block of the proton current with an IC50 value of 8.5 μM. Biological evaluation of twenty-three additional analogs of 13 led to the discovery of six other compounds that blocked the proton current by more than 50% at 50 μM concentration. This allowed for an investigation of structure-activity relationships. The antiproliferative activity of the selected promising hHV1 inhibitors was investigated in the cell lines MDA-MB-231 and THP-1, where compound 13 inhibited growth with an IC50 value of 9.0 and 8.1 μM, respectively. The identification of a new structural class of HV1 inhibitors contributes to our understanding of the structural requirements for inhibition of this ion channel and opens up the possibility of investigating the role of HV1 inhibitors in various pathological conditions and in cancer therapy.
Collapse
Affiliation(s)
- Martina Piga
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Zoltan Varga
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Adam Feher
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Ferenc Papp
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Eva Korpos
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
- HUN-REN−UD
Cell Biology and Signaling Research Group, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Kavya C. Bangera
- Faculty
of Medicine, University of Debrecen, Egyetem tér 1, Debrecen H-4032, Hungary
| | - Rok Frlan
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Janez Ilaš
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Jaka Dernovšek
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Tihomir Tomašič
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Nace Zidar
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| |
Collapse
|
5
|
Theparambil SM, Begum G, Rose CR. pH regulating mechanisms of astrocytes: A critical component in physiology and disease of the brain. Cell Calcium 2024; 120:102882. [PMID: 38631162 PMCID: PMC11423562 DOI: 10.1016/j.ceca.2024.102882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024]
Abstract
Strict homeostatic control of pH in both intra- and extracellular compartments of the brain is fundamentally important, primarily due to the profound impact of free protons ([H+]) on neuronal activity and overall brain function. Astrocytes, crucial players in the homeostasis of various ions in the brain, actively regulate their intracellular [H+] (pHi) through multiple membrane transporters and carbonic anhydrases. The activation of astroglial pHi regulating mechanisms also leads to corresponding alterations in the acid-base status of the extracellular fluid. Notably, astrocyte pH regulators are modulated by various neuronal signals, suggesting their pivotal role in regulating brain acid-base balance in both health and disease. This review presents the mechanisms involved in pH regulation in astrocytes and discusses their potential impact on extracellular pH under physiological conditions and in brain disorders. Targeting astrocytic pH regulatory mechanisms represents a promising therapeutic approach for modulating brain acid-base balance in diseases, offering a potential critical contribution to neuroprotection.
Collapse
Affiliation(s)
- Shefeeq M Theparambil
- Faculty of Health and Medicine, Department of Biomedical and Life Sciences, Lancaster University, Lancaster, LA1 4YW, Lancaster, UK.
| | - Gulnaz Begum
- Department of Neurology, The Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| |
Collapse
|
6
|
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
|
7
|
Zhang L, Wu X, Cao X, Rao K, Hong L. Trp207 regulation of voltage-dependent activation of human H v1 proton channel. J Biol Chem 2024; 300:105674. [PMID: 38272234 PMCID: PMC10875263 DOI: 10.1016/j.jbc.2024.105674] [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: 11/21/2023] [Revised: 01/08/2024] [Accepted: 01/13/2024] [Indexed: 01/27/2024] Open
Abstract
In voltage-gated Na+ and K+ channels, the hydrophobicity of noncharged residues in the S4 helix has been shown to regulate the S4 movement underlying the process of voltage-sensing domain (VSD) activation. In voltage-gated proton channel Hv1, there is a bulky noncharged tryptophan residue located at the S4 transmembrane segment. This tryptophan remains entirely conserved across all Hv1 members but is not seen in other voltage-gated ion channels, indicating that the tryptophan contributes different roles in VSD activation. The conserved tryptophan of human voltage-gated proton channel Hv1 is Trp207 (W207). Here, we showed that W207 modifies human Hv1 voltage-dependent activation, and small residues replacement at position 207 strongly perturbs Hv1 channel opening and closing, and the size of the side chain instead of the hydrophobic group of W207 regulates the transition between closed and open states of the channel. We conclude that the large side chain of tryptophan controls the energy barrier during the Hv1 VSD transition.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Xin Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Xinyu Cao
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Khushi Rao
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, Illinois, USA; Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, USA.
| |
Collapse
|
8
|
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
|
9
|
Shen R, Roux B, Perozo E. Anionic omega currents from single countercharge mutants in the voltage-sensing domain of Ci-VSP. J Gen Physiol 2024; 156:e202213311. [PMID: 38019193 PMCID: PMC10686229 DOI: 10.1085/jgp.202213311] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/08/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023] Open
Abstract
The S4 segment of voltage-sensing domains (VSDs) directly responds to voltage changes by reorienting within the electric field as a permion. A narrow hydrophobic "gasket" or charge transfer center at the core of most VSDs focuses the electric field into a narrow region and catalyzes the sequential and reversible translocation of S4 positive gating charge residues across the electric field while preventing the permeation of physiological ions. Mutating specific S4 gating charges can cause ionic leak currents through the VSDs. These gating pores or omega currents play important pathophysiological roles in many diseases of excitability. Here, we show that mutating D129, a key countercharge residue in the Ciona intestinalis voltage-sensing phosphatase (Ci-VSP), leads to the generation of unique anionic omega currents. Neutralizing D129 causes a dramatic positive shift of activation, facilitates the formation of a continuous water path through the VSD, and creates a positive electrostatic potential landscape inside the VSD that contributes to its unique anionic selectivity. Increasing the population or dwell time of the conducting state by a high external pH or an engineered Cd2+ bridge markedly increases the current magnitude. Our findings uncover a new role of countercharge residues in the impermeable VSD of Ci-VSP and offer insights into mechanisms of the conduction of anionic omega currents linked to countercharge residue mutations.
Collapse
Affiliation(s)
- Rong Shen
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Benoît Roux
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Eduardo Perozo
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
10
|
Zhao C, Webster PD, De Angeli A, Tombola F. Mechanically-primed voltage-gated proton channels from angiosperm plants. Nat Commun 2023; 14:7515. [PMID: 37980353 PMCID: PMC10657467 DOI: 10.1038/s41467-023-43280-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 11/06/2023] [Indexed: 11/20/2023] Open
Abstract
Voltage-gated and mechanically-gated ion channels are distinct classes of membrane proteins that conduct ions across gated pores and are turned on by electrical or mechanical stimuli, respectively. Here, we describe an Hv channel (a.k.a voltage-dependent H+ channel) from the angiosperm plant A. thaliana that gates with a unique modality as it is turned on by an electrical stimulus only after exposure to a mechanical stimulus, a process that we call priming. The channel localizes in the vascular tissue and has homologs in vascular plants. We find that mechanical priming is not required for activation of non-angiosperm Hvs. Guided by AI-generated structural models of plant Hv homologs, we identify a set of residues playing a crucial role in mechanical priming. We propose that Hvs from angiosperm plants require priming because of a network of hydrophilic/charged residues that locks the channels in a silent resting conformation. Mechanical stimuli destabilize the network allowing the conduction pathway to turn on. In contrast to many other channels and receptors, Hv proteins are not thought to possess mechanisms such as inactivation or desensitization. Our findings demonstrate that angiosperm Hv channels are electrically silent until a mechanical stimulation turns on their voltage-dependent activity.
Collapse
Affiliation(s)
- Chang Zhao
- Department of Physiology and Biophysics, University of California, Irvine, CA, 92697, USA
| | - Parker D Webster
- Department of Physiology and Biophysics, University of California, Irvine, CA, 92697, USA
| | - Alexis De Angeli
- IPSiM, University of Montpellier, CNRS, INRAE, Institut Agro, Montpellier, France.
| | - Francesco Tombola
- Department of Physiology and Biophysics, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
11
|
Lazaridis T. Proton Paths in Models of the Hv1 Proton Channel. J Phys Chem B 2023; 127:7937-7945. [PMID: 37695850 DOI: 10.1021/acs.jpcb.3c03960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
The voltage-gated proton channel (Hv1) plays an essential role in numerous biological processes, but a detailed molecular understanding of its function is lacking. The lack of reliable structures for the open and resting states is a major handicap. Several models have been built based on homologous voltage sensors and the structure of a chimera between the mouse homologue and a phosphatase voltage sensor, but their validity is uncertain. In addition, differing views exist regarding the mode of proton translocation, the role of specific residues, and the mechanism of pH effects on voltage gating. Here we use classical proton hopping simulations under a voltage biasing force to evaluate some of the proposed structural models and explore the mechanism of proton conduction. Paradoxically, some models proposed for the closed state allow for proton permeation more easily than models for the open state. An open state model with a D112-R211 salt bridge (R3D) allows proton transport more easily than models with a D112-R208 salt bridge (R2D). However, its permeation rate seems too high, considering experimental conductances. In all cases, the proton permeates through a water wire, bypassing the salt-bridged D112 rather than being shuttled by D112. Attempts to protonate D112 are rejected due to its strong interaction with an arginine. Consistent with proton selectivity, no Na+ permeation was observed in the R2D models. As a negative control, simulations with the Kv1.2-Kv2.1 paddle-chimera voltage sensor, which is not expected to conduct protons, did not show proton permeation under the same conditions. Hydrogen bond connectivity graphs show a constriction at D112, but cannot discriminate between open and closed states.
Collapse
Affiliation(s)
- Themis Lazaridis
- Department of Chemistry, City College of New York/CUNY, 160 Convent Avenue, New York, New York 10031, United States
- Graduate Programs in Chemistry, Biochemistry, and Physics, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, New York 10016, United States
| |
Collapse
|
12
|
Benko F, Urminská D, Ďuračka M, Tvrdá E. Signaling Roleplay between Ion Channels during Mammalian Sperm Capacitation. Biomedicines 2023; 11:2519. [PMID: 37760960 PMCID: PMC10525812 DOI: 10.3390/biomedicines11092519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
In order to accomplish their primary goal, mammalian spermatozoa must undergo a series of physiological, biochemical, and functional changes crucial for the acquisition of fertilization ability. Spermatozoa are highly polarized cells, which must swiftly respond to ionic changes on their passage through the female reproductive tract, and which are necessary for male gametes to acquire their functional competence. This review summarizes the current knowledge about specific ion channels and transporters located in the mammalian sperm plasma membrane, which are intricately involved in the initiation of changes within the ionic milieu of the sperm cell, leading to variations in the sperm membrane potential, membrane depolarization and hyperpolarization, changes in sperm motility and capacitation to further lead to the acrosome reaction and sperm-egg fusion. We also discuss the functionality of selected ion channels in male reproductive health and/or disease since these may become promising targets for clinical management of infertility in the future.
Collapse
Affiliation(s)
- Filip Benko
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (D.U.); (E.T.)
| | - Dana Urminská
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (D.U.); (E.T.)
| | - Michal Ďuračka
- AgroBioTech Research Centre, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
| | - Eva Tvrdá
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (D.U.); (E.T.)
| |
Collapse
|
13
|
Kawanabe A, Takeshita K, Takata M, Fujiwara Y. ATP modulates the activity of the voltage-gated proton channel through direct binding interaction. J Physiol 2023; 601:4073-4089. [PMID: 37555355 DOI: 10.1113/jp284175] [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: 11/29/2022] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
ATP is an important molecule implicated in diverse biochemical processes, including the modulation of ion channel and transporter activity. The voltage-gated proton channel (Hv1) controls proton flow through the transmembrane pathway in response to membrane potential, and various molecules regulate its activity. Although it is believed that ATP is not essential for Hv1 activity, a report has indicated that cytosolic ATP may modulate Hv1. However, the detailed molecular mechanism underlying the effect of ATP on Hv1 is unknown, and whether ATP is involved in the physiological regulation of Hv1 activity remains unclear. Here, we report that cytosolic ATP is required to maintain Hv1 activity. To gain insight into the underlying mechanism, we analysed the effects of ATP on the mouse Hv1 channel (mHv1) using electrophysiological and microscale thermophoresis (MST) methods. Intracellular ATP accelerated the activation kinetics of mHv1, thereby increasing the amplitude of the proton current within the physiological concentration range. The increase in proton current was reproduced with a non-hydrolysable ATP analogue, indicating that ATP directly influences Hv1 activity without an enzymatic reaction. The direct molecular interaction between the purified mHv1 protein and ATP was analysed and demonstrated through MST. In addition, ATP facilitation was observed for the endogenous proton current flowing through Hv1 in the physiological concentration range of ATP. These results suggest that ATP influences Hv1 activity via direct molecular interactions and is required for the physiological function of Hv1. KEY POINTS: We found that ATP is required to maintain the activity of voltage-gated proton channels (Hv1) and investigated the underlying molecular mechanism. Application of intracellular ATP increased the amplitude of the proton current flowing through Hv1, accompanied by an acceleration of activation kinetics. The direct interaction between purified Hv1 protein and ATP was quantitatively analysed using microscale thermophoresis. ATP enhanced endogenous proton currents in breast cancer cell lines. These results suggest that ATP influences Hv1 activity via direct molecular interactions and that its functional characteristics are required for the physiological activity of Hv1.
Collapse
Affiliation(s)
- Akira Kawanabe
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
| | | | - Maki Takata
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
| | - Yuichiro Fujiwara
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
| |
Collapse
|
14
|
Wu X, Singla S, Liu JJ, Hong L. The role of macrophage ion channels in the progression of atherosclerosis. Front Immunol 2023; 14:1225178. [PMID: 37588590 PMCID: PMC10425548 DOI: 10.3389/fimmu.2023.1225178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/10/2023] [Indexed: 08/18/2023] Open
Abstract
Atherosclerosis is a complex inflammatory disease that affects the arteries and can lead to severe complications such as heart attack and stroke. Macrophages, a type of immune cell, play a crucial role in atherosclerosis initiation and progression. Emerging studies revealed that ion channels regulate macrophage activation, polarization, phagocytosis, and cytokine secretion. Moreover, macrophage ion channel dysfunction is implicated in macrophage-derived foam cell formation and atherogenesis. In this context, exploring the regulatory role of ion channels in macrophage function and their impacts on the progression of atherosclerosis emerges as a promising avenue for research. Studies in the field will provide insights into novel therapeutic targets for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xin Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Sidhant Singla
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Jianhua J. Liu
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | - Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
15
|
Shen Y, Luo Y, Liao P, Zuo Y, Jiang R. Role of the Voltage-Gated Proton Channel Hv1 in Nervous Systems. Neurosci Bull 2023; 39:1157-1172. [PMID: 37029856 PMCID: PMC10313628 DOI: 10.1007/s12264-023-01053-6] [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: 10/12/2022] [Accepted: 02/20/2023] [Indexed: 04/09/2023] Open
Abstract
Hv1 is the only voltage-gated proton-selective channel in mammalian cells. It contains a conserved voltage-sensor domain, shared by a large class of voltage-gated ion channels, but lacks a pore domain. Its primary role is to extrude protons from the cytoplasm upon pH reduction and membrane depolarization. The best-known function of Hv1 is the regulation of cytosolic pH and the nicotinamide adenine dinucleotide phosphate oxidase-dependent production of reactive oxygen species. Accumulating evidence indicates that Hv1 is expressed in nervous systems, in addition to immune cells and others. Here, we summarize the molecular properties, distribution, and physiological functions of Hv1 in the peripheral and central nervous systems. We describe the recently discovered functions of Hv1 in various neurological diseases, including brain or spinal cord injury, ischemic stroke, demyelinating diseases, and pain. We also summarize the current advances in the discovery and application of Hv1-targeted small molecules in neurological diseases. Finally, we discuss the current limitations of our understanding of Hv1 and suggest future research directions.
Collapse
Affiliation(s)
- Yu Shen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yuncheng Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yunxia Zuo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610000, China.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Szanto TG, Feher A, Korpos E, Gyöngyösi A, Kállai J, Mészáros B, Ovari K, Lányi Á, Panyi G, Varga Z. 5-Chloro-2-Guanidinobenzimidazole (ClGBI) Is a Non-Selective Inhibitor of the Human H V1 Channel. Pharmaceuticals (Basel) 2023; 16:ph16050656. [PMID: 37242439 DOI: 10.3390/ph16050656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/13/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
5-chloro-2-guanidinobenzimidazole (ClGBI), a small-molecule guanidine derivative, is a known effective inhibitor of the voltage-gated proton (H+) channel (HV1, Kd ≈ 26 μM) and is widely used both in ion channel research and functional biological assays. However, a comprehensive study of its ion channel selectivity determined by electrophysiological methods has not been published yet. The lack of selectivity may lead to incorrect conclusions regarding the role of hHv1 in physiological or pathophysiological responses in vitro and in vivo. We have found that ClGBI inhibits the proliferation of lymphocytes, which absolutely requires the functioning of the KV1.3 channel. We, therefore, tested ClGBI directly on hKV1.3 using a whole-cell patch clamp and found an inhibitory effect similar in magnitude to that seen on hHV1 (Kd ≈ 72 μM). We then further investigated ClGBI selectivity on the hKV1.1, hKV1.4-IR, hKV1.5, hKV10.1, hKV11.1, hKCa3.1, hNaV1.4, and hNaV1.5 channels. Our results show that, besides HV1 and KV1.3, all other off-target channels were inhibited by ClGBI, with Kd values ranging from 12 to 894 μM. Based on our comprehensive data, ClGBI has to be considered a non-selective hHV1 inhibitor; thus, experiments aiming at elucidating the significance of these channels in physiological responses have to be carefully evaluated.
Collapse
Affiliation(s)
- Tibor G Szanto
- Department of Biophysics & Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Adam Feher
- Department of Biophysics & Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Eva Korpos
- Department of Biophysics & Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Adrienn Gyöngyösi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Judit Kállai
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Beáta Mészáros
- Department of Biophysics & Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Krisztian Ovari
- Department of Biophysics & Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Árpád Lányi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Gyorgy Panyi
- Department of Biophysics & Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Zoltan Varga
- Department of Biophysics & Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
19
|
Boytsov D, Brescia S, Chaves G, Koefler S, Hannesschlaeger C, Siligan C, Goessweiner-Mohr N, Musset B, Pohl P. Trapped Pore Waters in the Open Proton Channel H V 1. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205968. [PMID: 36683221 DOI: 10.1002/smll.202205968] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/13/2022] [Indexed: 06/17/2023]
Abstract
The voltage-gated proton channel, HV 1, is crucial for innate immune responses. According to alternative hypotheses, protons either hop on top of an uninterrupted water wire or bypass titratable amino acids, interrupting the water wire halfway across the membrane. To distinguish between both hypotheses, the water mobility for the putative case of an uninterrupted wire is estimated. The predicted single-channel water permeability 2.3 × 10-12 cm3 s-1 reflects the permeability-governing number of hydrogen bonds between water molecules in single-file configuration and pore residues. However, the measured unitary water permeability does not confirm the predicted value. Osmotic deflation of reconstituted lipid vesicles reveals negligible water permeability of the HV 1 wild-type channel and the D174A mutant open at 0 mV. The conductance of 1400 H+ s-1 per wild-type channel agrees with the calculated diffusion limit for a ≈2 Å capture radius for protons. Removal of a charged amino acid (D174) at the pore mouth decreases H+ conductance by reducing the capture radius. At least one intervening amino acid contributes to H+ conductance while interrupting the water wire across the membrane.
Collapse
Affiliation(s)
- Danila Boytsov
- Institute of Biophysics, Johannes Kepler University, 4020 Linz, 40, Gruberstr, Austria
| | - Stefania Brescia
- Institute of Biophysics, Johannes Kepler University, 4020 Linz, 40, Gruberstr, Austria
| | - Gustavo Chaves
- Institute of Physiology, Pathophysiology and Biophysics, CPPB, Paracelsus Medical University, 90419, Nuremberg, Germany
| | - Sabina Koefler
- Institute of Biophysics, Johannes Kepler University, 4020 Linz, 40, Gruberstr, Austria
| | | | - Christine Siligan
- Institute of Biophysics, Johannes Kepler University, 4020 Linz, 40, Gruberstr, Austria
| | | | - Boris Musset
- Institute of Physiology, Pathophysiology and Biophysics, CPPB, Paracelsus Medical University, 90419, Nuremberg, Germany
| | - Peter Pohl
- Institute of Biophysics, Johannes Kepler University, 4020 Linz, 40, Gruberstr, Austria
| |
Collapse
|
20
|
El Chemaly A, Jaquet V, Cambet Y, Caillon A, Cherpin O, Balafa A, Krause KH, Demaurex N. Discovery and validation of new Hv1 proton channel inhibitors with onco-therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119415. [PMID: 36640925 DOI: 10.1016/j.bbamcr.2022.119415] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 01/13/2023]
Abstract
The voltage-gated hydrogen channel Hv1 encoded in humans by the HVCN1 gene is a highly selective proton channel that allows large fluxes of protons across biological membranes. Hv1 form functional dimers of four transmembrane spanning proteins resembling the voltage sensing domain of potassium channels. Each subunit is highly selective for protons and is controlled by changes in the transmembrane voltage and pH gradient. Hv1 is most expressed in phagocytic cells where it sustains NADPH oxidase-dependent bactericidal function and was reported to facilitate antibody production by B cells and to promote the maturation and motility of spermatocytes. Hv1 contributes to neuroinflammation following brain damage and favors cancer progression possibly by extruding protons generated during aerobic glycolysis of cancer cells. Lack of specific Hv1 inhibitors has hampered translation of this knowledge to treat immune, fertility, or malignancy diseases. In this study, we show that the genetic deletion of Hv1 delays tumor development in a mouse model of granulocytic sarcoma and report the discovery and characterization of two novel bioavailable inhibitors of Hv1 channels that we validate by orthogonal assays and electrophysiological recordings.
Collapse
Affiliation(s)
- Antoun El Chemaly
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Vincent Jaquet
- Department of Pathology and Immunology, University of Geneva, Geneva 1211, Switzerland; READs unit, University of Geneva, Geneva 1211, Switzerland
| | - Yves Cambet
- READs unit, University of Geneva, Geneva 1211, Switzerland
| | - Aurélie Caillon
- Department of Pathology and Immunology, University of Geneva, Geneva 1211, Switzerland
| | - Ophélie Cherpin
- Department of Pathology and Immunology, University of Geneva, Geneva 1211, Switzerland
| | - Alexia Balafa
- Department of Pathology and Immunology, University of Geneva, Geneva 1211, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva, Geneva 1211, Switzerland
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva 1211, Switzerland.
| |
Collapse
|
21
|
Yan S, Wang B, Lin H. Tracking the Delocalized Proton in Concerted Proton Transfer in Bulk Water. J Chem Theory Comput 2023; 19:448-459. [PMID: 36630655 DOI: 10.1021/acs.jctc.2c01097] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A solvated proton in water is often characterized as a charge or structural defect, and it is important to track its evolution on-the-fly in certain dynamics simulations. Previously, we introduced the proton indicator, a pseudo-atom, whose position approximates the location of the excess proton modeled as a structural defect. The proton indicator generally yields a smooth trajectory of a hydrated proton diffusing in aqueous solutions, including in the events of stepwise proton transfer via the Grotthuss mechanism; however, the proton indicator did not perform well in the events of concerted proton transfer, for which it occasionally yielded large position displacements between two successive time steps. To overcome this hurdle, we develop a new algorithm of a proton indicator with greatly enhanced performance for concerted proton transfer in bulk water. A protocol is proposed to exhaustively explore the hydrogen-bonding network of the water wires over which the excess proton is delocalized and to properly account for the contributions of the water molecules in this network as the geometry evolves. The new proton indicator (called Indicator 2.0) is assessed in dynamics simulations of an excess proton in bulk water and in specially constructed model systems of more complex architectures. The results demonstrate that the new indicator yields a smooth trajectory in both stepwise and concerted proton transfers.
Collapse
Affiliation(s)
- Shengheng Yan
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen360015P. R. China
| | - Binju Wang
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen360015P. R. China
| | - Hai Lin
- Department of Chemistry, CB 194, University of Colorado Denver, P.O. Box 173364, Denver, Colorado80217, United States
| |
Collapse
|
22
|
Lazaridis T. Molecular origins of asymmetric proton conduction in the influenza M2 channel. Biophys J 2023; 122:90-98. [PMID: 36403086 PMCID: PMC9822799 DOI: 10.1016/j.bpj.2022.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 11/20/2022] Open
Abstract
The M2 proton channel of influenza A is embedded into the viral envelope and allows acidification of the virion when the external pH is lowered. In contrast, no outward proton conductance is observed when the internal pH is lowered, although outward current is observed at positive voltage. Residues Trp41 and Asp44 are known to play a role in preventing pH-driven outward conductance, but the mechanism for this is unclear. We investigate this issue using classical molecular dynamics simulations with periodic proton hops. When all key His37 residues are neutral, inward proton movement is much more facile than outward movement if the His are allowed to shuttle the proton. The preference for inward movement increases further as the charge on the His37 increases. Analysis of the trajectories reveals three factors accounting for this asymmetry. First, in the outward direction, Asp44 traps the hydronium by strong electrostatic interactions. Secondly, Asp44 and Trp41 orient the hydronium with the protons pointing inward, hampering outward Grotthus hopping. As a result, the effective barrier is lower in the inward direction. Trp41 adds to the barrier by weakly H-bonding to potential H+ acceptors. Finally, for charged His, the H3O+ in the inner vestibule tends to get trapped at lipid-lined fenestrations of the cone-shaped channel. Simulations qualitatively reproduce the experimentally observed higher outward conductance of mutants. The ability of positive voltage, unlike proton gradient, to induce an outward current appears to arise from its ability to bias H3O+ and the waters around it toward more H-outward orientations.
Collapse
Affiliation(s)
- Themis Lazaridis
- Department of Chemistry, City College of New York/CUNY, New York, New York; Graduate Programs in Chemistry, Biochemistry, and Physics, The Graduate Center, City University of New York, New York, New York.
| |
Collapse
|
23
|
Zhao R, Lopez B, Schwingshackl A, Goldstein SA. Protection from acute lung injury by a peptide designed to inhibit the voltage-gated proton channel. iScience 2022; 26:105901. [PMID: 36660473 PMCID: PMC9843441 DOI: 10.1016/j.isci.2022.105901] [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: 02/27/2022] [Revised: 12/06/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022] Open
Abstract
There are no targeted medical therapies for Acute Lung Injury (ALI) or its most severe form acute respiratory distress syndrome (ARDS). Infections are the most common cause of ALI/ARDS and these disorders present clinically with alveolar inflammation and barrier dysfunction due to the influx of neutrophils and inflammatory mediator secretion. We designed the C6 peptide to inhibit voltage-gated proton channels (Hv1) and demonstrated that it suppressed the release of reactive oxygen species (ROS) and proteases from neutrophils in vitro. We now show that intravenous C6 counteracts bacterial lipopolysaccharide (LPS)-induced ALI in mice, and suppresses the accumulation of neutrophils, ROS, and proinflammatory cytokines in bronchoalveolar lavage fluid. Confirming the salutary effects of C6 are via Hv1, genetic deletion of the channel similarly protects mice from LPS-induced ALI. This report reveals that Hv1 is a key regulator of ALI, that Hv1 is a druggable target, and that C6 is a viable agent to treat ALI/ARDS.
Collapse
Affiliation(s)
- Ruiming Zhao
- Departments of Pediatrics, Physiology & Biophysics, and Pharmaceutical Sciences, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Benjamin Lopez
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andreas Schwingshackl
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA 90095, USA,Corresponding author
| | - Steve A.N. Goldstein
- Departments of Pediatrics, Physiology & Biophysics, and Pharmaceutical Sciences, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, Irvine, CA 92697, USA,Corresponding author
| |
Collapse
|
24
|
Ekimova M, Kleine C, Ludwig J, Ochmann M, Agrenius TEG, Kozari E, Pines D, Pines E, Huse N, Wernet P, Odelius M, Nibbering ETJ. From Local Covalent Bonding to Extended Electric Field Interactions in Proton Hydration. Angew Chem Int Ed Engl 2022; 61:e202211066. [PMID: 36102247 PMCID: PMC9827956 DOI: 10.1002/anie.202211066] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Indexed: 01/12/2023]
Abstract
Seemingly simple yet surprisingly difficult to probe, excess protons in water constitute complex quantum objects with strong interactions with the extended and dynamically changing hydrogen-bonding network of the liquid. Proton hydration plays pivotal roles in energy transport in hydrogen fuel cells and signal transduction in transmembrane proteins. While geometries and stoichiometry have been widely addressed in both experiment and theory, the electronic structure of these specific hydrated proton complexes has remained elusive. Here we show, layer by layer, how utilizing novel flatjet technology for accurate x-ray spectroscopic measurements and combining infrared spectral analysis and calculations, we find orbital-specific markers that distinguish two main electronic-structure effects: Local orbital interactions determine covalent bonding between the proton and neigbouring water molecules, while orbital-energy shifts measure the strength of the extended electric field of the proton.
Collapse
Affiliation(s)
- Maria Ekimova
- Max Born Institut für Nichtlineare Optik und KurzzeitspektroskopieMax Born Strasse 2A12489BerlinGermany
| | - Carlo Kleine
- Max Born Institut für Nichtlineare Optik und KurzzeitspektroskopieMax Born Strasse 2A12489BerlinGermany
| | - Jan Ludwig
- Max Born Institut für Nichtlineare Optik und KurzzeitspektroskopieMax Born Strasse 2A12489BerlinGermany
| | - Miguel Ochmann
- Institute for Nanostructure and Solid State Physics, Center for Free-Electron Laser ScienceLuruper Chaussee 14922761HamburgGermany
| | - Thomas E. G. Agrenius
- Department of PhysicsStockholm UniversityAlbaNova University Center106 91StockholmSweden
| | - Eve Kozari
- Department of ChemistryBen Gurion University of the NegevP.O.B. 653Beersheva84105Israel
| | - Dina Pines
- Department of ChemistryBen Gurion University of the NegevP.O.B. 653Beersheva84105Israel
| | - Ehud Pines
- Department of ChemistryBen Gurion University of the NegevP.O.B. 653Beersheva84105Israel
| | - Nils Huse
- Institute for Nanostructure and Solid State Physics, Center for Free-Electron Laser ScienceLuruper Chaussee 14922761HamburgGermany
| | - Philippe Wernet
- Department of Physics and AstronomyUppsala UniversityBox 516 Lägerhyddsvägen 1751 20UppsalaSweden
| | - Michael Odelius
- Department of PhysicsStockholm UniversityAlbaNova University Center106 91StockholmSweden
| | - Erik T. J. Nibbering
- Max Born Institut für Nichtlineare Optik und KurzzeitspektroskopieMax Born Strasse 2A12489BerlinGermany
| |
Collapse
|
25
|
Cordero-Martínez J, Jimenez-Gutierrez GE, Aguirre-Alvarado C, Alacántara-Farfán V, Chamorro-Cevallos G, Roa-Espitia AL, Hernández-González EO, Rodríguez-Páez L. Participation of signaling proteins in sperm hyperactivation. Syst Biol Reprod Med 2022; 68:315-330. [DOI: 10.1080/19396368.2022.2122761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Joaquín Cordero-Martínez
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | | | - Charmina Aguirre-Alvarado
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Unidad de Investigación Médica en Inmunología e Infectología Centro Médico Nacional La Raza, IMSS, Ciudad de México, Mexico
| | - Verónica Alacántara-Farfán
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Germán Chamorro-Cevallos
- Laboratorio de Toxicología Preclínica Departamento de Farmacia Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ana L. Roa-Espitia
- Departamento de Biología Celular Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional, México City, Mexico
| | - Enrique O. Hernández-González
- Departamento de Biología Celular Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional, México City, Mexico
| | - Lorena Rodríguez-Páez
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
26
|
Swain DK, Sharma P, Shah N, Sethi M, Mahajan A, Gupta S, Mishra AK, Yadav S. Introduction to the pathways involved in the activation and regulation of sperm motility: A review of the relevance of ion channels. Anim Reprod Sci 2022; 246:107052. [PMID: 35987804 DOI: 10.1016/j.anireprosci.2022.107052] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022]
Abstract
To participate in sperm-oocyte fusion, spermatozoa need to be motile. In the testes, spermatozoa are immotile, although these gametes acquire the capacity for motility during the transit through the epididymis. During the period of epididymal transport from the male genital tract to the female genital tract, spermatozoa exhibit various types of motility that are regulated by complex signalling and communication mechanisms. Because motility is very dynamic, it can be affected by small changes in the external or internal environment of spermatozoa within a very short time. This indicates that regulatory membrane proteins, known as sperm ion channels, are involved in the regulation of sperm motility. Research results from studies, where there was use of electrophysiological, pharmacological, molecular and knock-out approaches, indicate ion channels are possibly involved in the regulation of sperm membrane polarisation, intracellular pH, motility, energy homeostasis, membrane integrity, capacitation, hyperactivity, acrosome reaction and fertilisation processes. In this review, there is summarisation of the key functions that ion channels have in the regulation, initiation, maintenance, and modulation of sperm motility. In addition, in this review there is highlighting of novel insights about the pathways of ion channels that are activated in spermatozoa while these gametes are located in the oviduct leading to the fertilisation capacity of these cells.
Collapse
Affiliation(s)
- Dilip Kumar Swain
- Sperm Signaling Laboratory, Department of Veterinary Physiology, College of Veterinary Science & Animal Husbandry, U.P. Pandit Deendayal Upadhayaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan, Mathura 281001, Uttar Pradesh, India.
| | - Pratishtha Sharma
- Sperm Signaling Laboratory, Department of Veterinary Physiology, College of Veterinary Science & Animal Husbandry, U.P. Pandit Deendayal Upadhayaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan, Mathura 281001, Uttar Pradesh, India
| | - Nadeem Shah
- Department of Veterinary Gynaecology and Obstetrics, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Manisha Sethi
- Department of Veterinary Gynaecology and Obstetrics, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Abhishek Mahajan
- Sperm Signaling Laboratory, Department of Veterinary Physiology, College of Veterinary Science & Animal Husbandry, U.P. Pandit Deendayal Upadhayaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan, Mathura 281001, Uttar Pradesh, India
| | - Shashikant Gupta
- Animal Reproduction Division, ICAR-Indian Veterinary Research Institute, Izzatnagar, Bareilly 243122, Uttar Pradesh, India
| | | | - Sarvajeet Yadav
- Sperm Signaling Laboratory, Department of Veterinary Physiology, College of Veterinary Science & Animal Husbandry, U.P. Pandit Deendayal Upadhayaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan, Mathura 281001, Uttar Pradesh, India
| |
Collapse
|
27
|
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
|
28
|
Saltarella I, Altamura C, Lamanuzzi A, Apollonio B, Vacca A, Frassanito MA, Desaphy JF. Ion Channels in Multiple Myeloma: Pathogenic Role and Therapeutic Perspectives. Int J Mol Sci 2022; 23:ijms23137302. [PMID: 35806308 PMCID: PMC9266328 DOI: 10.3390/ijms23137302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Ion channels are pore-forming proteins that allow ions to flow across plasma membranes and intracellular organelles in both excitable and non-excitable cells. They are involved in the regulation of several biological processes (i.e., proliferation, cell volume and shape, differentiation, migration, and apoptosis). Recently, the aberrant expression of ion channels has emerged as an important step of malignant transformation, tumor progression, and drug resistance, leading to the idea of “onco-channelopathy”. Here, we review the contribution of ion channels and transporters in multiple myeloma (MM), a hematological neoplasia characterized by the expansion of tumor plasma cells (MM cells) in the bone marrow (BM). Deregulation of ion channels sustains MM progression by modulating intracellular pathways that promote MM cells’ survival, proliferation, and drug resistance. Finally, we focus on the promising role of ion channels as therapeutic targets for the treatment of MM patients in a combination strategy with currently used anti-MM drugs to improve their cytotoxic activity and reduce adverse effects.
Collapse
Affiliation(s)
- Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Section of Pharmacology, University of Bari Aldo Moro Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, Section of Pharmacology, University of Bari Aldo Moro Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Aurelia Lamanuzzi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine, University of Bari Aldo Moro Medical School, I-70124 Bari, Italy
| | - Benedetta Apollonio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine, University of Bari Aldo Moro Medical School, I-70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine, University of Bari Aldo Moro Medical School, I-70124 Bari, Italy
| | - Maria Antonia Frassanito
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari Aldo Moro Medical School, I-70124 Bari, Italy
| | - Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, Section of Pharmacology, University of Bari Aldo Moro Medical School, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| |
Collapse
|
29
|
Molecular determinants of inhibition of the human proton channel hHv1 by the designer peptide C6 and a bivalent derivative. Proc Natl Acad Sci U S A 2022; 119:e2120750119. [PMID: 35648818 PMCID: PMC9191634 DOI: 10.1073/pnas.2120750119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We designed C6 peptide to address the absence of specific inhibitors of human voltage-gated proton channels (hHv1). Two C6 bind to the two hHv1 voltage sensors at the resting state, inhibiting activation on depolarization. Here, we identify the C6–hHv1 binding interface using tethered-toxin variants and channel mutants, unveil an important role for negatively charged lipids, and present a model of the C6–hHv1 complex. Inspired by nature, we create a peptide with two C6 epitopes (C62) that binds to both channel subunits simultaneously, yielding picomolar affinity and significantly improved inhibition at high potentials. C6 and C62 are peptides designed to regulate hHv1, a channel involved in innate immune-system inflammatory pathophysiology, sperm capacitation, cancer-cell proliferation, and tissue damage in ischemic stroke. The human voltage-gated proton channel (hHv1) is important for control of intracellular pH. We designed C6, a specific peptide inhibitor of hHv1, to evaluate the roles of the channel in sperm capacitation and in the inflammatory immune response of neutrophils [R. Zhao et al., Proc. Natl. Acad. Sci. U.S.A. 115, E11847–E11856 (2018)]. One C6 binds with nanomolar affinity to each of the two S3–S4 voltage-sensor loops in hHv1 in cooperative fashion so that C6-bound channels require greater depolarization to open and do so more slowly. As depolarization drives hHv1 sensors outwardly, C6 affinity decreases, and inhibition is partial. Here, we identified residues essential to C6–hHv1 binding by scanning mutagenesis, five in the hHv1 S3–S4 loops and seven on C6. A structural model of the C6–hHv1 complex was then generated by molecular dynamics simulations and validated by mutant-cycle analysis. Guided by this model, we created a bivalent C6 peptide (C62) that binds simultaneously to both hHv1 subunits and fully inhibits current with picomolar affinity. The results help delineate the structural basis for C6 state-dependent inhibition, support an anionic lipid-mediated binding mechanism, and offer molecular insight into the effectiveness of engineered C6 as a therapeutic agent or lead.
Collapse
|
30
|
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: 13] [Impact Index Per Article: 6.5] [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
|
31
|
Chatterjee S, Zamani E, Farzin S, Evazzade I, Obewhere OA, Johnson TJ, Alexandrov V, Dishari SK. Molecular-Level Control over Ionic Conduction and Ionic Current Direction by Designing Macrocycle-Based Ionomers. JACS AU 2022; 2:1144-1159. [PMID: 35647599 PMCID: PMC9131371 DOI: 10.1021/jacsau.2c00143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 06/15/2023]
Abstract
Poor ionic conductivity of the catalyst-binding, sub-micrometer-thick ionomer layers in energy conversion and storage devices is a huge challenge. However, ionomers are rarely designed keeping in mind the specific issues associated with nanoconfinement. Here, we designed nature-inspired ionomers (calix-2) having hollow, macrocyclic, calix[4]arene-based repeat units with precise, sub-nanometer diameter. In ≤100 nm-thick films, the in-plane proton conductivity of calix-2 was up to 8 times higher than the current benchmark ionomer Nafion at 85% relative humidity (RH), while it was 1-2 orders of magnitude higher than Nafion at 20-25% RH. Confocal laser scanning microscopy and other synthetic techniques allowed us to demonstrate the role of macrocyclic cavities in boosting the proton conductivity. The systematic self-assembly of calix-2 chains into ellipsoids in thin films was evidenced from atomic force microscopy and grazing incidence small-angle X-ray scattering measurements. Moreover, the likelihood of alignment and stacking of macrocyclic units, the presence of one-dimensional water wires across this macrocycle stacks, and thus the formation of long-range proton conduction pathways were suggested by atomistic simulations. We not only did see an unprecedented improvement in thin-film proton conductivity but also saw an improvement in proton conductivity of bulk membranes when calix-2 was added to the Nafion matrices. Nafion-calix-2 composite membranes also took advantage of the asymmetric charge distribution across calix[4]arene repeat units collectively and exhibited voltage-gating behavior. The inclusion of molecular macrocyclic cavities into the ionomer chemical structure can thus emerge as a promising design concept for highly efficient ion-conducting and ion-permselective materials for sustainable energy applications.
Collapse
Affiliation(s)
| | | | | | - Iman Evazzade
- Department of Chemical and Biomolecular
Engineering, University of Nebraska−Lincoln, Lincoln 68588, Nebraska, United States
| | - Oghenetega Allen Obewhere
- Department of Chemical and Biomolecular
Engineering, University of Nebraska−Lincoln, Lincoln 68588, Nebraska, United States
| | - Tyler James Johnson
- Department of Chemical and Biomolecular
Engineering, University of Nebraska−Lincoln, Lincoln 68588, Nebraska, United States
| | - Vitaly Alexandrov
- Department of Chemical and Biomolecular
Engineering, University of Nebraska−Lincoln, Lincoln 68588, Nebraska, United States
| | - Shudipto Konika Dishari
- Department of Chemical and Biomolecular
Engineering, University of Nebraska−Lincoln, Lincoln 68588, Nebraska, United States
| |
Collapse
|
32
|
Kim JH, Chae MR, Wijerathne TD, Cooray AD, Kim CY, Lee SW, Lee KP. In vitro assessment of Prunus japonica seed extract on human spermatozoa hypermotility and intracellular alkalization. Andrologia 2022; 54:e14471. [PMID: 35590125 DOI: 10.1111/and.14471] [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: 01/17/2022] [Revised: 03/25/2022] [Accepted: 04/17/2022] [Indexed: 11/29/2022] Open
Abstract
Prunus japonica var. nakaii is used in traditional Korean medicine to treat various conditions; however, it has not been investigated for treating male infertility. In this study, we investigated the in vitro effects of the ethanolic extract of P. japonica seeds on human sperm motility and identified its mechanism of action. Eleven male volunteers were selected, and the effects of the extract on human spermatozoa were assessed through a computer-assisted semen analysis. The P. japonica seed extract increased the percentage of total and progressive motility of spermatozoa. To understand the mechanism of action, we monitored intracellular alkalization using flow cytometry and obtained electrophysiological recordings of human voltage-gated proton channels hHv1 that were overexpressed in HEK-293 cells. The extract shifted the activation curves in a concentration-dependent manner. Two major constituents of the extract, linoleic acid and oleic acid, exhibited proton channel activity. Our in vitro experiments suggested that P. japonica seed extract could be potentially used to rescue sperm motility in idiopathic infertility patients via pharmacological modulation of the proton channels during capacitation. Therefore, our results indicate the therapeutic potential of P. japonica seed extract for treating male infertility.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, South Korea
| | - Mee Ree Chae
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Tharaka Darshana Wijerathne
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, South Korea
| | - Akila Dushyantha Cooray
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, South Korea
| | - Chul Young Kim
- College of Pharmacy, Hanyang University, Ansan, South Korea
| | - Sung Won Lee
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyu Pil Lee
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
33
|
Insight into the function of a unique voltage-sensor protein (TMEM266) and its short form in mouse cerebellum. Biochem J 2022; 479:1127-1145. [PMID: 35574701 DOI: 10.1042/bcj20220033] [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: 01/26/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
Abstract
Voltage-sensing proteins generally consist of voltage-sensor domains and pore-gate domains, forming the voltage-gated ion channels. However, there are several unconventional voltage-sensor proteins that lack pore-gate domains, conferring them unique voltage-sensing machinery. TMEM266, which is expressed in cerebellum granule cells, is one of the interesting voltage-sensing proteins that has a putative intracellular coiled-coil and a functionally unidentified cytosolic region instead of a pore-gate domain. Here, we approached the molecular function of TMEM266 by performing co-immunoprecipitation experiments. We unexpectedly discovered that TMEM266 proteins natively interact with the novel short form splice variants that only have voltage-sensor domains and putative cytosolic coiled-coil region in cerebellum. The crystal structure of coiled-coil region of TMEM266 suggested that these coiled-coil regions play significant roles in forming homodimers. In vitro expression experiments supported the idea that short form TMEM266 (sTMEM266) or full length TMEM266 (fTMEM266) form homodimers. We also performed proximity labeling mass spectrometry analysis for fTMEM266 and sTMEM266 using Neuro-2A, neuroblastoma cells, and fTMEM266 showed more interacting molecules than sTMEM266, suggesting that the C-terminal cytosolic region in fTMEM266 binds to various targets. Finally, TMEM266-deficient animals showed the moderate abnormality in open-field test. The present study provides clues about the novel voltage-sensing mechanism mediated by TMEM266.
Collapse
|
34
|
Reduced H + channel activity disrupts pH homeostasis and calcification in coccolithophores at low ocean pH. Proc Natl Acad Sci U S A 2022; 119:e2118009119. [PMID: 35522711 PMCID: PMC9171652 DOI: 10.1073/pnas.2118009119] [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] [Indexed: 11/18/2022] Open
Abstract
Coccolithophore calcification is a major ocean biogeochemical process. While this process is likely to be sensitive to acidification-driven changes in ocean carbonate chemistry, incomplete understanding of the underlying mechanisms and constraints is a major bottleneck in predicting ocean acidification effects on calcification. We report severe disruption of pH homeostasis linked to a loss of H+ channel function in the coccolithophore Coccolithus braarudii acclimated to seawater pH values that are likely to be encountered currently in localized regions and more widely in future oceans. This disruption leads to specific defects in coccolith morphology. These findings provide mechanistic insight into how calcification in different coccolithophores is affected by changes in seawater carbonate chemistry. Coccolithophores are major producers of ocean biogenic calcite, but this process is predicted to be negatively affected by future ocean acidification scenarios. Since coccolithophores calcify intracellularly, the mechanisms through which changes in seawater carbonate chemistry affect calcification remain unclear. Here we show that voltage-gated H+ channels in the plasma membrane of Coccolithus braarudii serve to regulate pH and maintain calcification under normal conditions but have greatly reduced activity in cells acclimated to low pH. This disrupts intracellular pH homeostasis and impairs the ability of C. braarudii to remove H+ generated by the calcification process, leading to specific coccolith malformations. These coccolith malformations can be reproduced by pharmacological inhibition of H+ channels. Heavily calcified coccolithophore species such as C. braarudii, which make the major contribution to carbonate export to the deep ocean, have a large intracellular H+ load and are likely to be most vulnerable to future decreases in ocean pH.
Collapse
|
35
|
Wang G, Nauseef WM. Neutrophil dysfunction in the pathogenesis of cystic fibrosis. Blood 2022; 139:2622-2631. [PMID: 35213685 PMCID: PMC9053701 DOI: 10.1182/blood.2021014699] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) figure prominently in host defense against infection and in noninfectious inflammation. Mobilized early in an inflammatory response, PMNs mediate immediate cellular defense against microbes and orchestrate events that culminate in cessation of inflammation and restoration of homeostasis. Failure to terminate the inflammatory response and its causes can fuel exuberant inflammation characteristic of many human diseases, including cystic fibrosis (CF), an autosomal recessive genetic disease caused by mutations in the CF transmembrane conductance regulator. CF affects multiple end organs, with persistent bacterial infection and chronic neutrophilic inflammation in airways predominating the clinical picture. To match the diverse microbial challenges that they may encounter, PMNs possess a variety of antimicrobial systems to slow or kill invading microorganisms confined in their phagosomes. Prominent among PMN defense systems is their ability to generate hypochlorous acid, a potent microbicide, by reacting oxidants generated by the NADPH oxidase with myeloperoxidase (MPO) released from azurophilic granules in the presence of chloride (Cl-). Products of the MPO-H2O2-Cl system oxidize susceptible biomolecules and support robust antimicrobial action against many, but not all, potential human pathogens. Underscoring that the MPO-H2O2-Cl system is integral to optimal host defense and proper regulation of inflammation, individuals with defects in any component of this system, as seen in chronic granulomatous disease or MPO deficiency, incur increased rates or severity of infection and signs of dysregulated inflammatory responses. We focus attention in this review on the molecular basis for and the clinical consequences of defects in the MPO-H2O2-Cl system because of the compromised Cl transport seen in CF. We will discuss first how the MPO-H2O2-Cl system in healthy PMNs participates in host defense and resolution of inflammation and then review how a defective MPO-H2O2-Cl system contributes to the increased susceptibility to infection and dysregulated inflammation associated with the clinical manifestations of CF.
Collapse
Affiliation(s)
- Guoshun Wang
- Department of Microbiology, Immunology, and Parasitology, and
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA
| | - William M Nauseef
- Inflammation Program, Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA; and
- Veterans Administration Medical Center, Iowa City, IA
| |
Collapse
|
36
|
Coe D, Poobalasingam T, Fu H, Bonacina F, Wang G, Morales V, Moregola A, Mitro N, Cheung KC, Ward EJ, Nadkarni S, Aksentijevic D, Bianchi K, Norata GD, Capasso M, Marelli-Berg FM. Loss of hydrogen voltage-gated channel-1 expression reveals heterogeneous metabolic adaptation to intracellular acidification by T-cells. JCI Insight 2022; 7:147814. [PMID: 35472029 PMCID: PMC9220931 DOI: 10.1172/jci.insight.147814] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Hvcn1 is a voltage-gated proton channel, which reduces cytosol acidification and facilitates the production of reactive oxygen species (ROS). The increased expression of this channel in some cancers, has led to proposing Hvcn1 antagonists as potential therapeutics.While its role in most leukocytes has been studied in-depth, the function of Hvcn1 in T-cells remains poorly defined. We show that HVCN1 plays a non-redundant role in protecting naïve T-cells from intracellular acidification during priming. Despite sharing overall functional impairment in vivo and in vitro, Hvcn1-deficient CD4+ and CD8+ T-cells display profound differences during the transition from naïve to primed T-cells, including in the preservation of TCR signaling, cellular division and death. These selective features result, at least in part, from a substantially different metabolic response to intracellular acidification associated with priming. While Hvcn1-deficient naïve CD4+ T-cells reprogram to rescue the glycolytic pathway, naïve CD8+ T-cells, which express high levels of this channel in the mitochondria, respond by metabolically compensating mitochondrial dysfunction, at least in part via AMPK activation.These observations imply heterogeneity between adaptation of naïve CD4+ and CD8+ T-cells to intracellular acidification during activation.
Collapse
Affiliation(s)
- David Coe
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | | - Hongmei Fu
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Guosu Wang
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Valle Morales
- Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Annalisa Moregola
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | | | - Eleanor J Ward
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Suchita Nadkarni
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Dunja Aksentijevic
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | | | | |
Collapse
|
37
|
Zheng W, Shen C, Wang L, Rawson S, Xie WJ, Nist-Lund C, Wu J, Shen Z, Xia S, Holt JR, Wu H, Fu TM. pH regulates potassium conductance and drives a constitutive proton current in human TMEM175. SCIENCE ADVANCES 2022; 8:eabm1568. [PMID: 35333573 PMCID: PMC8956256 DOI: 10.1126/sciadv.abm1568] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 02/03/2022] [Indexed: 06/01/2023]
Abstract
Human TMEM175, a noncanonical potassium (K+) channel in endolysosomes, contributes to their pH stability and is implicated in the pathogenesis of Parkinson's disease (PD). Structurally, the TMEM175 family exhibits an architecture distinct from canonical potassium channels, as it lacks the typical TVGYG selectivity filter. Here, we show that human TMEM175 not only exhibits pH-dependent structural changes that reduce K+ permeation at acidic pH but also displays proton permeation. TMEM175 constitutively conducts K+ at pH 7.4 but displays reduced K+ permeation at lower pH. In contrast, proton current through TMEM175 increases with decreasing pH because of the increased proton gradient. Molecular dynamics simulation, structure-based mutagenesis, and electrophysiological analysis suggest that K+ ions and protons share the same permeation pathway. The M393T variant of human TMEM175 associated with PD shows reduced function in both K+ and proton permeation. Together, our structural and electrophysiological analysis reveals a mechanism of TMEM175 regulation by pH.
Collapse
Affiliation(s)
- Wang Zheng
- Departments of Otolaryngology and Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chen Shen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Longfei Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Shaun Rawson
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Wen Jun Xie
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Carl Nist-Lund
- Departments of Otolaryngology and Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jason Wu
- Departments of Otolaryngology and Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zhangfei Shen
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Shiyu Xia
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Jeffrey R. Holt
- Departments of Otolaryngology and Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Tian-Min Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
38
|
Wu X, Li Y, Maienschein-Cline M, Feferman L, Wu L, Hong L. RNA-Seq Analyses Reveal Roles of the HVCN1 Proton Channel in Cardiac pH Homeostasis. Front Cell Dev Biol 2022; 10:860502. [PMID: 35372367 PMCID: PMC8967321 DOI: 10.3389/fcell.2022.860502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/16/2022] [Indexed: 12/13/2022] Open
Abstract
The voltage-gated proton channel HVCN1 is a member of the voltage-gated ion channel family. HVCN1 channel controls acid extrusion and regulates pH homeostasis in various cell types. Recent evidence indicated that the HVCN1 channel was associated with cardiac function. To investigate the role of HVCN1 in cardiac myocytes, we performed an RNA sequencing analysis of murine hearts and showed that HVCN1 null hearts exhibited a differential transcriptome profile compared with wild-type hearts. The RNA-seq data indicating impaired pH homeostasis in HVCN1 null hearts were the downregulated NADPH oxidoreductases (NOXs) and decreased expression of Cl−/HCO3− exchanger, indicating HVCN1 is a regulator of gene transcriptional networks controlling NOX signaling and CO2 homeostasis in the heart. Additionally, HVCN1 null hearts exhibited differential expression of cardiac ion channels, suggesting a potential role of HVCN1 in cardiac electrophysiological remodeling. The study highlights the importance of HVCN1 in cardiac function and may present a novel target associated with heart diseases.
Collapse
Affiliation(s)
- Xin Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Yawei Li
- Department of Preventive Medicine, Northwestern University, Chicago, IL, United States
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Leonid Feferman
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Longjun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Liang Hong,
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Roy S, Zheng L, Silberbush O, Engel M, Atsmon-Raz Y, Miller Y, Migliore A, Beratan DN, Ashkenasy N. Mechanism of Side Chain-Controlled Proton Conductivity in Bioinspired Peptidic Nanostructures. J Phys Chem B 2021; 125:12741-12752. [PMID: 34780197 DOI: 10.1021/acs.jpcb.1c08857] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bioinspired peptide assemblies are promising candidates for use as proton-conducting materials in electrochemical devices and other advanced technologies. Progress toward applications requires establishing foundational structure-function relationships for transport in these materials. This experimental-theoretical study sheds light on how the molecular structure and proton conduction are linked in three synthetic cyclic peptide nanotube assemblies that comprise the three canonical basic amino acids (lysine, arginine, and histidine). Experiments find an order of magnitude higher proton conductivity for lysine-containing peptide assemblies compared to histidine and arginine containing assemblies. The simulations indicate that, upon peptide assembly, the basic amino acid side chains are close enough to enable direct proton transfer. The proton transfer kinetics is determined in the simulations to be governed by the structure and flexibility of the side chains. Together, experiments and theory indicate that the proton mobility is the main determinant of proton conductivity, critical for the performance of peptide-based devices.
Collapse
Affiliation(s)
- Subhasish Roy
- Department of Materials Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Lianjun Zheng
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Ohad Silberbush
- Department of Materials Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Maor Engel
- Department of Materials Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Yoav Atsmon-Raz
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Yifat Miller
- Department of Chemistry, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel.,Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| | - Agostino Migliore
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States.,Department of Chemical Sciences, University of Padova, Via Marzolo, 1, Padova 35131, Italy
| | - David N Beratan
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States.,Department of Physics, Duke University, Durham, North Carolina 27708, United States.,Department of Biochemistry, Duke University, Durham, North Carolina 27710, United States
| | - Nurit Ashkenasy
- Department of Materials Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel.,Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 84105, Israel
| |
Collapse
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
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
|
46
|
Aldana A, Carneiro J, Martínez-Mekler G, Darszon A. Discrete Dynamic Model of the Mammalian Sperm Acrosome Reaction: The Influence of Acrosomal pH and Physiological Heterogeneity. Front Physiol 2021; 12:682790. [PMID: 34349664 PMCID: PMC8328089 DOI: 10.3389/fphys.2021.682790] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/28/2021] [Indexed: 01/31/2023] Open
Abstract
The acrosome reaction (AR) is an exocytotic process essential for mammalian fertilization. It involves diverse physiological changes (biochemical, biophysical, and morphological) that culminate in the release of the acrosomal content to the extracellular medium as well as a reorganization of the plasma membrane (PM) that allows sperm to interact and fuse with the egg. In spite of many efforts, there are still important pending questions regarding the molecular mechanism regulating the AR. Particularly, the contribution of acrosomal alkalinization to AR triggering physiological conditions is not well understood. Also, the dependence of the proportion of sperm capable of undergoing AR on the physiological heterogeneity within a sperm population has not been studied. Here, we present a discrete mathematical model for the human sperm AR based on the physiological interactions among some of the main components of this complex exocytotic process. We show that this model can qualitatively reproduce diverse experimental results, and that it can be used to analyze how acrosomal pH (pH a ) and cell heterogeneity regulate AR. Our results confirm that a pH a increase can on its own trigger AR in a subpopulation of sperm, and furthermore, it indicates that this is a necessary step to trigger acrosomal exocytosis through progesterone, a known natural inducer of AR. Most importantly, we show that the proportion of sperm undergoing AR is directly related to the detailed structure of the population physiological heterogeneity.
Collapse
Affiliation(s)
- Andrés Aldana
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jorge Carneiro
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova, Oeiras, Portugal
| | - Gustavo Martínez-Mekler
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
47
|
Zhao C, Hong L, Galpin JD, Riahi S, Lim VT, Webster PD, Tobias DJ, Ahern CA, Tombola F. HIFs: New arginine mimic inhibitors of the Hv1 channel with improved VSD-ligand interactions. J Gen Physiol 2021; 153:212451. [PMID: 34228044 PMCID: PMC8263924 DOI: 10.1085/jgp.202012832] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 06/14/2021] [Indexed: 12/14/2022] Open
Abstract
The human voltage-gated proton channel Hv1 is a drug target for cancer, ischemic stroke, and neuroinflammation. It resides on the plasma membrane and endocytic compartments of a variety of cell types, where it mediates outward proton movement and regulates the activity of NOX enzymes. Its voltage-sensing domain (VSD) contains a gated and proton-selective conduction pathway, which can be blocked by aromatic guanidine derivatives such as 2-guanidinobenzimidazole (2GBI). Mutation of Hv1 residue F150 to alanine (F150A) was previously found to increase 2GBI apparent binding affinity more than two orders of magnitude. Here, we explore the contribution of aromatic interactions between the inhibitor and the channel in the presence and absence of the F150A mutation, using a combination of electrophysiological recordings, classic mutagenesis, and site-specific incorporation of fluorinated phenylalanines via nonsense suppression methodology. Our data suggest that the increase in apparent binding affinity is due to a rearrangement of the binding site allowed by the smaller residue at position 150. We used this information to design new arginine mimics with improved affinity for the nonrearranged binding site of the wild-type channel. The new compounds, named “Hv1 Inhibitor Flexibles” (HIFs), consist of two “prongs,” an aminoimidazole ring, and an aromatic group connected by extended flexible linkers. Some HIF compounds display inhibitory properties that are superior to those of 2GBI, thus providing a promising scaffold for further development of high-affinity Hv1 inhibitors.
Collapse
Affiliation(s)
- Chang Zhao
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
| | - Liang Hong
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA
| | - Jason D Galpin
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
| | - Saleh Riahi
- Department of Chemistry, University of California, Irvine, Irvine, CA
| | - Victoria T Lim
- Department of Chemistry, University of California, Irvine, Irvine, CA
| | - Parker D Webster
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA
| | - Douglas J Tobias
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA.,Department of Chemistry, University of California, Irvine, Irvine, CA
| | - Christopher A Ahern
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
| | - Francesco Tombola
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
| |
Collapse
|
48
|
Zhao C, Hong L, Riahi S, Lim VT, Tobias DJ, Tombola F. A novel Hv1 inhibitor reveals a new mechanism of inhibition of a voltage-sensing domain. J Gen Physiol 2021; 153:212452. [PMID: 34228045 PMCID: PMC8263925 DOI: 10.1085/jgp.202012833] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 06/14/2021] [Indexed: 12/16/2022] Open
Abstract
Voltage-gated sodium, potassium, and calcium channels consist of four voltage-sensing domains (VSDs) that surround a central pore domain and transition from a down state to an up state in response to membrane depolarization. While many types of drugs bind pore domains, the number of organic molecules known to bind VSDs is limited. The Hv1 voltage-gated proton channel is made of two VSDs and does not contain a pore domain, providing a simplified model for studying how small ligands interact with VSDs. Here, we describe a ligand, named HIF, that interacts with the Hv1 VSD in the up and down states. We find that HIF rapidly inhibits proton conduction in the up state by blocking the open channel, as previously described for 2-guanidinobenzimidazole and its derivatives. HIF, however, interacts with a site slowly accessible in the down state. Functional studies and MD simulations suggest that this interaction traps the compound in a narrow pocket lined with charged residues within the VSD intracellular vestibule, which results in slow recovery from inhibition. Our findings point to a “wrench in gears” mechanism whereby side chains within the binding pocket trap the compound as the teeth of interlocking gears. We propose that the use of screening strategies designed to target binding sites with slow accessibility, similar to the one identified here, could lead to the discovery of new ligands capable of interacting with VSDs of other voltage-gated ion channels in the down state.
Collapse
Affiliation(s)
- Chang Zhao
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
| | - Liang Hong
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA
| | - Saleh Riahi
- Department of Chemistry, University of California, Irvine, Irvine, CA
| | - Victoria T Lim
- Department of Chemistry, University of California, Irvine, Irvine, CA
| | - Douglas J Tobias
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA.,Department of Chemistry, University of California, Irvine, Irvine, CA
| | - Francesco Tombola
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA
| |
Collapse
|
49
|
Zhao R, Dai H, Arias RJ, De Blas GA, Orta G, Pavarotti MA, Shen R, Perozo E, Mayorga LS, Darszon A, Goldstein SAN. Direct activation of the proton channel by albumin leads to human sperm capacitation and sustained release of inflammatory mediators by neutrophils. Nat Commun 2021; 12:3855. [PMID: 34158477 PMCID: PMC8219737 DOI: 10.1038/s41467-021-24145-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 06/02/2021] [Indexed: 02/05/2023] Open
Abstract
Human voltage-gated proton channels (hHv1) extrude protons from cells to compensate for charge and osmotic imbalances due metabolism, normalizing intracellular pH and regulating protein function. Human albumin (Alb), present at various levels throughout the body, regulates oncotic pressure and transports ligands. Here, we report Alb is required to activate hHv1 in sperm and neutrophils. Dose-response studies reveal the concentration of Alb in semen is too low to activate hHv1 in sperm whereas the higher level in uterine fluid yields proton efflux, allowing capacitation, the acrosomal reaction, and oocyte fertilization. Likewise, Alb activation of hHv1 in neutrophils is required to sustain production and release of reactive oxygen species during the immune respiratory burst. One Alb binds to both voltage sensor domains (VSDs) in hHv1, enhancing open probability and increasing proton current. A computational model of the Alb-hHv1 complex, validated by experiments, identifies two sites in Alb domain II that interact with the VSDs, suggesting an electrostatic gating modification mechanism favoring the active "up" sensor conformation. This report shows how sperm are triggered to fertilize, resolving how hHv1 opens at negative membrane potentials in sperm, and describes a role for Alb in physiology that will operate in the many tissues expressing hHv1.
Collapse
Affiliation(s)
- Ruiming Zhao
- Departments of Pediatrics and Physiology & Biophysics, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, USA
| | - Hui Dai
- Departments of Pediatrics and Physiology & Biophysics, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, USA
| | - Rodolfo J Arias
- Instituto de Histología y Embriología de Mendoza (IHEM/CONICET-UNCuyo), School of Medicine, National University of Cuyo, Mendoza, CP, Argentina
| | - Gerardo A De Blas
- Instituto de Histología y Embriología de Mendoza (IHEM/CONICET-UNCuyo), School of Medicine, National University of Cuyo, Mendoza, CP, Argentina
- Laboratorio de Telediagnóstico e Investigación Traslacional (LaTIT). Área de Farmacología. Departamento de Patología, School of Medicine, National University of Cuyo, Mendoza, CP, Argentina
| | - Gerardo Orta
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Morelos, México
| | - Martín A Pavarotti
- Instituto de Histología y Embriología de Mendoza (IHEM/CONICET-UNCuyo), School of Medicine, National University of Cuyo, Mendoza, CP, Argentina
| | - Rong Shen
- Department of Biochemistry and Molecular Biology, Gordon Center for Integrative Science, University of Chicago, Chicago, IL, USA
| | - Eduardo Perozo
- Department of Biochemistry and Molecular Biology, Gordon Center for Integrative Science, University of Chicago, Chicago, IL, USA
| | - Luis S Mayorga
- Instituto de Histología y Embriología de Mendoza (IHEM/CONICET-UNCuyo), School of Medicine, National University of Cuyo, Mendoza, CP, Argentina
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Morelos, México
| | - Steve A N Goldstein
- Departments of Pediatrics and Physiology & Biophysics, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA, USA.
| |
Collapse
|
50
|
Kozari E, Sigalov M, Pines D, Fingerhut BP, Pines E. Infrared and NMR Spectroscopic Fingerprints of the Asymmetric H 7+ O 3 Complex in Solution. Chemphyschem 2021; 22:716-725. [PMID: 33599024 PMCID: PMC8252526 DOI: 10.1002/cphc.202001046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/13/2021] [Indexed: 12/17/2022]
Abstract
Infrared (IR) absorption in the 1000-3700 cm-1 range and 1 H NMR spectroscopy reveal the existence of an asymmetric protonated water trimer, H7+ O3, in acetonitrile. The core H7+ O3 motif persists in larger protonated water clusters in acetonitrile up to at least 8 water molecules. Quantum mechanics/molecular mechanics (QM/MM) molecular dynamics (MD) simulations reveal irreversible proton transport promoted by propagating the asymmetric H7+ O3 structure in solution. The QM/MM calculations allow for the successful simulation of the measured IR absorption spectra of H7+ O3 in the OH stretch region, which reaffirms the assignment of the H7+ O3 spectra to a hybrid-complex structure: a protonated water dimer strongly hydrogen-bonded to a third water molecule with the proton exchanging between the two possible shared-proton Zundel-like centers. The H7+ O3 structure lends itself to promoting irreversible proton transport in presence of even one additional water molecule. We demonstrate how continuously evolving H7+ O3 structures may support proton transport within larger water solvates.
Collapse
Affiliation(s)
- Eve Kozari
- Department of ChemistryBen-Gurion University of the NegevP.O. Box 653Beer-Sheva84105Israel
| | - Mark Sigalov
- Department of ChemistryBen-Gurion University of the NegevP.O. Box 653Beer-Sheva84105Israel
| | - Dina Pines
- Department of ChemistryBen-Gurion University of the NegevP.O. Box 653Beer-Sheva84105Israel
| | - Benjamin P. Fingerhut
- Max-Born-Institut für Nichtlineare Optik und KurzzeitspektroskopieBerlin12489Germany
| | - Ehud Pines
- Department of ChemistryBen-Gurion University of the NegevP.O. Box 653Beer-Sheva84105Israel
| |
Collapse
|