1
|
Okochi Y, Jinno Y, Okamura Y. Dimerization is required for the glycosylation of S1-S2 linker of sea urchin voltage-gated proton channel Hv1. Biophys J 2024; 123:4221-4232. [PMID: 39086135 DOI: 10.1016/j.bpj.2024.07.034] [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: 04/03/2024] [Revised: 06/08/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Multimerization of ion channels is essential for establishing the ion-selective pathway and tuning the gating regulated by membrane potential, second messengers, and temperature. Voltage-gated proton channel, Hv1, consists of voltage-sensor domain and coiled-coil domain. Hv1 forms dimer, whereas voltage-dependent channel activity is self-contained in monomer unlike many ion channels, which assemble to form ion-conductive pathways among multiple subunits. Dimerization of Hv1 is necessary for cooperative gating, but other roles of dimerization in physiological aspects are still largely unclear. In this study, we show that dimerization of Hv1 takes place in ER. Sea urchin Hv1 (Strongylocentrotus purpuratus Hv1: SpHv1) was glycosylated in the consensus sequence for N-linked glycosylation within the S1-S2 extracellular loop. However, glycosylation was not observed in the monomeric SpHv1 that lacks the coiled-coil domain. A version of mHv1 in which the S1-S2 loop was replaced by that of SpHv1 showed glycosylation and its monomeric form was not glycosylated. Tandem dimer of monomeric SpHv1 underwent glycosylation, suggesting that dimerization of Hv1 is required for glycosylation. Moreover, when monomeric Hv1 has a dilysine motif in the C-terminal end, which is known to act as a retrieval signal from Golgi to ER, prolonging the time of residency in ER, it was glycosylated. Overall, our results suggest that monomeric SpHv1 does not stay long in ER, thereby escaping glycosylation, while the dimerization causes the proteins to stay longer in ER. Thus, the findings highlight the novel significance of dimerization of Hv1: regulation of biogenesis and maturation of the proteins in intracellular compartments.
Collapse
Affiliation(s)
- Yoshifumi Okochi
- Integrative Physiology, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Yuka Jinno
- Integrative Physiology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yasushi Okamura
- Integrative Physiology, Graduate School of Medicine, Osaka University, Suita, Japan; Graduate School of Frontier Biosciences, Osaka University, Suita, Japan.
| |
Collapse
|
2
|
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
|
3
|
Fujiwara Y. Temperature Dependent Activity of the Voltage-Gated Proton Channel. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:109-125. [PMID: 39289277 DOI: 10.1007/978-981-97-4584-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Voltage-gated proton channel (Hv) has activity of proton transport following electrochemical gradient of proton. Hv is expressed in neutrophils and macrophages of which functions are physiologically temperature-sensitive. Hv is also expressed in human sperm cells and regulates their locomotion. H+ transport through Hv is both regulated by membrane potential and pH difference across biological membrane. It is also reported that properties of Hv such as proton conductance and gating are highly temperature-dependent. Hv consists of the N-terminal cytoplasmic domain, the voltage sensor domain (VSD), and the C-terminal coiled-coil domain, and H+ permeates through VSD voltage-dependently. The functional unit of Hv is a dimer via the interaction between C-terminal coiled-coils assembly domain. We have reported that the coiled-coil domain of Hv has the nature of dissociation around our bodily temperature and mutational change of the coiled-coil affected temperature-sensitive gating, especially its temperature threshold. The temperature-sensitive gating is assessed from two separate points: temperature threshold and temperature dependence. In this chapter, I describe physiological roles and molecular structure mechanisms of Hv by mainly focusing on thermosensitive properties.
Collapse
Affiliation(s)
- Yuichiro Fujiwara
- Molecular Physiology & Biophysics, Faculty of Medicine/Graduate School of Medicine, Kagawa University, Miki-cho, Kagawa, Japan.
| |
Collapse
|
4
|
Taya T, Teruyama F, Gojo S. Host-directed therapy for bacterial infections -Modulation of the phagolysosome pathway. Front Immunol 2023; 14:1227467. [PMID: 37841276 PMCID: PMC10570837 DOI: 10.3389/fimmu.2023.1227467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Bacterial infections still impose a significant burden on humanity, even though antimicrobial agents have long since been developed. In addition to individual severe infections, the f fatality rate of sepsis remains high, and the threat of antimicrobial-resistant bacteria grows with time, putting us at inferiority. Although tremendous resources have been devoted to the development of antimicrobial agents, we have yet to recover from the lost ground we have been driven into. Looking back at the evolution of treatment for cancer, which, like infectious diseases, has the similarity that host immunity eliminates the lesion, the development of drugs to eliminate the tumor itself has shifted from a single-minded focus on drug development to the establishment of a treatment strategy in which the de-suppression of host immunity is another pillar of treatment. In infectious diseases, on the other hand, the development of therapies that strengthen and support the immune system has only just begun. Among innate immunity, the first line of defense that bacteria encounter after invading the host, the molecular mechanisms of the phagolysosome pathway, which begins with phagocytosis to fusion with lysosome, have been elucidated in detail. Bacteria have a large number of strategies to escape and survive the pathway. Although the full picture is still unfathomable, the molecular mechanisms have been elucidated for some of them, providing sufficient clues for intervention. In this article, we review the host defense mechanisms and bacterial evasion mechanisms and discuss the possibility of host-directed therapy for bacterial infection by intervening in the phagolysosome pathway.
Collapse
Affiliation(s)
- Toshihiko Taya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fumiya Teruyama
- Pharmacology Research Department, Tokyo New Drug Research Laboratories, Kowa Company, Ltd., Tokyo, Japan
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
Cozzolino M, Gyöngyösi A, Korpos E, Gogolak P, Naseem MU, Kállai J, Lanyi A, Panyi G. The Voltage-Gated Hv1 H+ Channel Is Expressed in Tumor-Infiltrating Myeloid-Derived Suppressor Cells. Int J Mol Sci 2023; 24:ijms24076216. [PMID: 37047188 PMCID: PMC10094655 DOI: 10.3390/ijms24076216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key determinants of the immunosuppressive microenvironment in tumors. As ion channels play key roles in the physiology/pathophysiology of immune cells, we aimed at studying the ion channel repertoire in tumor-derived polymorphonuclear (PMN-MDSC) and monocytic (Mo-MDSC) MDSCs. Subcutaneous tumors in mice were induced by the Lewis lung carcinoma cell line (LLC). The presence of PMN-MDSC (CD11b+/Ly6G+) and Mo-MDSCs (CD11b+/Ly6C+) in the tumor tissue was confirmed using immunofluorescence microscopy and cells were identified as CD11b+/Ly6G+ PMN-MDSCs and CD11b+/Ly6C+/F4/80−/MHCII− Mo-MDSCs using flow cytometry and sorting. The majority of the myeloid cells infiltrating the LLC tumors were PMN-MDSC (~60%) as compared to ~10% being Mo-MDSCs. We showed that PMN- and Mo-MDSCs express the Hv1 H+ channel both at the mRNA and at the protein level and that the biophysical and pharmacological properties of the whole-cell currents recapitulate the hallmarks of Hv1 currents: ~40 mV shift in the activation threshold of the current per unit change in the extracellular pH, high H+ selectivity, and sensitivity to the Hv1 inhibitor ClGBI. As MDSCs exert immunosuppression mainly by producing reactive oxygen species which is coupled to Hv1-mediated H+ currents, Hv1 might be an attractive target for inhibition of MDSCs in tumors.
Collapse
Affiliation(s)
- Marco Cozzolino
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
| | - Adrienn Gyöngyösi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Eva Korpos
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Peter Gogolak
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Muhammad Umair Naseem
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
| | - Judit Kállai
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Arpad Lanyi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (A.G.); (P.G.); (J.K.); (A.L.)
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (M.C.); (E.K.); (M.U.N.)
- Correspondence: ; Tel.: +36-52-352201
| |
Collapse
|
10
|
Naish E, Wood AJT, Stewart AP, Routledge M, Morris AC, Chilvers ER, Lodge KM. The formation and function of the neutrophil phagosome. Immunol Rev 2023; 314:158-180. [PMID: 36440666 PMCID: PMC10952784 DOI: 10.1111/imr.13173] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neutrophils are the most abundant circulating leukocyte and are crucial to the initial innate immune response to infection. One of their key pathogen-eliminating mechanisms is phagocytosis, the process of particle engulfment into a vacuole-like structure called the phagosome. The antimicrobial activity of the phagocytic process results from a collaboration of multiple systems and mechanisms within this organelle, where a complex interplay of ion fluxes, pH, reactive oxygen species, and antimicrobial proteins creates a dynamic antimicrobial environment. This complexity, combined with the difficulties of studying neutrophils ex vivo, has led to gaps in our knowledge of how the neutrophil phagosome optimizes pathogen killing. In particular, controversy has arisen regarding the relative contribution and integration of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-derived antimicrobial agents and granule-delivered antimicrobial proteins. Clinical syndromes arising from dysfunction in these systems in humans allow useful insight into these mechanisms, but their redundancy and synergy add to the complexity. In this article, we review the current knowledge regarding the formation and function of the neutrophil phagosome, examine new insights into the phagosomal environment that have been permitted by technological advances in recent years, and discuss aspects of the phagocytic process that are still under debate.
Collapse
Affiliation(s)
- Emily Naish
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Alexander JT Wood
- Medical SchoolUniversity of Western AustraliaPerthAustralia
- Department of Critical CareUniversity of MelbourneMelbourneAustralia
| | | | - Matthew Routledge
- Department of MedicineUniversity of CambridgeCambridgeUK
- Division of Immunology, Department of PathologyUniversity of CambridgeCambridgeUK
| | - Andrew Conway Morris
- Department of MedicineUniversity of CambridgeCambridgeUK
- Division of Immunology, Department of PathologyUniversity of CambridgeCambridgeUK
| | - Edwin R Chilvers
- National Heart and Lung InstituteImperial College LondonLondonUK
| | | |
Collapse
|
11
|
Neal ML, Beier EE, Hossain MM, Boyle A, Zheng J, Kim C, Mhatre-Winters I, Wu LJ, Richardson JR. Voltage-Gated Proton Channel Hv1 Regulates Neuroinflammation and Dopaminergic Neurodegeneration in Parkinson's Disease Models. Antioxidants (Basel) 2023; 12:582. [PMID: 36978830 PMCID: PMC10044828 DOI: 10.3390/antiox12030582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Although the precise mechanisms for neurodegeneration in Parkinson's disease (PD) are unknown, evidence suggests that neuroinflammation is a critical factor in the pathogenic process. Here, we sought to determine whether the voltage-gated proton channel, Hv1 (HVCN1), which is expressed in microglia and regulates NADPH oxidase, is associated with dopaminergic neurodegeneration. We utilized data mining to evaluate the mRNA expression of HVCN1 in the brains of PD patients and controls and uncovered increased expression of the gene encoding Hv1, HVCN1, in the brains of PD patients compared to controls, specifically in male PD patients. In an acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 4 × 16 mg/kg) mouse model of PD, Hvcn1 gene expression was increased 2-fold in the striatum. MPTP administration to wild-type (WT) mice resulted in a ~65% loss of tyrosine hydroxylase positive neurons (TH+) in the substantia nigra (SN), while a ~39% loss was observed in Hv1 knockout (KO) mice. Comparable neuroprotective effects of Hv1 deficiency were found in a repeated-dose LPS model. Neuroprotection was associated with decreased pro-inflammatory cytokine levels and pro-oxidant factors in both neurotoxicant animal models. These in vivo results were confirmed in primary microglial cultures, with LPS treatment increasing Hvcn1 mRNA levels and Hv1 KO microglia failing to exhibit the LPS-mediated inflammatory response. Conditioned media from Hv1 KO microglia treated with LPS resulted in an attenuated loss of cultured dopamine neuron cell viability compared to WT microglia. Taken together, these data suggest that Hv1 is upregulated and mediates microglial pro-inflammatory cytokine production in parkinsonian models and therefore represents a novel target for neuroprotection.
Collapse
Affiliation(s)
- Matthew L. Neal
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Eric E. Beier
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Muhammad M. Hossain
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Alexa Boyle
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH 44201, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Chunki Kim
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Isha Mhatre-Winters
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jason R. Richardson
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH 44201, USA
| |
Collapse
|
12
|
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
|
13
|
Zheng J, Murugan M, Wang L, Wu LJ. Microglial voltage-gated proton channel Hv1 in spinal cord injury. Neural Regen Res 2022; 17:1183-1189. [PMID: 34782552 PMCID: PMC8643068 DOI: 10.4103/1673-5374.327325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/12/2020] [Accepted: 05/20/2021] [Indexed: 11/23/2022] Open
Abstract
After spinal cord injury, microglia as the first responders to the lesion display both beneficial and detrimental characteristics. Activated microglia phagocyte and eliminate cell debris, release cytokines to recruit peripheral immune cells to the injury site. Excessively activated microglia can aggravate the secondary damage by producing extravagant reactive oxygen species and pro-inflammatory cytokines. Recent studies demonstrated that the voltage-gated proton channel Hv1 is selectively expressed in microglia and regulates microglial activation upon injury. In mouse models of spinal cord injury, Hv1 deficiency ameliorates microglia activation, resulting in alleviated production of reactive oxygen species and pro-inflammatory cytokines. The reduced secondary damage subsequently decreases neuronal loss and correlates with improved locomotor recovery. This review provides a brief historical perspective of advances in investigating voltage-gated proton channel Hv1 and home in on microglial Hv1. We discuss recent studies on the roles of Hv1 activation in pathophysiological activities of microglia, such as production of NOX-dependent reactive oxygen species, microglia polarization, and tissue acidosis, particularly in the context of spinal cord injury. Further, we highlight the rationale for targeting Hv1 for the treatment of spinal cord injury and related disorders.
Collapse
Affiliation(s)
- Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Madhuvika Murugan
- Department of Neurosurgery, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
14
|
Alvear-Arias JJ, Carrillo C, Villar JP, Garcia-Betancourt R, Peña-Pichicoi A, Fernandez A, Fernandez M, Carmona EM, Pupo A, Neely A, Alvarez O, Garate J, Barajas-Martinez H, Larsson HP, Lopez-Rodriguez A, Latorre R, Gonzalez C. Expression of H v1 proton channels in myeloid-derived suppressor cells (MDSC) and its potential role in T cell regulation. Proc Natl Acad Sci U S A 2022; 119:e2104453119. [PMID: 35377790 PMCID: PMC9169626 DOI: 10.1073/pnas.2104453119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 02/14/2022] [Indexed: 12/07/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous cell population with high immunosuppressive activity that proliferates in infections, inflammation, and tumor microenvironments. In tumors, MDSC exert immunosuppression mainly by producing reactive oxygen species (ROS), a process triggered by the NADPH oxidase 2 (NOX2) activity. NOX2 is functionally coupled with the Hv1 proton channel in certain immune cells to support sustained free-radical production. However, a functional expression of the Hv1 channel in MDSC has not yet been reported. Here, we demonstrate that mouse MDSC express functional Hv1 proton channel by immunofluorescence microscopy, flow cytometry, and Western blot, besides performing a biophysical characterization of its macroscopic currents via patch-clamp technique. Our results show that the immunosuppression by MDSC is conditional to their ability to decrease the proton concentration elevated by the NOX2 activity, rendering Hv1 a potential drug target for cancer treatment.
Collapse
Affiliation(s)
- Juan J. Alvear-Arias
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Christian Carrillo
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Javiera Paz Villar
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Richard Garcia-Betancourt
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Antonio Peña-Pichicoi
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Audry Fernandez
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Miguel Fernandez
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Emerson M. Carmona
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Amaury Pupo
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Alan Neely
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Osvaldo Alvarez
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
| | - Jose Garate
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | | | - H. Peter Larsson
- Department of Physiology & Biophysics, University of Miami, Coral Gables, FL 33101
| | | | - Ramon Latorre
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
| | - Carlos Gonzalez
- Millenium Institute Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad de Valparaíso, Valparaíso 236 0102, Chile
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
| |
Collapse
|
15
|
Droste A, Chaves G, Stein S, Trzmiel A, Schweizer M, Karl H, Musset B. Zinc accelerates respiratory burst termination in human PMN. Redox Biol 2021; 47:102133. [PMID: 34562872 PMCID: PMC8476447 DOI: 10.1016/j.redox.2021.102133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 11/25/2022] Open
Abstract
The respiratory burst of phagocytes is essential for human survival. Innate immune defence against pathogens relies strongly on reactive oxygen species (ROS) production by the NADPH oxidase (NOX2). ROS kill pathogens while the translocation of electrons across the plasma membrane via NOX2 depolarizes the cell. Simultaneously, protons are released into the cytosol. Here, we compare freshly isolated human polymorphonuclear leukocytes (PMN) to the granulocytes-like cell line PLB 985. We are recording ROS production while inhibiting the charge compensating and pH regulating voltage-gated proton channel (HV1). The data suggests that human PMN and the PLB 985 generate ROS via a general mechanism, consistent of NOX2 and HV1. Additionally, we advanced a mathematical model based on the biophysical properties of NOX2 and HV1. Our results strongly suggest the essential interconnection of HV1 and NOX2 during the respiratory burst of phagocytes. Zinc chelation during the time course of the experiments postulates that zinc leads to an irreversible termination of the respiratory burst over time. Flow cytometry shows cell death triggered by high zinc concentrations and PMA. Our data might help to elucidate the complex interaction of proteins during the respiratory burst and contribute to decipher its termination.
Collapse
Affiliation(s)
- Annika Droste
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany; Department of Gynecology and Obstetrics, Johannes Gutenberg University, Mainz, Germany
| | - Gustavo Chaves
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
| | - Stefan Stein
- Flow Cytometry Unit, Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Annette Trzmiel
- Flow Cytometry Unit, Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Matthias Schweizer
- Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| | - Hubert Karl
- Department efi, Technische Hochschule Nürnberg Georg Simon Ohm, Nuremberg, Germany
| | - Boris Musset
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany; Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
16
|
Du H, Pang H, Gao Y, Zhou Y, Li SJ. Deficiency of voltage-gated proton channel Hv1 aggravates ovalbumin-induced allergic lung asthma in mice. Int Immunopharmacol 2021; 96:107640. [PMID: 33866247 DOI: 10.1016/j.intimp.2021.107640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Asthma is a chronic airway inflammation that caused by many factors. The voltage-gated proton channel Hv1 has been proposed to extrude excessive protons produced by NADPH oxidase (NOX) from cytosol to maintain its activity during respiratory bursts. Here, we showed that loss of Hv1 aggravates ovalbumin (OVA)-induced allergic lung asthma in mice. The numbers of total cells, eosinophils and neutrophils in bronchoalveolar lavage fluid (BALF) of Hv1-deficiency (KO) mice are obviously increased after OVA challenge compared with that of wild-type (WT) mice. Histopathological staining reveals that Hv1-deficiency aggravates OVA-induced inflammatory cell infiltration and goblet cell hyperplasia in lung tissues. The expression of IL-4, IL-5 and IL-13 are markedly increased in lung tissues of OVA-challenged KO mice compared with that of WT mice. Furthermore, the expression levels of NOX2, NOX4 and DUOX1 are dramatically increased, while the expression levels of SOD2 and catalase are significantly reduced in lung tissues of OVA-challenged KO mice compared with that of WT mice. The production of ROS in lung tissues of KO mice is significantly higher than that of WT mice after OVA challenge. Our data suggest that Hv1-deficiency might aggravate the development of allergic asthma through increasing ROS production.
Collapse
Affiliation(s)
- Hongyan Du
- Department of Biophysics, School of Physical Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Huimin Pang
- Department of Biophysics, School of Physical Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Yingtang Gao
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin Third Central Hospital affiliated to Nankai University, 83 Jintang Road, Hedong District, Tianjin 300170, China
| | - Yongfa Zhou
- Department of Biophysics, School of Physical Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Shu Jie Li
- Department of Biophysics, School of Physical Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China; Qilu Institute of Technology, Shandong 250200, P. R. China.
| |
Collapse
|
17
|
Perelman SS, James DBA, Boguslawski KM, Nelson CW, Ilmain JK, Zwack EE, Prescott RA, Mohamed A, Tam K, Chan R, Narechania A, Pawline MB, Vozhilla N, Moustafa AM, Kim SY, Dittmann M, Ekiert DC, Bhabha G, Shopsin B, Planet PJ, Koralov SB, Torres VJ. Genetic variation of staphylococcal LukAB toxin determines receptor tropism. Nat Microbiol 2021; 6:731-745. [PMID: 33875847 PMCID: PMC8597016 DOI: 10.1038/s41564-021-00890-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 03/11/2021] [Indexed: 02/02/2023]
Abstract
Staphylococcus aureus has evolved into diverse lineages, known as clonal complexes (CCs), which exhibit differences in the coding sequences of core virulence factors. Whether these alterations affect functionality is poorly understood. Here, we studied the highly polymorphic pore-forming toxin LukAB. We discovered that the LukAB toxin variants produced by S. aureus CC30 and CC45 kill human phagocytes regardless of whether CD11b, the previously established LukAB receptor, is present, and instead target the human hydrogen voltage-gated channel 1 (HVCN1). Biochemical studies identified the domain within human HVCN1 that drives LukAB species specificity, enabling the generation of humanized HVCN1 mice with enhanced susceptibility to CC30 LukAB and to bloodstream infection caused by CC30 S. aureus strains. Together, this work advances our understanding of an important S. aureus toxin and underscores the importance of considering genetic variation in characterizing virulence factors and understanding the tug of war between pathogens and the host.
Collapse
Affiliation(s)
- Sofya S Perelman
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - David B A James
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Kristina M Boguslawski
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Chase W Nelson
- Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Juliana K Ilmain
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Erin E Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rachel A Prescott
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Adil Mohamed
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Kayan Tam
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rita Chan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Apurva Narechania
- Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
| | - Miranda B Pawline
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY, USA
| | - Nikollaq Vozhilla
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ahmed M Moustafa
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sang Y Kim
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Office of Collaborative Sciences, NYU Grossman School of Medicine, New York, NY, USA
| | - Meike Dittmann
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Damian C Ekiert
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Skirball Institute of Biomolecular Medicine and Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Gira Bhabha
- Skirball Institute of Biomolecular Medicine and Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, NY, USA
| | - Paul J Planet
- Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sergei B Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
18
|
Okochi Y, Okamura Y. Regulation of Neutrophil Functions by Hv1/VSOP Voltage-Gated Proton Channels. Int J Mol Sci 2021; 22:ijms22052620. [PMID: 33807711 PMCID: PMC7961965 DOI: 10.3390/ijms22052620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
The voltage-gated proton channel, Hv1, also termed VSOP, was discovered in 2006. It has long been suggested that proton transport through voltage-gated proton channels regulate reactive oxygen species (ROS) production in phagocytes by counteracting the charge imbalance caused by the activation of NADPH oxidase. Discovery of Hv1/VSOP not only confirmed this process in phagocytes, but also led to the elucidation of novel functions in phagocytes. The compensation of charge by Hv1/VSOP sustains ROS production and is also crucial for promoting Ca2+ influx at the plasma membrane. In addition, proton extrusion into neutrophil phagosomes by Hv1/VSOP is necessary to maintain neutral phagosomal pH for the effective killing of bacteria. Contrary to the function of Hv1/VSOP as a positive regulator for ROS generation, it has been revealed that Hv1/VSOP also acts to inhibit ROS production in neutrophils. Hv1/VSOP inhibits hypochlorous acid production by regulating degranulation, leading to reduced inflammation upon fungal infection, and suppresses the activation of extracellular signal-regulated kinase (ERK) signaling by inhibiting ROS production. Thus, Hv1/VSOP is a two-way player regulating ROS production. Here, we review the functions of Hv1/VSOP in neutrophils and discuss future perspectives.
Collapse
Affiliation(s)
- Yoshifumi Okochi
- Integrative Physiology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita 5650871, Osaka, Japan;
- Correspondence:
| | - Yasushi Okamura
- Integrative Physiology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita 5650871, Osaka, Japan;
- Graduate School of Frontier Bioscience, Osaka University, 2-2 Yamada-oka, Suita 5650871, Osaka, Japan
| |
Collapse
|
19
|
Kawai T, Takao K, Akter S, Abe M, Sakimura K, Miyakawa T, Okamura Y. Heterogeneity of microglial proton channel in different brain regions and its relationship with aging. J Neurochem 2021; 157:624-641. [PMID: 33404063 DOI: 10.1111/jnc.15292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/13/2022]
Abstract
The properties of microglia largely differ depending on aging as well as on brain regions. However, there are few studies that investigated the functional importance of such heterogeneous properties of microglia at the molecular level. Voltage-gated proton channel, Hv1/VSOP, could be one of the candidates which confers functional heterogeneity among microglia since it regulates brain oxidative stress in age-dependent manner. In this study, we found that Hv1/VSOP shows brain region-dependent heterogeneity of gene expression with the highest level in the striatum. We studied the importance of Hv1/VSOP in two different brain regions, the cerebral cortex and striatum, and examined their relationship with aging (using mice of different ages). In the cortex, we observed the age-dependent impact of Hv1/VSOP on oxidative stress, microglial morphology, and gene expression profile. On the other hand, we found that the age-dependent significance of Hv1/VSOP was less obvious in the striatum than the cortex. Finally, we performed a battery of behavioral experiments on Hv1/VSOP-deficient mice both at young and aged stages to examine the effect of aging on Hv1/VSOP function. Hv1/VSOP-deficient mice specifically showed a marked difference in behavior in light/dark transition test only at aged stages, indicating that anxiety state is altered in aged Hv1/VSOP mice. This study suggests that a combination of brain region heterogeneity and animal aging underscores the functional importance of Hv1/VSOP in microglia.
Collapse
Affiliation(s)
- Takafumi Kawai
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan
| | - Keizo Takao
- Section of Behavior Patterns, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Japan.,Life Science Research Center, University of Toyama, Toyama, Japan
| | - Sharmin Akter
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tsuyoshi Miyakawa
- Section of Behavior Patterns, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Japan.,Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Yasushi Okamura
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan.,Graduate School of Frontier Bioscience, Osaka University, Suita, Japan
| |
Collapse
|
20
|
Thermodynamics and Mechanism of the Membrane Permeation of Hv1 Channel Blockers. J Membr Biol 2020; 254:5-16. [PMID: 33196887 DOI: 10.1007/s00232-020-00149-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023]
Abstract
The voltage-gated proton channel Hv1 mediates efflux of protons from the cell. Hv1 integrally contributes to various physiological processes including pH homeostasis and the respiratory burst of phagocytes. Inhibition of Hv1 may provide therapeutic avenues for the treatment of inflammatory diseases, breast cancer, and ischemic brain damage. In this work, we investigate two prototypical Hv1 inhibitors, 2-guanidinobenzimidazole (2GBI), and 5-chloro-2-guanidinobenzimidazole (GBIC), from an experimentally screened class of guanidine derivatives. Both compounds block proton conduction by binding the same site located on the intracellular side of the channel. However, when added to the extracellular medium, the compounds strongly differ in their ability to inhibit proton conduction, suggesting substantial differences in membrane permeability. Here, we compute the potential of mean force for each compound to permeate through the membrane using atomistic molecular dynamics simulations with the adaptive biasing force method. Our results rationalize the putative distinction between these two blockers with respect to their abilities to permeate the cellular membrane.
Collapse
|
21
|
Murugan M, Zheng J, Wu G, Mogilevsky R, Zheng X, Hu P, Wu J, Wu LJ. The voltage-gated proton channel Hv1 contributes to neuronal injury and motor deficits in a mouse model of spinal cord injury. Mol Brain 2020; 13:143. [PMID: 33081841 PMCID: PMC7574559 DOI: 10.1186/s13041-020-00682-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/06/2020] [Indexed: 01/30/2023] Open
Abstract
Traumatic injury to the spinal cord initiates a series of pathological cellular processes that exacerbate tissue damage at and beyond the original site of injury. This secondary damage includes oxidative stress and inflammatory cascades that can lead to further neuronal loss and motor deficits. Microglial activation is an essential component of these secondary signaling cascades. The voltage-gated proton channel, Hv1, functionally expressed in microglia has been implicated in microglia polarization and oxidative stress in ischemic stroke. Here, we investigate whether Hv1 mediates microglial/macrophage activation and aggravates secondary damage following spinal cord injury (SCI). Following contusion SCI, wild-type (WT) mice showed significant tissue damage, white matter damage and impaired motor recovery. However, mice lacking Hv1 (Hv1−/−) showed significant white matter sparing and improved motor recovery. The improved motor recovery in Hv1−/− mice was associated with decreased interleukin-1β, reactive oxygen/ nitrogen species production and reduced neuronal loss. Further, deficiency of Hv1 directly influenced microglia activation as noted by decrease in microglia numbers, soma size and reduced outward rectifier K+ current density in Hv1−/− mice compared to WT mice at 7 d following SCI. Our results therefore implicate that Hv1 may be a promising potential therapeutic target to alleviate secondary damage following SCI caused by microglia/macrophage activation.
Collapse
Affiliation(s)
- Madhuvika Murugan
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA.,Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA.,Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Gongxiong Wu
- One Harvard Street Institute of Health, Brookline, MA, 02446, USA
| | - Rochelle Mogilevsky
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Xin Zheng
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Peiwen Hu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Junfang Wu
- Department of Anesthesiology, University of Maryland, Baltimore, MD, 21201, USA.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA. .,Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA. .,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA. .,Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
22
|
Kawai T, Kayama K, Tatsumi S, Akter S, Miyawaki N, Okochi Y, Abe M, Sakimura K, Yamamoto H, Kihara S, Okamura Y. Regulation of hepatic oxidative stress by voltage-gated proton channels (Hv1/VSOP) in Kupffer cells and its potential relationship with glucose metabolism. FASEB J 2020; 34:15805-15821. [PMID: 33040408 DOI: 10.1096/fj.202001056rrr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 01/20/2023]
Abstract
Voltage-gated proton channels (Hv1/VSOP), encoded by Hvcn1, are important regulator of reactive oxygen species (ROS) production in many types of immune cells. While in vitro studies indicate that Hv1/VSOP regulates ROS production by maintaining pH homeostasis, there are few studies investigating the functional importance of Hv1/VSOP in vivo. In the present study, we first show that Hv1/VSOP is functionally expressed in liver resident macrophage, Kupffer cells, regulating the hepatic oxidative stress in vivo. Our immunocytochemistry and electrophysiology data showed that Hvcn1 is specifically expressed in Kupffer cells, but not in hepatocytes. Furthermore, Hvcn1-deficiency drastically altered the hepatic oxidative stress. The Hvcn1-deficient mice showed high blood glucose and serum insulin but normal insulin sensitivity, indicating that these phenotypes were not linked to insulin resistance. Transcriptome analysis indicated that the gene expression of glycogen phosphorylase (Pygl) and Glucose-6-phosphatase, catalytic subunit (G6pc) were upregulated in Hvcn1-deficient liver tissues, and quantitative PCR confirmed the result for Pygl. Furthermore, we observed higher amount of glucose-6-phosphate, a key sugar intermediate for glucose in Hvcn1-deficient liver than WT, suggesting that glucose production in liver is accelerated in Hvcn1-deficient mice. The present study sheds light on the functional importance of Kupffer cells in hepatic oxidative stress and its potential relationship with glucose metabolism.
Collapse
Affiliation(s)
- Takafumi Kawai
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan
| | - Kento Kayama
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan.,Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shoki Tatsumi
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan.,Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Sharmin Akter
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan
| | - Nana Miyawaki
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan
| | - Yoshifumi Okochi
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyasu Yamamoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shinji Kihara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasushi Okamura
- Integrative Physiology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
23
|
Voltage-dependent structural models of the human Hv1 proton channel from long-timescale molecular dynamics simulations. Proc Natl Acad Sci U S A 2020; 117:13490-13498. [PMID: 32461356 DOI: 10.1073/pnas.1920943117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The voltage-gated Hv1 proton channel is a ubiquitous membrane protein that has roles in a variety of cellular processes, including proton extrusion, pH regulation, production of reactive oxygen species, proliferation of cancer cells, and increased brain damage during ischemic stroke. A crystal structure of an Hv1 construct in a putative closed state has been reported, and structural models for the channel open state have been proposed, but a complete characterization of the Hv1 conformational dynamics under an applied membrane potential has been elusive. We report structural models of the Hv1 voltage-sensing domain (VSD), both in a hyperpolarized state and a depolarized state resulting from voltage-dependent conformational changes during a 10-μs-timescale atomistic molecular dynamics simulation in an explicit membrane environment. In response to a depolarizing membrane potential, the S4 helix undergoes an outward displacement, leading to changes in the VSD internal salt-bridge network, resulting in a reshaping of the permeation pathway and a significant increase in hydrogen bond connectivity throughout the channel. The total gating charge displacement associated with this transition is consistent with experimental estimates. Molecular docking calculations confirm the proposed mechanism for the inhibitory action of 2-guanidinobenzimidazole (2GBI) derived from electrophysiological measurements and mutagenesis. The depolarized structural model is also consistent with the formation of a metal bridge between residues located in the core of the VSD. Taken together, our results suggest that these structural models are representative of the closed and open states of the Hv1 channel.
Collapse
|
24
|
Okochi Y, Umemoto E, Okamura Y. Hv1/VSOP regulates neutrophil directional migration and ERK activity by tuning ROS production. J Leukoc Biol 2020; 107:819-831. [DOI: 10.1002/jlb.2a0320-110rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Yoshifumi Okochi
- Integrative Physiology, Graduate School of Medicine Osaka University Osaka Japan
| | - Eiji Umemoto
- Laboratory of Microbiology and Immunology School of Pharmaceutical Sciences University of Shizuoka Japan
| | - Yasushi Okamura
- Integrative Physiology, Graduate School of Medicine Osaka University Osaka Japan
- Graduate School of Frontier Biosciences Osaka University Osaka Japan
| |
Collapse
|
25
|
Chaves G, Bungert-Plümke S, Franzen A, Mahorivska I, Musset B. Zinc modulation of proton currents in a new voltage-gated proton channel suggests a mechanism of inhibition. FEBS J 2020; 287:4996-5018. [PMID: 32160407 PMCID: PMC7754295 DOI: 10.1111/febs.15291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/10/2020] [Accepted: 03/10/2020] [Indexed: 02/03/2023]
Abstract
The HV1 voltage‐gated proton (HV1) channel is a key component of the cellular proton extrusion machinery and is pivotal for charge compensation during the respiratory burst of phagocytes. The best‐described physiological inhibitor of HV1 is Zn2+. Externally applied ZnCl2 drastically reduces proton currents reportedly recorded in Homo sapiens, Rattus norvegicus, Mus musculus, Oryctolagus cuniculus, Rana esculenta, Helix aspersa, Ciona intestinalis, Coccolithus pelagicus, Emiliania huxleyi, Danio rerio, Helisoma trivolvis, and Lingulodinium polyedrum, but with considerable species variability. Here, we report the effects of Zn2+ and Cd2+ on HV1 from Nicoletia phytophila, NpHV1. We introduced mutations at potential Zn2+ coordination sites and measured Zn2+ inhibition in different extracellular pH, with Zn2+ concentrations up to 1000 μm. Zn2+ inhibition in NpHV1 was quantified by the slowing of the activation time constant and a positive shift of the conductance–voltage curve. Replacing aspartate in the S3‐S4 loop with histidine (D145H) enhanced both the slowing of activation kinetics and the shift in the voltage–conductance curve, such that Zn2+ inhibition closely resembled that of the human channel. Histidine is much more effective than aspartate in coordinating Zn2+ in the S3‐S4 linker. A simple Hodgkin Huxley model of NpHV1 suggests a decrease in the opening rate if it is inhibited by zinc or cadmium. Limiting slope measurements and high‐resolution clear native gel electrophoresis (hrCNE) confirmed that NpHV1 functions as a dimer. The data support the hypothesis that zinc is coordinated in between the dimer instead of the monomer. Zinc coordination sites may be potential targets for drug development.
Collapse
Affiliation(s)
- Gustavo Chaves
- Institut für Physiologie und Pathophysiologie, Paracelsus Universität Salzburg Standort Nürnberg, Nuremberg, Germany
| | - Stefanie Bungert-Plümke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4) Forschungszentrum Jülich, Jülich, Germany
| | - Arne Franzen
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4) Forschungszentrum Jülich, Jülich, Germany
| | - Iryna Mahorivska
- Institut für Physiologie und Pathophysiologie, Paracelsus Universität Salzburg Standort Nürnberg, Nuremberg, Germany
| | - Boris Musset
- Institut für Physiologie und Pathophysiologie, Paracelsus Universität Salzburg Standort Nürnberg, Nuremberg, Germany
| |
Collapse
|
26
|
Tang D, Yang Y, Xiao Z, Xu J, Yang Q, Dai H, Liang S, Tang C, Dong H, Liu Z. Scorpion toxin inhibits the voltage-gated proton channel using a Zn 2+ -like long-range conformational coupling mechanism. Br J Pharmacol 2020; 177:2351-2364. [PMID: 31975366 DOI: 10.1111/bph.14984] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Blocking the voltage-gated proton channel HV 1 is a promising strategy for the treatment of diseases like ischaemia stroke and cancer. However, few HV 1 channel antagonists have been reported. Here, we have identified a novel HV 1 channel antagonist from scorpion venom and have elucidated its action mechanism. EXPERIMENTAL APPROACH HV 1 and NaV channels were heterologously expressed in mammalian cell lines and their currents recorded using whole-cell patch clamp. Site-directed mutagenesis was used to generate mutants. Toxins were recombinantly produced in Escherichia coli. AGAP/W38F-HV 1 interaction was modelled by molecular dynamics simulations. KEY RESULTS The scorpion toxin AGAP (anti-tumour analgesic peptide) potently inhibited HV 1 currents. One AGAP mutant has reduced NaV channel activity but intact HV 1 activity (AGAP/W38F). AGAP/W38F inhibited HV 1 channel activation by trapping its S4 voltage sensor in a deactivated state and inhibited HV 1 currents with less pH dependence than Zn2+ . Mutation analysis showed that the binding pockets of AGAP/W38F and Zn2+ in HV 1 channel partly overlapped (common sites are His140 and His193). The E153A mutation at the intracellular Coulombic network (ICN) in HV 1 channel markedly reduced AGAP/W38F inhibition, as observed for Zn2+ . Experimental data and MD simulations suggested that AGAP/W38F inhibited HV 1 channel using a Zn2+ -like long-range conformational coupling mechanism. CONCLUSION AND IMPLICATIONS Our results suggest that the Zn2+ binding pocket in HV 1 channel might be a hotspot for modulators and valuable for designing HV 1 channel ligands. Moreover, AGAP/W38F is a useful molecular probe to study HV 1 channel and a lead compound for drug development.
Collapse
Affiliation(s)
- Dongfang Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yuqin Yang
- Kuang Yaming Honors School, Nanjing University, Nanjing, China.,Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Zhen Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jiahui Xu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Qiuchu Yang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Han Dai
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Songping Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Hao Dong
- Kuang Yaming Honors School, Nanjing University, Nanjing, China.,Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
27
|
Pang H, Li J, Du H, Gao Y, Lv J, Liu Y, Li SJ. Loss of voltage-gated proton channel Hv1 leads to diet-induced obesity in mice. BMJ Open Diabetes Res Care 2020; 8:8/1/e000951. [PMID: 32049639 PMCID: PMC7039614 DOI: 10.1136/bmjdrc-2019-000951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE The voltage-gated proton channel Hv1 has been proposed to mediate NADPH oxidase (NOX) function by regulating intracellular pH during respiratory bursts. In our previous work, we showed that Hv1 is expressed in pancreatic β cells and positively regulates insulin secretion. Here, we investigated the role of Hv1 in adipose tissue differentiation, metabolic homeostasis and insulin sensitivity using Hv1 knockout (KO) mice. DESIGN Mice with genetic deletion of Hv1 are treated with high-fat diet (HFD) similar to wild-type (WT) mice. Body weight gain, adiposity, insulin sensitivity and gene expressions in both adipose tissue and liver were analyzed. RESULTS Mice with genetic deletion of Hv1 display overt obesity with higher body weight gain and accumulation of adipose tissue compared with similarly HFD-treated WT. Hv1-deficient mice develop more glucose intolerance than WT, but no significant difference in insulin resistance, after fed with HFD. Deficiency of Hv1 results in a remarkable increase in epididymal adipocyte weight and size, while the gene expressions of proinflammatory factors and cytokines are obviously enhanced in the HFD-fed mice. Furthermore, the gene expression of Hv1 is increased in the HFD-fed mice, which is accompanied by the increase of NOX2 and NOX4. In addition, there is more severely diet-induced steatosis and inflammation in liver in KO mice. CONCLUSION Our data demonstrated that lacking of Hv1 results in diet-induced obesity in mice through inflammation and hepatic steatosis. This study suggested that Hv1 acts as a positive regulator of metabolic homeostasis and a potential target for antiobesity drugs in therapy and may serve as an adaptive mechanism in cooperating with NOX to mediate reactive oxygen species for adipogenesis and insulin sensitivity.
Collapse
Affiliation(s)
- Huimin Pang
- Department of Biophysics, School of Physics Science,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, P. R. China
| | - Jinwen Li
- Department of Biophysics, School of Physics Science,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, P. R. China
| | - Hongyan Du
- Department of Biophysics, School of Physics Science,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, P. R. China
| | - Yingtang Gao
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Tianjin Third Central Hospital affiliated to Nankai University, 83 Jintang Road, Hedong District, Tianjin 300170, China
| | - Jili Lv
- Department of Biophysics, School of Physics Science,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, P. R. China
| | - Yanxia Liu
- Department of Biophysics, School of Physics Science,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, P. R. China
| | - Shu Jie Li
- Department of Biophysics, School of Physics Science,The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
28
|
Pang H, Wang X, Zhao S, Xi W, Lv J, Qin J, Zhao Q, Che Y, Chen L, Li SJ. Loss of the voltage-gated proton channel Hv1 decreases insulin secretion and leads to hyperglycemia and glucose intolerance in mice. J Biol Chem 2020; 295:3601-3613. [PMID: 31949049 DOI: 10.1074/jbc.ra119.010489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/16/2020] [Indexed: 11/06/2022] Open
Abstract
Insulin secretion by pancreatic islet β-cells is regulated by glucose levels and is accompanied by proton generation. The voltage-gated proton channel Hv1 is present in pancreatic β-cells and extremely selective for protons. However, whether Hv1 is involved in insulin secretion is unclear. Here we demonstrate that Hv1 promotes insulin secretion of pancreatic β-cells and glucose homeostasis. Hv1-deficient mice displayed hyperglycemia and glucose intolerance because of reduced insulin secretion but retained normal peripheral insulin sensitivity. Moreover, Hv1 loss contributed much more to severe glucose intolerance as the mice got older. Islets of Hv1-deficient and heterozygous mice were markedly deficient in glucose- and K+-induced insulin secretion. In perifusion assays, Hv1 deletion dramatically reduced the first and second phase of glucose-stimulated insulin secretion. Islet insulin and proinsulin content was reduced, and histological analysis of pancreas slices revealed an accompanying modest reduction of β-cell mass in Hv1 knockout mice. EM observations also indicated a reduction in insulin granule size, but not granule number or granule docking, in Hv1-deficient mice. Mechanistically, Hv1 loss limited the capacity for glucose-induced membrane depolarization, accompanied by a reduced ability of glucose to raise Ca2+ levels in islets, as evidenced by decreased durations of individual calcium oscillations. Moreover, Hv1 expression was significantly reduced in pancreatic β-cells from streptozotocin-induced diabetic mice, indicating that Hv1 deficiency is associated with β-cell dysfunction and diabetes. We conclude that Hv1 regulates insulin secretion and glucose homeostasis through a mechanism that depends on intracellular Ca2+ levels and membrane depolarization.
Collapse
Affiliation(s)
- Huimin Pang
- Department of Biophysics, School of Physics Science, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Xudong Wang
- Department of Biophysics, School of Physics Science, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Shiqun Zhao
- Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Wang Xi
- Department of Biophysics, School of Physics Science, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Jili Lv
- Department of Biophysics, School of Physics Science, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Jiwei Qin
- Department of Biophysics, School of Physics Science, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Qing Zhao
- Department of Biophysics, School of Physics Science, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Yongzhe Che
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Liangyi Chen
- Institute of Molecular Medicine, Peking University, Beijing 100871, China.
| | - Shu Jie Li
- Department of Biophysics, School of Physics Science, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
29
|
Yu Y, Luo X, Li C, Ding F, Wang M, Xie M, Yu Z, Ransom BR, Wang W. Microglial Hv1 proton channels promote white matter injuries after chronic hypoperfusion in mice. J Neurochem 2019; 152:350-367. [PMID: 31769505 DOI: 10.1111/jnc.14925] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 12/30/2022]
Abstract
Microglia are critical in damage/repair processes during ischemic white matter injury (WMI). Voltage-gated proton channel (Hv1) is expressed in microglia and contributes to nicotinamide adenine dinucleotide phosphate oxidase complex-dependent production of reactive oxygen species (ROS). Recent findings have shown that Hv1 is involved in regulating luminal pH of M1-polarized microglial phagosomes and inhibits endocytosis in microglia. We previously reported that Hv1 facilitated production of ROS and pro-inflammatory cytokines in microglia and enhanced damage to oligodendrocyte progenitor cells from oxygen and glucose deprivation. To investigate the role of Hv1 in hypoperfusion-induced WMI, we employed mice that were genetically devoid of Hv1 (Hv1-/- ), as well as a model of subcortical vascular dementia via bilateral common carotid artery stenosis. Integrity of myelin was assessed using immunofluorescent staining and transmission electron microscopy, while cognitive impairment was assessed using an eight-arm radial maze test. Hv1 deficiency was found to attenuate bilateral common carotid artery stenosis-induced disruption of white matter integrity and impairment of working memory. Immunofluorescent staining and western blotting were used to assay changes in oligodendrocytes, OPCs, and microglial polarization. Compared with that in wild-type (WT) mice, Hv1-/- mice exhibited reduced ROS generation, decreased pro-inflammatory cytokines production, and an M2-dominant rather than M1-dominant microglial polarization. Furthermore, Hv1-/- mice exhibited enhanced OPC proliferation and differentiation into oligodendrocytes. Results of mouse-derived microglia-OPC co-cultures suggested that PI3K/Akt signaling was involved in Hv1-deficiency-induced M2-type microglial polarization and concomitant OPC differentiation. These results suggest that microglial Hv1 is a promising therapeutic target for reducing ischemic WMI and cognitive impairment.
Collapse
Affiliation(s)
- Ying Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyu Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengfei Ding
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bruce R Ransom
- Department of Neurology, University of Washington School of Medicine HMC, Seattle, WA, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Nauseef WM. The phagocyte NOX2 NADPH oxidase in microbial killing and cell signaling. Curr Opin Immunol 2019; 60:130-140. [PMID: 31302569 DOI: 10.1016/j.coi.2019.05.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/01/2019] [Accepted: 05/11/2019] [Indexed: 12/17/2022]
Abstract
The phagocyte NADPH oxidase possesses a transmembrane electron transferase comprised of gp91phox (aka NOX2) and p22phox and two multicomponent cytosolic complexes, which in stimulated phagocytes translocate to assemble a functional enzyme complex at plasma or phagosomal membranes. The NOX2-centered NADPH oxidase shuttles electrons from cytoplasmic NADPH to molecular oxygen in phagosomes or the extracellular space to produce oxidants that support optimal antimicrobial activity by phagocytes. Additionally, NOX2-generated oxidants have been implicated in both autocrine and paracrine signaling in a variety of biological contexts. However, when interpreting experimental results, investigators must recognize the complexity inherent in the biochemistry of oxidant-mediated attack of microbial targets and the technical limitations of the probes currently used to detect intracellular oxidants.
Collapse
Affiliation(s)
- William M Nauseef
- Inflammation Program, Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 501 EMRB, 431 Newton Road, Iowa City, IA 52242-1101, United States.
| |
Collapse
|
31
|
Wang X, Gao YT, Jiang D, Wang Y, Du H, Lv J, Li SJ. Hv1-deficiency protects β cells from glucotoxicity through regulation of NOX4 level. Biochem Biophys Res Commun 2019; 513:434-438. [PMID: 30967259 DOI: 10.1016/j.bbrc.2019.03.195] [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: 03/24/2019] [Accepted: 03/29/2019] [Indexed: 11/27/2022]
Abstract
High glucose (HG)-induced oxidative stress contributes to the dysfunction of pancreatic β cells in diabetes. The voltage-gated proton channel Hv1 has been proposed to support reactive oxygen species (ROS) production during respiratory bursts. However, the effect of Hv1 on glucotoxicity in pancreatic β cells is not clear yet. In this study, we examined the protective effects of Hv1-deficiency in HG cultured β cells. Following 48 h of treatment with 30 mM high glucose, Hv1 KO β cells showed higher cell viability, lower cell apoptosis and a more stable insulin gene expression level compared to WT β cells. In both control and HG cultured β cells, deficiency of Hv1 decreased the glucose- and PMA-induced ROS production. Finally, HG incubation led to NOX4 upregulation in WT β cells, which could be inhibited by HV1 deficiency. In conclusion, Hv1-deficiency prevents the HG treatment-induced NOX4 upregulation and protects β cells from glucotoxicity.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Biophysics, School of Physics Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, PR China
| | - Ying-Tang Gao
- Key Laboratory of Artificial Cell, Third Central Clinical College of Tianjin Medical University, Tianjin, 300170, PR China
| | - Dan Jiang
- Department of Biophysics, School of Physics Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, PR China
| | - Yuzhou Wang
- Laboratory Animal Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Hongyan Du
- Department of Biophysics, School of Physics Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, PR China
| | - Jili Lv
- Department of Biophysics, School of Physics Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, PR China
| | - Shu Jie Li
- Department of Biophysics, School of Physics Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
32
|
Ratanayotha A, Kawai T, Okamura Y. Real-time functional analysis of Hv1 channel in neutrophils: a new approach from zebrafish model. Am J Physiol Regul Integr Comp Physiol 2019; 316:R819-R831. [PMID: 30943046 DOI: 10.1152/ajpregu.00326.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Voltage-gated proton channel (Hv1) has been studied in various immune cells, including neutrophils. However, most studies have taken an in vitro approach using isolated cells or primary cultured cells of mammals; therefore, limited evidence is available on the function of Hv1 in a physiological context. In this study, we have developed the in vivo system that enables real-time functional analysis of Hv1 using zebrafish embryos (Danio rerio). Hvcn1-deficiency (hvcn1-/-) in zebrafish completely abolished voltage-gated proton current, which is typically observed in wild-type neutrophils. Importantly, hvcn1-deficiency significantly reduced reactive oxygen species production and calcium response of zebrafish neutrophils, comparable to the results observed in mammalian models. These findings verify zebrafish Hv1 (DrHv1) as the primary contributor for native Hv1-derived proton current in neutrophils and suggest the conserved function of Hv1 in the immune cells across vertebrate animals. Taking advantage of Hv1 zebrafish model, we compared real-time behaviors of neutrophils between wild-type and hvcn1-/- zebrafish in response to tissue injury and acute bacterial infection. Notably, we observed a significant increase in the number of phagosomes in hvcn1-/- neutrophils, raising a possible link between Hv1 and phagosomal maturation. Furthermore, survival analysis of zebrafish larvae potentially supports a protective role of Hv1 in the innate immune response against systemic bacterial infection. This study represents the influence of Hv1 on neutrophil behaviors and highlights the benefits of in vivo approach toward the understanding of Hv1 in a physiological context.
Collapse
Affiliation(s)
- Adisorn Ratanayotha
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University , Suita, Osaka , Japan
| | - Takafumi Kawai
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University , Suita, Osaka , Japan
| | - Yasushi Okamura
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University , Suita, Osaka , Japan
| |
Collapse
|
33
|
Izquierdo P, Attwell D, Madry C. Ion Channels and Receptors as Determinants of Microglial Function. Trends Neurosci 2019; 42:278-292. [PMID: 30678990 DOI: 10.1016/j.tins.2018.12.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022]
Abstract
Microglia provide immune surveillance of the CNS. They display diverse behaviors, including nondirectional and directed motility of their processes, phagocytosis of targets such as dying neurons or superfluous synapses, and generation of reactive oxygen species (ROS) and cytokines. Many of these functions are mediated by ion channels and cell surface receptors, the expression of which varies with the many morphological and functional states that microglial cells can adopt. Recent progress in understanding microglial function has been facilitated by applying classical cell physiological techniques in situ, such as patch-clamping and live imaging, and cell-specific transcriptomic analyses. Here, we review the contribution of microglial ion channels and receptors to microglial and brain function.
Collapse
Affiliation(s)
- Pablo Izquierdo
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower St, London, WC1E 6BT, UK
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower St, London, WC1E 6BT, UK.
| | - Christian Madry
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117 Berlin, Germany.
| |
Collapse
|
34
|
Role of human Hv1 channels in sperm capacitation and white blood cell respiratory burst established by a designed peptide inhibitor. Proc Natl Acad Sci U S A 2018; 115:E11847-E11856. [PMID: 30478045 DOI: 10.1073/pnas.1816189115] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Using a de novo peptide inhibitor, Corza6 (C6), we demonstrate that the human voltage-gated proton channel (hHv1) is the main pathway for H+ efflux that allows capacitation in sperm and permits sustained reactive oxygen species (ROS) production in white blood cells (WBCs). C6 was identified by a phage-display strategy whereby ∼1 million novel peptides were fabricated on an inhibitor cysteine knot (ICK) scaffold and sorting on purified hHv1 protein. Two C6 peptides bind to each dimeric channel, one on the S3-S4 loop of each voltage sensor domain (VSD). Binding is cooperative with an equilibrium affinity (K d) of ∼1 nM at -50 mV. As expected for a VSD-directed toxin, C6 inhibits by shifting hHv1 activation to more positive voltages, slowing opening and speeding closure, effects that diminish with membrane depolarization.
Collapse
|
35
|
Immler R, Simon SI, Sperandio M. Calcium signalling and related ion channels in neutrophil recruitment and function. Eur J Clin Invest 2018; 48 Suppl 2:e12964. [PMID: 29873837 PMCID: PMC6221920 DOI: 10.1111/eci.12964] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/04/2018] [Indexed: 12/15/2022]
Abstract
The recruitment of neutrophils to sites of inflammation, their battle against invading microorganisms through phagocytosis and the release of antimicrobial agents is a highly coordinated and tightly regulated process that involves the interplay of many different receptors, ion channels and signalling pathways. Changes in intracellular calcium levels, caused by cytosolic Ca2+ store depletion and the influx of extracellular Ca2+ via ion channels, play a critical role in synchronizing neutrophil activation and function. In this review, we provide an overview of how Ca2+ signalling is initiated in neutrophils and how changes in intracellular Ca2+ levels modulate neutrophil function.
Collapse
Affiliation(s)
- Roland Immler
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Klinikum der Universität, Ludwig-Maximilians-Universität München, Germany
| | - Scott I. Simon
- Department of Biomedical Engineering, Graduate Group in Immunology, University of California, Davis, CA, USA
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Klinikum der Universität, Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
36
|
Ratanayotha A, Kawai T, Higashijima SI, Okamura Y. Molecular and functional characterization of the voltage-gated proton channel in zebrafish neutrophils. Physiol Rep 2018; 5:5/15/e13345. [PMID: 28774948 PMCID: PMC5555884 DOI: 10.14814/phy2.13345] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 11/24/2022] Open
Abstract
Voltage‐gated proton channels (Hv1/VSOP) are expressed in various cells types, including phagocytes, and are involved in diverse physiological processes. Although hvcn1, the gene encoding Hv1, has been identified across a wide range of species, most of the knowledge about its physiological function and expression profile is limited to mammals. In this study, we investigated the basic properties of DrHv1, the Hv1 ortholog in zebrafish (Danio rerio) which is an excellent animal model owing to the transparency, as well as its functional expression in native cells. Electrophysiological analysis using a heterologous expression system confirmed the properties of a voltage‐gated proton channel are conserved in DrHv1 with differences in threshold and activation kinetics as compared to mouse (Mus musculus) Hv1 (mHv1). RT‐PCR analysis revealed that hvcn1 is expressed in zebrafish neutrophils, as is the case in mammals. Subsequent electrophysiological analysis confirmed the functional expression of DrHv1 in zebrafish neutrophils, which suggests Hv1 function in phagocytes is conserved among vertebrates. We also found that DrHv1 is comparatively resistant to extracellular Zn2+, which is a potent inhibitor of mammalian Hv1, and this phenomenon appears to reflect variation in the Zn2+‐coordinating residue (histidine) within the extracellular linker region in mammalian Hv1. Notably, the serum Zn2+ concentration is much higher in zebrafish than in mouse, raising the possibility that Zn2+ sensitivity was acquired in accordance with a change in the serum Zn2+ concentration. This study highlights the biological variation and importance of Hv1 in different animal species.
Collapse
Affiliation(s)
- Adisorn Ratanayotha
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takafumi Kawai
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shin-Ichi Higashijima
- Laboratory of Behavioral Neurobiology, Department of Biodesign Research, Okazaki Institute for Integrative Bioscience, Okazaki, Aichi, Japan
| | - Yasushi Okamura
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
37
|
Wang X, Xi W, Qin J, Lv J, Wang Y, Zhang T, Li SJ. Deficiency of voltage-gated proton channel Hv1 attenuates streptozotocin-induced β-cell damage. Biochem Biophys Res Commun 2018; 498:975-980. [DOI: 10.1016/j.bbrc.2018.03.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 10/17/2022]
|
38
|
Cooperative electrogenic proton transport pathways in the plasma membrane of the proton-secreting osteoclast. Pflugers Arch 2018; 470:851-866. [PMID: 29550927 DOI: 10.1007/s00424-018-2137-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/13/2018] [Accepted: 03/06/2018] [Indexed: 02/05/2023]
Abstract
A proton is a ubiquitous signaling ion. Many transmembrane H+ transport pathways either maintain pH homeostasis or generate acidic compartments. The osteoclast is a bone-resorbing cell, which degrades bone tissues by secreting protons and lysosomal enzymes into the resorption pit. The plasma membrane facing bone tissue (ruffled border), generated partly by fusion of lysosomes, may mimic H+ flux mechanisms regulating acidic vesicles. We identified three electrogenic H+-fluxes in osteoclast plasma membranes-a vacuolar H+-ATPase (V-ATPase), a voltage-gated proton channel (Hv channel) and an acid-inducible H+-leak-whose electrophysiological profiles and regulation mechanisms differed. V-ATPase and Hv channel, both may have intracellular reservoirs, but the recruitment/internalization is regulated independently. V-ATPase mediates active H+ efflux, acidifying the resorption pit, while acid-inducible H+ leak, activated at an extracellular pH < 5.5, diminishes pit acidification, possibly to protect bone from excess degradation. The two-way H+ flux mechanisms in opposite directions may have advantages in fine regulation of pit pH. Hv channel mediates passive H+ efflux. Although its working ranges are limited, the amount of H+ extrusion is 100 times larger than those of the V-ATPase and may support reactive oxygen species production during osteoclastogenesis. Extracellular Ca2+, H+ and inorganic phosphate, which accumulate in the resorption pit, will either stimulate or inhibit these H+ fluxes. Skeletal integration is disrupted by too much or too less of bone resorption. Diversities in plasma membrane H+ flux pathways, which may co-operate or compete, are essential to adjust osteoclast functions in variable conditions.
Collapse
|
39
|
Boonamnaj P, Sompornpisut P. Insight into the Role of the Hv1 C-Terminal Domain in Dimer Stabilization. J Phys Chem B 2018; 122:1037-1048. [PMID: 29290112 DOI: 10.1021/acs.jpcb.7b08669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The voltage-gated proton-selective channel (Hv1) conducts protons in response to changes in membrane potential. The Hv1 protein forms dimers in the membrane. Crystal structures of Hv1 channels have revealed that the primary contacts between the two monomers are in the C-terminal domain (CTD), which forms a coiled-coil structure. The role of Hv1-CTD in channel assembly and activity is not fully understood. Here, molecular dynamics (MD) simulations of full-length and truncated CTD models of human and mouse Hv1 channels reveal a strong contribution of the CTD to the packing of the transmembrane domains. Simulations of the CTD models highlight four fundamental interactions of the key residues contributing to dimer stability. These include salt bridges, hydrophobic interactions, hydrogen bonds, and a disulfide bond across the dimer interface. At neutral pH, salt-bridge interactions increase dimer stability and the dimer becomes less stable at acidic pH. Hydrophobic core packing of the heptad pattern is important for stability, as shown by favorable nonpolar binding free energies rather than by electrostatic components. Moreover, free-energy calculations indicate that a more uniform hydrophobic core in the coiled-coil structure of the Hv1-NIN, a channel carrying the triple mutation M234N-N235I-V236N, leads to an increase in dimer stability with respect to the wild-type. A Cys disulfide bond has a strong impact on dimer stability by holding the dimer together and facilitating the interactions described above. These results are consistent with dissociative temperatures and energy barriers of dimer dissociation obtained from the temperature-accelerated MD.
Collapse
Affiliation(s)
- Panisak Boonamnaj
- Department of Chemistry, Faculty of Science, Chulalongkorn University , Bangkok 10330, Thailand
| | - Pornthep Sompornpisut
- Department of Chemistry, Faculty of Science, Chulalongkorn University , Bangkok 10330, Thailand
| |
Collapse
|
40
|
Kawai T, Tatsumi S, Kihara S, Sakimura K, Okamura Y. Mechanistic insight into the suppression of microglial ROS production by voltage-gated proton channels (VSOP/Hv1). Channels (Austin) 2017; 12:1-8. [PMID: 28961043 PMCID: PMC5972804 DOI: 10.1080/19336950.2017.1385684] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Voltage-gated proton channels (VSOP/Hv1) reportedly promote reactive oxygen species (ROS) production in several immune cell types. However, we recently reported that primary microglia from VSOP/Hv1-deficient mice show higher ROS production than those from WT mice. Microglia may show a distinct activation status between WT and VSOP/Hv1-deficient cells, leading to a distinct level of ROS production between them. This is unlikely, however, because ROS production in VSOP/Hv1-deficient microglia remained higher than in WT microglia when the cells were exposed to LPS. Further, this increase in ROS production in VSOP/Hv1-deficient cells was not observed in macrophages, which suggests microglia have a unique mechanism of VSOP/Hv1-dependent ROS regulation. The mechanism underlying this unconventional ROS regulation by VSOP/Hv1 in microglia is discussed.
Collapse
Affiliation(s)
- Takafumi Kawai
- a Integrative Physiology, Department of Physiology, Graduate School of Medicine , Osaka University , Osaka , JAPAN
| | - Shoki Tatsumi
- a Integrative Physiology, Department of Physiology, Graduate School of Medicine , Osaka University , Osaka , JAPAN.,b Graduate School of Frontier Biosciences, Osaka University , Osaka , JAPAN.,c Department of Biomedical Informatics , Division of Health Sciences, Graduate School of Medicine, Osaka University , Osaka , JAPAN
| | - Shinji Kihara
- c Department of Biomedical Informatics , Division of Health Sciences, Graduate School of Medicine, Osaka University , Osaka , JAPAN
| | - Kenji Sakimura
- d Department of Cellular Neurobiology , Brain Research Institute, Niigata University , Niigata , JAPAN
| | - Yasushi Okamura
- a Integrative Physiology, Department of Physiology, Graduate School of Medicine , Osaka University , Osaka , JAPAN.,b Graduate School of Frontier Biosciences, Osaka University , Osaka , JAPAN
| |
Collapse
|
41
|
Chae MR, Kang SJ, Lee KP, Choi BR, Kim HK, Park JK, Kim CY, Lee SW. Onion (Allium cepa L.) peel extract (OPE) regulates human sperm motility via protein kinase C-mediated activation of the human voltage-gated proton channel. Andrology 2017; 5:979-989. [PMID: 28805023 DOI: 10.1111/andr.12406] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 06/13/2017] [Accepted: 06/27/2017] [Indexed: 01/14/2023]
Abstract
Onion (Allium cepa L.) and quercetin protect against oxidative damage and have positive effects on multiple functional parameters of spermatozoa, including viability and motility. However, the associated underlying mechanisms of action have not yet been identified. The aim of this study was to investigate the effect of onion peel extract (OPE) on voltage-gated proton (Hv1) channels, which play a critical role in rapid proton extrusion. This process underlies a wide range of physiological processes, particularly male fertility. The whole-cell patch-clamp technique was used to record the changes in Hv1 currents in HEK293 cells transiently transfected with human Hv1 (HVCN1). The effects of OPE on human sperm motility were also analyzed. OPE significantly activated the outward-rectifying proton currents in a concentration-dependent manner, with an EC50 value of 30 μg/mL. This effect was largely reversible upon washout. Moreover, OPE induced an increase in the proton current amplitude and decreased the time constant of activation at 0 mV from 4.9 ± 1.7 to 0.6 ± 0.1 sec (n = 6). In the presence of OPE, the half-activation voltage (V1/2 ) shifted in the negative direction, from 20.1 ± 5.8 to 5.2 ± 8.7 mV (n = 6), but the slope was not significantly altered. The OPE-induced current was profoundly inhibited by 10 μm Zn2+ , the most potent Hv1 channel inhibitor, and was also inhibited by treatment with GF109203X, a specific protein kinase C (PKC) inhibitor. Furthermore, sperm motility was significantly increased in the OPE-treated groups. OPE exhibits protective effects on sperm motility, at least partially via regulation of the proton channel. Moreover, similar effects were exerted by quercetin, the major flavonoid in OPE. These results suggest OPE, which is rich in the potent Hv1 channel activator quercetin, as a possible new candidate treatment for human infertility.
Collapse
Affiliation(s)
- M R Chae
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - S J Kang
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - K P Lee
- Laboratory of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| | - B R Choi
- Department of Urology, Medical School and Institute for Medical Sciences, Chonbuk National University, Research Institute and Clinical Trial Center of Medical Device of Chonbuk National University Hospital, Jeonju, Korea
| | - H K Kim
- College of Pharmacy, Kyungsung University, Busan, Korea
| | - J K Park
- Department of Urology, Medical School and Institute for Medical Sciences, Chonbuk National University, Research Institute and Clinical Trial Center of Medical Device of Chonbuk National University Hospital, Jeonju, Korea
| | - C Y Kim
- College of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Korea
| | - S W Lee
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
42
|
Kawai T, Okochi Y, Ozaki T, Imura Y, Koizumi S, Yamazaki M, Abe M, Sakimura K, Yamashita T, Okamura Y. Unconventional role of voltage‐gated proton channels (
VSOP
/Hv1) in regulation of microglial
ROS
production. J Neurochem 2017. [DOI: 10.1111/jnc.14106] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Takafumi Kawai
- Integrative Physiology Department of Physiology Graduate School of Medicine & Frontier Biosciences Osaka University Suita Osaka Japan
| | - Yoshifumi Okochi
- Integrative Physiology Department of Physiology Graduate School of Medicine & Frontier Biosciences Osaka University Suita Osaka Japan
| | - Tomohiko Ozaki
- Department of Molecular Neuroscience Graduate School of Medicine Osaka University Suita Osaka Japan
| | - Yoshio Imura
- Department of Neuropharmacology Interdisciplinary Graduate School of Medicine University of Yamanashi Chuo Yamanashi Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology Interdisciplinary Graduate School of Medicine University of Yamanashi Chuo Yamanashi Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology Brain Research Institute Niigata University Niigata Japan
| | - Manabu Abe
- Department of Cellular Neurobiology Brain Research Institute Niigata University Niigata Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology Brain Research Institute Niigata University Niigata Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience Graduate School of Medicine Osaka University Suita Osaka Japan
| | - Yasushi Okamura
- Integrative Physiology Department of Physiology Graduate School of Medicine & Frontier Biosciences Osaka University Suita Osaka Japan
| |
Collapse
|
43
|
DeCoursey TE. The intimate and controversial relationship between voltage-gated proton channels and the phagocyte NADPH oxidase. Immunol Rev 2017; 273:194-218. [PMID: 27558336 DOI: 10.1111/imr.12437] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
One of the most fascinating and exciting periods in my scientific career entailed dissecting the symbiotic relationship between two membrane transporters, the Nicotinamide adenine dinucleotide phosphate reduced form (NADPH) oxidase complex and voltage-gated proton channels (HV 1). By the time I entered this field, there had already been substantial progress toward understanding NADPH oxidase, but HV 1 were known only to a tiny handful of cognoscenti around the world. Having identified the first proton currents in mammalian cells in 1991, I needed to find a clear function for these molecules if the work was to become fundable. The then-recent discoveries of Henderson, Chappell, and colleagues in 1987-1988 that led them to hypothesize interactions of both molecules during the respiratory burst of phagocytes provided an excellent opportunity. In a nutshell, both transporters function by moving electrical charge across the membrane: NADPH oxidase moves electrons and HV 1 moves protons. The consequences of electrogenic NADPH oxidase activity on both membrane potential and pH strongly self-limit this enzyme. Fortunately, both consequences specifically activate HV 1, and HV 1 activity counteracts both consequences, a kind of yin-yang relationship. Notwithstanding a decade starting in 1995 when many believed the opposite, these are two separate molecules that function independently despite their being functionally interdependent in phagocytes. The relationship between NADPH oxidase and HV 1 has become a paradigm that somewhat surprisingly has now extended well beyond the phagocyte NADPH oxidase - an industrial strength producer of reactive oxygen species (ROS) - to myriad other cells that produce orders of magnitude less ROS for signaling purposes. These cells with their seven NADPH oxidase (NOX) isoforms provide a vast realm of mechanistic obscurity that will occupy future studies for years to come.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL, USA
| |
Collapse
|
44
|
Sakata S, Miyawaki N, McCormack TJ, Arima H, Kawanabe A, Özkucur N, Kurokawa T, Jinno Y, Fujiwara Y, Okamura Y. Comparison between mouse and sea urchin orthologs of voltage-gated proton channel suggests role of S3 segment in activation gating. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2972-2983. [PMID: 27637155 DOI: 10.1016/j.bbamem.2016.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 09/06/2016] [Accepted: 09/09/2016] [Indexed: 10/21/2022]
Abstract
The voltage-gated proton channel, Hv1, is expressed in blood cells, airway epithelium, sperm and microglia, playing important roles in diverse biological contexts including phagocytosis or sperm maturation through its regulation of membrane potential and pH. The gene encoding Hv1, HVCN1, is widely found across many species and is also conserved in unicellular organisms such as algae or dinoflagellates where Hv1 plays role in calcification or bioluminescence. Voltage-gated proton channels exhibit a large variation of activation rate among different species. Here we identify an Hv1 ortholog from sea urchin, Strongylocentrotus purpuratus, SpHv1. SpHv1 retains most of key properties of Hv1 but exhibits 20-60 times more rapid activation kinetics than mammalian orthologs upon heterologous expression in HEK293T cells. Comparison between SpHv1 and mHv1 highlights novel roles of the third transmembrane segment S3 in activation gating of Hv1.
Collapse
Affiliation(s)
- Souhei Sakata
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Institute of Academic Initiative, Osaka University, Suita, Osaka 565-0871, Japan; National Institute of Natural Sciences, Okazaki, Aichi, Japan
| | - Nana Miyawaki
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Thomas J McCormack
- Section of Developmental Neurophysiology, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), Okazaki, Aichi, Japan
| | - Hiroki Arima
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Akira Kawanabe
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Nurdan Özkucur
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Department of Anatomy, Medical Theoretical Center, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Tatsuki Kurokawa
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Section of Developmental Neurophysiology, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), Okazaki, Aichi, Japan
| | - Yuka Jinno
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuichiro Fujiwara
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasushi Okamura
- Lab. of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Section of Developmental Neurophysiology, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), Okazaki, Aichi, Japan; National Institute of Natural Sciences, Okazaki, Aichi, Japan; Graduate School of Frontier Bioscience, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
45
|
Affiliation(s)
- William M Nauseef
- Inflammation Program and Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Veterans Administration Medical Center, Iowa City, IA, USA
| |
Collapse
|
46
|
Fernández A, Pupo A, Mena-Ulecia K, Gonzalez C. Pharmacological Modulation of Proton Channel Hv1 in Cancer Therapy: Future Perspectives. Mol Pharmacol 2016; 90:385-402. [PMID: 27260771 DOI: 10.1124/mol.116.103804] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 06/02/2016] [Indexed: 12/23/2022] Open
Abstract
The pharmacological modulation of the immunosuppressive tumor microenvironment has emerged as a relevant component for cancer therapy. Several approaches aiming to deplete innate and adaptive suppressive populations, to circumvent the impairment in antigen presentation, and to ultimately increase the frequency of activated tumor-specific T cells are currently being explored. In this review, we address the potentiality of targeting the voltage-gated proton channel, Hv1, as a novel strategy to modulate the tumor microenvironment. The function of Hv1 in immune cells such as macrophages, neutrophils, dendritic cells, and T cells has been associated with the maintenance of NADPH oxidase activity and the generation of reactive oxygen species, which are required for the host defense against pathogens. We discuss evidence suggesting that the Hv1 proton channel could also be important for the function of these cells within the tumor microenvironment. Furthermore, as summarized here, tumor cells express Hv1 as a primary mechanism to extrude the increased amount of protons generated metabolically, thus maintaining physiologic values for the intracellular pH. Therefore, because this channel might be relevant for both tumor cells and immune cells supporting tumor growth, the pharmacological inhibition of Hv1 could be an innovative approach for cancer therapy. With that focus, we analyzed the available compounds that inhibit Hv1, highlighted the need to develop better drugs suitable for patients, and commented on the future perspectives of targeting Hv1 in the context of cancer therapy.
Collapse
Affiliation(s)
- Audry Fernández
- Interdisciplinary Center for Neurosciences of Valparaíso, Faculty of Sciences, University of Valparaíso, Chile
| | - Amaury Pupo
- Interdisciplinary Center for Neurosciences of Valparaíso, Faculty of Sciences, University of Valparaíso, Chile
| | - Karel Mena-Ulecia
- Interdisciplinary Center for Neurosciences of Valparaíso, Faculty of Sciences, University of Valparaíso, Chile
| | - Carlos Gonzalez
- Interdisciplinary Center for Neurosciences of Valparaíso, Faculty of Sciences, University of Valparaíso, Chile
| |
Collapse
|
47
|
Kawanabe A, Okamura Y. Effects of unsaturated fatty acids on the kinetics of voltage-gated proton channels heterologously expressed in cultured cells. J Physiol 2016; 594:595-610. [PMID: 26563684 DOI: 10.1113/jp271274] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/28/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Arachidonic acid (AA) greatly enhances the activity of the voltage-gated proton (Hv) channel, although its mechanism of action and physiological function remain unclear. In the present study, we analysed the effects of AA on proton currents through Hv channels heterologously expressed in HEK293T cells. The dramatic increase in proton current amplitude elicited by AA was accompanied by accelerated activation kinetics and a leftward shift in the voltage-dependence of activation. Mutagenesis studies suggest the two aforementioned effects of AA reflect two distinct structural mechanisms. Application of phospholipase A2 , which liberates AA from phospholipids in the membrane, also enhances Hv channel activity, supporting the idea that AA modulates Hv channel activity within physiological contexts. Unsaturated fatty acids are key components of the biological membranes of all cells, and precursors of mediators for cell signalling. Arachidonic acid (AA) is an unsaturated fatty acid known to modulate the activities of various ion channels, including the voltage-gated proton (Hv) channel, which supports the rapid production of reactive oxygen species (ROS) in phagocytes through regulation of pH and membrane potential. However, the molecular mechanisms and physiological functions of the effects of AA on Hv channels remain unclear. In the present study, we report an electrophysiological analysis of the effects of AA on the mouse Hv channel (mHv1) heterologously expressed in HEK293T cells. Application of AA to excised inside-out patch membranes rapidly induced a robust increase in the amplitude of the proton current through mHv1. The current increase was accompanied by accelerated activation kinetics and a small leftward shift of the current-voltage relationship. In monomeric channels lacking the coiled-coil region of the channel protein, the shift in the current-voltage relationship was diminished but activation and deactivation remained accelerated. Studies with several AA derivatives showed that double bonds and hydrophilic head groups are essential for the effect of AA, although charge was not important. The application of phospholipase A2 (PLA2), which generates AA from cell membrane phospholipids, stimulated mHv1 activity to a similar extent as direct application of ∼ 20 μM AA, suggesting that endogenous AA may regulate Hv channel activity.
Collapse
Affiliation(s)
- Akira Kawanabe
- Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Yasushi Okamura
- Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| |
Collapse
|
48
|
Abstract
Hv1 is a voltage-gated proton-selective channel that plays critical parts in host defense, sperm motility, and cancer progression. Hv1 contains a conserved voltage-sensor domain (VSD) that is shared by a large family of voltage-gated ion channels, but it lacks a pore domain. Voltage sensitivity and proton conductivity are conferred by a unitary VSD that consists of four transmembrane helices. The architecture of Hv1 differs from that of cation channels that form a pore in the center among multiple subunits (as in most cation channels) or homologous repeats (as in voltage-gated sodium and calcium channels). Hv1 forms a dimer in which a cytoplasmic coiled coil underpins the two protomers and forms a single, long helix that is contiguous with S4, the transmembrane voltage-sensing segment. The closed-state structure of Hv1 was recently solved using X-ray crystallography. In this article, we discuss the gating mechanism of Hv1 and focus on cooperativity within dimers and their sensitivity to metal ions.
Collapse
Affiliation(s)
- Yasushi Okamura
- Department of Integrative Physiology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; , ,
| | | | | |
Collapse
|
49
|
Morera FJ, Saravia J, Pontigo JP, Vargas-Chacoff L, Contreras GF, Pupo A, Lorenzo Y, Castillo K, Tilegenova C, Cuello LG, Gonzalez C. Voltage-dependent BK and Hv1 channels expressed in non-excitable tissues: New therapeutics opportunities as targets in human diseases. Pharmacol Res 2015; 101:56-64. [PMID: 26305431 DOI: 10.1016/j.phrs.2015.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/14/2015] [Accepted: 08/14/2015] [Indexed: 11/28/2022]
Abstract
Voltage-gated ion channels are the molecular determinants of cellular excitability. This group of ion channels is one of the most important pharmacological targets in excitable tissues such as nervous system, cardiac and skeletal muscle. Moreover, voltage-gated ion channels are expressed in non-excitable cells, where they mediate key cellular functions through intracellular biochemical mechanisms rather than rapid electrical signaling. This review aims at illustrating the pharmacological impact of these ion channels, highlighting in particular the structural details and physiological functions of two of them - the high conductance voltage- and Ca(2+)-gated K(+) (BK) channels and voltage-gated proton (Hv1) channels- in non-excitable cells. BK channels have been implicated in a variety of physiological processes ranging from regulation of smooth muscle tone to modulation of hormone and neurotransmitter release. Interestingly, BK channels are also involved in modulating K(+) transport in the mammalian kidney and colon epithelium with a potential role in the hyperkalemic phenotype observed in patients with familial hyperkalemic hypertension type 2, and in the pathophysiology of hypertension. In addition, BK channels are responsible for resting and stimulated Ca(2+)-activated K(+) secretion in the distal colon. Hv1 channels have been detected in many cell types, including macrophages, blood cells, lung epithelia, skeletal muscle and microglia. These channels have a central role in the phagocytic system. In macrophages, Hv1 channels participate in the generation of reactive oxygen species in the respiratory burst during the process of phagocytosis.
Collapse
Affiliation(s)
- Francisco J Morera
- Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile.
| | - Julia Saravia
- Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Juan Pablo Pontigo
- Institute of Marine Sciences and Limnology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Vargas-Chacoff
- Institute of Marine Sciences and Limnology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Gustavo F Contreras
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile
| | - Amaury Pupo
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile
| | - Yenisleidy Lorenzo
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile
| | - Karen Castillo
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile
| | - Cholpon Tilegenova
- Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubcock, TX, USA
| | - Luis G Cuello
- Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubcock, TX, USA.
| | - Carlos Gonzalez
- Interdisciplinary Center for Neuroscience of Valparaiso, Faculty of Sciences, Universidad de Valparaiso, Valparaiso, Chile.
| |
Collapse
|
50
|
Liu J, Tian D, Murugan M, Eyo UB, Dreyfus CF, Wang W, Wu LJ. Microglial Hv1 proton channel promotes cuprizone-induced demyelination through oxidative damage. J Neurochem 2015; 135:347-56. [PMID: 26173779 DOI: 10.1111/jnc.13242] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/14/2015] [Accepted: 07/06/2015] [Indexed: 12/21/2022]
Abstract
NADPH oxidase (NOX)-dependent reactive oxygen species (ROS) production in inflammatory cells including microglia plays an important role in demyelination and free radical-mediated tissue injury in multiple sclerosis (MS). However, the mechanism underlying microglial ROS production and demyelination remains largely unknown. The voltage-gated proton channel, Hv1, is selectively expressed in microglia and is required for NOX-dependent ROS generation in the brain. In the present study, we sought to determine the role of microglial Hv1 proton channels in a mouse model of cuprizone-induced demyelination, a model for MS. Following cuprizone exposure, wild-type mice presented obvious demyelination, decreased myelin basic protein expression, loss of mature oligodendrocytes, and impaired motor coordination in comparison to mice on a normal chow diet. However, mice lacking Hv1 (Hv1(-/-) ) are partially protected from demyelination and motor deficits compared with those in wild-type mice. These rescued phenotypes in Hv1(-/-) mice in cuprizone-induced demyelination is accompanied by reduced ROS production, ameliorated microglial activation, increased oligodendrocyte progenitor cell (NG2) proliferation, and increased number of mature oligodendrocytes. These results demonstrate that the Hv1 proton channel is required for cuprizone-induced microglial oxidative damage and subsequent demyelination. Our study suggests that the microglial Hv1 proton channel is a unique target for controlling NOX-dependent ROS production in the pathogenesis of MS.
Collapse
Affiliation(s)
- Junli Liu
- Cancer center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey, USA
| | - Daishi Tian
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey, USA.,Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Madhuvika Murugan
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey, USA
| | - Ukpong B Eyo
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey, USA
| | - Cheryl F Dreyfus
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long-Jun Wu
- Cancer center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|