1
|
Shen M, Huang Y, Cai Z, Cherny VV, DeCoursey TE, Shen J. Interior pH-sensing residue of human voltage-gated proton channel H v1 is histidine 168. Biophys J 2024; 123:4211-4220. [PMID: 39054673 DOI: 10.1016/j.bpj.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/07/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
The molecular mechanisms governing the human voltage-gated proton channel hHv1 remain elusive. Here, we used membrane-enabled hybrid-solvent continuous constant pH molecular dynamics (CpHMD) simulations with pH replica exchange to further evaluate the structural models of hHv1 in the closed (hyperpolarized) and open (depolarized) states recently obtained with MD simulations and explore potential pH-sensing residues. The CpHMD titration at a set of symmetric pH conditions revealed three residues that can gain or lose protons upon channel depolarization. Among them, residue H168 at the intracellular end of the S3 helix switches from the deprotonated to the protonated state and its protonation is correlated with the increased tilting of the S3 helix during the transition from the closed to the open state. Thus, the simulation data suggest H168 as an interior pH sensor, in support of a recent finding based on electrophysiological experiments of Hv1 mutants. We propose that protonation of H168 acts as a key that unlocks the closed channel configuration by increasing the flexibility of the S2-S3 linker, which increases the tilt angle of S3 and enhances the mobility of the S4 helix, thus promoting channel opening. Our work represents an important step toward deciphering the pH-dependent gating mechanism of hHv1.
Collapse
Affiliation(s)
- Mingzhe Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Yandong Huang
- College of Computer Engineering, Jimei University, Xiamen, Fujian Province, China.
| | - Zhitao Cai
- College of Computer Engineering, Jimei University, Xiamen, Fujian Province, China
| | - Vladimir V Cherny
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, Illinois
| | - Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, Illinois
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland.
| |
Collapse
|
2
|
Gonzales GA, Huang S, Wilkinson L, Nguyen JA, Sikdar S, Piot C, Naumenko V, Rajwani J, Wood CM, Dinh I, Moore M, Cedeño E, McKenna N, Polyak MJ, Amidian S, Ebacher V, Rosin NL, Carneiro MB, Surewaard B, Peters NC, Mody CH, Biernaskie J, Yates RM, Mahoney DJ, Canton J. The pore-forming apolipoprotein APOL7C drives phagosomal rupture and antigen cross-presentation by dendritic cells. Sci Immunol 2024; 9:eadn2168. [PMID: 39485861 DOI: 10.1126/sciimmunol.adn2168] [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: 11/28/2023] [Revised: 07/23/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024]
Abstract
Conventional dendritic cells (cDCs) generate protective cytotoxic T lymphocyte (CTL) responses against extracellular pathogens and tumors. This is achieved through a process known as cross-presentation (XP), and, despite its biological importance, the mechanism(s) driving XP remains unclear. Here, we show that a cDC-specific pore-forming protein called apolipoprotein L 7C (APOL7C) is up-regulated in response to innate immune stimuli and is recruited to phagosomes. Association of APOL7C with phagosomes led to phagosomal rupture and escape of engulfed antigens to the cytosol, where they could be processed via the endogenous MHC class I antigen processing pathway. Accordingly, mice deficient in APOL7C did not efficiently prime CD8+ T cells in response to immunization with bead-bound and cell-associated antigens. Together, our data indicate the presence of dedicated apolipoproteins that mediate the delivery of phagocytosed proteins to the cytosol of activated cDCs to facilitate XP.
Collapse
Affiliation(s)
- Gerone A Gonzales
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Song Huang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Liam Wilkinson
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jenny A Nguyen
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Saif Sikdar
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Cécile Piot
- Immunobiology Laboratory, Francis Crick Institute, London, UK
| | - Victor Naumenko
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
| | - Jahanara Rajwani
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Cassandra M Wood
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Irene Dinh
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Melanie Moore
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eymi Cedeño
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Neil McKenna
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Maria J Polyak
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Sara Amidian
- Cell Imaging Core, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Nicole L Rosin
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matheus B Carneiro
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Bas Surewaard
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Nathan C Peters
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Christopher H Mody
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Douglas J Mahoney
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
| | - Johnathan Canton
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Calgary, Alberta, Canada
| |
Collapse
|
3
|
Becker S, Swoboda A, Siemer H, Schimmelpfennig S, Sargin S, Shahin V, Schwab A, Najder K. Membrane potential dynamics of C5a-stimulated neutrophil granulocytes. Pflugers Arch 2024; 476:1007-1018. [PMID: 38613695 PMCID: PMC11139730 DOI: 10.1007/s00424-024-02947-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/15/2024]
Abstract
Neutrophil granulocytes play a crucial role in host defense against invading pathogens and in inflammatory diseases. The aim of this study was to elucidate membrane potential dynamics during the initial phase of neutrophil activation and its relation to migration and production of reactive oxygen species (ROS). We performed ROS production measurements of neutrophils from healthy C57BL/6J mice after TNFα-priming and/or C5a stimulation. The actin cytoskeleton was visualized with fluorescence microscopy. Furthermore, we combined migration assays and measurements of membrane potential dynamics after stimulating unprimed and/or TNFα-primed neutrophils with C5a. We show that C5a has a concentration-dependent effect on ROS production and chemokinetic migration. Chemokinetic migration and chemotaxis are impaired at C5a concentrations that induce ROS production. The actin cytoskeleton of unstimulated and of ROS-producing neutrophils is not distributed in a polarized way. Inhibition of the phagocytic NADPH oxidase NOX2 with diphenyleneiodonium (DPI) leads to a polarized distribution of the actin cytoskeleton and rescues chemokinetic migration of primed and C5a-stimulated neutrophils. Moreover, C5a evokes a pronounced depolarization of the cell membrane potential by 86.6 ± 4.2 mV starting from a resting membrane potential of -74.3 ± 0.7 mV. The C5a-induced depolarization occurs almost instantaneously (within less than one minute) in contrast to the more gradually developing depolarization induced by PMA (lag time of 3-4 min). This initial depolarization is accompanied by a decrease of the migration velocity. Collectively, our results show that stimulation with C5a evokes parallel changes in membrane potential dynamics, neutrophil ROS production and motility. Notably, the amplitude of membrane potential dynamics is comparable to that of excitable cells.
Collapse
Affiliation(s)
- Stina Becker
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Aljoscha Swoboda
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Henrik Siemer
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | | | - Sarah Sargin
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Victor Shahin
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University Hospital Münster, Münster, Germany.
| | - Karolina Najder
- Institute of Physiology II, University Hospital Münster, Münster, Germany.
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany.
| |
Collapse
|
4
|
DeCoursey TE. Transcendent Aspects of Proton Channels. Annu Rev Physiol 2024; 86:357-377. [PMID: 37931166 PMCID: PMC10938948 DOI: 10.1146/annurev-physiol-042222-023242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
A handful of biological proton-selective ion channels exist. Some open at positive or negative membrane potentials, others open at low or high pH, and some are light activated. This review focuses on common features that result from the unique properties of protons. Proton conduction through water or proteins differs qualitatively from that of all other ions. Extraordinary proton selectivity is needed to ensure that protons permeate and other ions do not. Proton selectivity arises from a proton pathway comprising a hydrogen-bonded chain that typically includes at least one titratable amino acid side chain. The enormously diverse functions of proton channels in disparate regions of the phylogenetic tree can be summarized by considering the chemical and electrical consequences of proton flux across membranes. This review discusses examples of cells in which proton efflux serves to increase pHi, decrease pHo, control the membrane potential, generate action potentials, or compensate transmembrane movement of electrical charge.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois, USA;
| |
Collapse
|
5
|
Kopach O, Sylantyev S, Bard L, Michaluk P, Heller JP, Gutierrez del Arroyo A, Ackland GL, Gourine AV, Rusakov DA. Human neutrophils communicate remotely via calcium-dependent glutamate-induced glutamate release. iScience 2023; 26:107236. [PMID: 37496680 PMCID: PMC10366500 DOI: 10.1016/j.isci.2023.107236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/25/2023] [Accepted: 06/23/2023] [Indexed: 07/28/2023] Open
Abstract
Neutrophils are white blood cells that are critical to acute inflammatory and adaptive immune responses. Their swarming-pattern behavior is controlled by multiple cellular cascades involving calcium-dependent release of various signaling molecules. Previous studies have reported that neutrophils express glutamate receptors and can release glutamate but evidence of direct neutrophil-neutrophil communication has been elusive. Here, we hold semi-suspended cultured human neutrophils in patch-clamp whole-cell mode to find that calcium mobilization induced by stimulating one neutrophil can trigger an N-methyl-D-aspartate (NMDA) receptor-driven membrane current and calcium signal in neighboring neutrophils. We employ an enzymatic-based imaging assay to image, in real time, glutamate release from neutrophils induced by glutamate released from their neighbors. These observations provide direct evidence for a positive-feedback inter-neutrophil communication that could contribute to mechanisms regulating communal neutrophil behavior.
Collapse
Affiliation(s)
- Olga Kopach
- Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Sergyi Sylantyev
- Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
- Rowett Institute, University of Aberdeen, Ashgrove Road West, Aberdeen AB25 2ZD, UK
| | - Lucie Bard
- Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Piotr Michaluk
- Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
- BRAINCITY, Laboratory of Neurobiology, Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Janosch P. Heller
- Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
- School of Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Ana Gutierrez del Arroyo
- Translational Medicine and Therapeutics, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Gareth L. Ackland
- Translational Medicine and Therapeutics, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Alexander V. Gourine
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Dmitri A. Rusakov
- Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
6
|
Stratmann AEP, Wohlgemuth L, Erber ME, Bernhard S, Hug S, Fauler M, Vidoni L, Mohamed AOK, Thomaß BD, Münnich F, Stukan L, Föhr KJ, Mannes M, Huber-Lang MS, Messerer DAC. Simultaneous Measurement of Changes in Neutrophil Granulocyte Membrane Potential, Intracellular pH, and Cell Size by Multiparametric Flow Cytometry. Biomedicines 2021; 9:1504. [PMID: 34829733 PMCID: PMC8614908 DOI: 10.3390/biomedicines9111504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 01/10/2023] Open
Abstract
Neutrophils provide rapid and efficient defense mechanisms against invading pathogens. Upon stimulation with proinflammatory mediators, including complement factors and bacterial peptides, neutrophils respond with changes in their membrane potential, intracellular pH, and cellular size. This study provides an approach to quantify these important changes simultaneously using multiparametric flow cytometry, thereby revealing a typical sequence of neutrophil activation consisting of depolarization, alkalization, and increase in cellular size. Additionally, the time resolution of the flow cytometric measurement is improved in order to allow changes that occur within seconds to be monitored, and thus to enhance the kinetic analysis of the neutrophil response. The method is appropriate for the reliable semiquantitative detection of small variations with respect to an increase, no change, and decrease in those parameters as demonstrated by the screening of various proinflammatory mediators. As a translational outlook, the findings are put into context in inflammatory conditions in vitro as well as in a clinically relevant whole blood model of endotoxemia. Taken together, the multiparametric analysis of neutrophil responsiveness regarding depolarization, alkalization, and changes in cellular size may contribute to a better understanding of neutrophils in health and disease, thus potentially yielding innovative mechanistic insights and possible novel diagnostic and/or prognostic approaches.
Collapse
Affiliation(s)
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Maike Elisabeth Erber
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Stefan Bernhard
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Stefan Hug
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany
| | - Laura Vidoni
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Adam Omar Khalaf Mohamed
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Bertram Dietrich Thomaß
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Frederik Münnich
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Laura Stukan
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| | - Karl Josef Föhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Markus Stefan Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
7
|
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
|
8
|
Hug S, Bernhard S, Stratmann AEP, Erber M, Wohlgemuth L, Knapp CL, Bauer JM, Vidoni L, Fauler M, Föhr KJ, Radermacher P, Hoffmann A, Huber-Lang M, Messerer DAC. Activation of Neutrophil Granulocytes by Platelet-Activating Factor Is Impaired During Experimental Sepsis. Front Immunol 2021; 12:642867. [PMID: 33796110 PMCID: PMC8007865 DOI: 10.3389/fimmu.2021.642867] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Platelet-activating factor (PAF) is an important mediator of the systemic inflammatory response. In the case of sepsis, proper activation and function of neutrophils as the first line of cellular defense are based on a well-balanced physiological response. However, little is known about the role of PAF in cellular changes of neutrophils during sepsis. Therefore, this study investigates the reaction patterns of neutrophils induced by PAF with a focus on membrane potential (MP), intracellular pH, and cellular swelling under physiological and pathophysiological conditions and hypothesizes that the PAF-mediated response of granulocytes is altered during sepsis. The cellular response of granulocytes including MP, intracellular pH, cellular swelling, and other activation markers were analyzed by multiparametric flow cytometry. In addition, the chemotactic activity and the formation of platelet-neutrophil complexes after exposure to PAF were investigated. The changes of the (electro-)physiological response features were translationally verified in a human ex vivo whole blood model of endotoxemia as well as during polymicrobial porcine sepsis. In neutrophils from healthy human donors, PAF elicited a rapid depolarization, an intracellular alkalization, and an increase in cell size in a time- and dose-dependent manner. Mechanistically, the alkalization was dependent on sodium-proton exchanger 1 (NHE1) activity, while the change in cellular shape was sodium flux- but only partially NHE1-dependent. In a pathophysiological altered environment, the PAF-induced response of neutrophils was modulated. Acidifying the extracellular pH in vitro enhanced PAF-mediated depolarization, whereas the increases in cell size and intracellular pH were largely unaffected. Ex vivo exposure of human whole blood to lipopolysaccharide diminished the PAF-induced intracellular alkalization and the change in neutrophil size. During experimental porcine sepsis, depolarization of the MP was significantly impaired. Additionally, there was a trend for increased cellular swelling, whereas intracellular alkalization remained stable. Overall, an impaired (electro-)physiological response of neutrophils to PAF stimulation represents a cellular hallmark of those cells challenged during systemic inflammation. Furthermore, this altered response may be indicative of and causative for the development of neutrophil dysfunction during sepsis.
Collapse
Affiliation(s)
- Stefan Hug
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Stefan Bernhard
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | | | - Maike Erber
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christiane Leonie Knapp
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Jonas Martin Bauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Laura Vidoni
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, University of Ulm, Ulm, Germany
| | - Karl Josef Föhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Development, University Hospital of Ulm, Ulm, Germany
| | - Andrea Hoffmann
- Institute of Anesthesiological Pathophysiology and Process Development, University Hospital of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany.,Institute of Anesthesiological Pathophysiology and Process Development, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
9
|
Messerer DAC, Schmidt H, Frick M, Huber-Lang M. Ion and Water Transport in Neutrophil Granulocytes and Its Impairment during Sepsis. Int J Mol Sci 2021; 22:1699. [PMID: 33567720 PMCID: PMC7914618 DOI: 10.3390/ijms22041699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Neutrophil granulocytes are the vanguard of innate immunity in response to numerous pathogens. Their activity drives the clearance of microbe- and damage-associated molecular patterns, thereby contributing substantially to the resolution of inflammation. However, excessive stimulation during sepsis leads to cellular unresponsiveness, immunological dysfunction, bacterial expansion, and subsequent multiple organ dysfunction. During the short lifespan of neutrophils, they can become significantly activated by complement factors, cytokines, and other inflammatory mediators. Following stimulation, the cells respond with a defined (electro-)physiological pattern, including depolarization, calcium influx, and alkalization as well as with increased metabolic activity and polarization of the actin cytoskeleton. Activity of ion transport proteins and aquaporins is critical for multiple cellular functions of innate immune cells, including chemotaxis, generation of reactive oxygen species, and phagocytosis of both pathogens and tissue debris. In this review, we first describe the ion transport proteins and aquaporins involved in the neutrophil ion-water fluxes in response to chemoattractants. We then relate ion and water flux to cellular functions with a focus on danger sensing, chemotaxis, phagocytosis, and oxidative burst and approach the role of altered ion transport protein expression and activity in impaired cellular functions and cell death during systemic inflammation as in sepsis.
Collapse
Affiliation(s)
- David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, 89081 Ulm, Germany;
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, 89081 Ulm, Germany
| | - Hanna Schmidt
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany; (H.S.); (M.F.)
- Department of Pediatrics and Adolescent Medicine, University Hospital of Ulm, 89081 Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany; (H.S.); (M.F.)
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, 89081 Ulm, Germany;
| |
Collapse
|
10
|
Cantin AM, Ouellet C, Cloutier A, McDonald PP. Airway Mucins Inhibit Oxidative and Non-Oxidative Bacterial Killing by Human Neutrophils. Front Pharmacol 2020; 11:554353. [PMID: 33101020 PMCID: PMC7554606 DOI: 10.3389/fphar.2020.554353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/11/2020] [Indexed: 11/13/2022] Open
Abstract
Neutrophil killing of bacteria is mediated by oxidative and non-oxidative mechanisms. Oxidants are generated through the NADPH oxidase complex, whereas antimicrobial proteins and peptides rank amongst non-oxidative host defenses. Mucus hypersecretion, deficient hydration and poor clearance from the airways are prominent features of cystic fibrosis (CF) lung disease. CF airways are commonly infected by Pseudomonas aeruginosa and Burkholderia cepacia complex bacteria. Whereas the former bacterium is highly sensitive to non-oxidative killing, the latter is only killed if the oxidative burst is intact. Despite an abundance of neutrophils, both pathogens thrive in CF airway secretions. In this study, we report that secreted mucins protect these CF pathogens against host defenses. Mucins were purified from CF sputum and from the saliva of healthy volunteers. Whereas mucins did not alter the phagocytosis of Pseudomonas aeruginosa and Burkholderia cenocepacia by neutrophils, they completely suppressed bacterial killing. Accordingly, mucins markedly inhibited non-oxidative bacterial killing by neutrophil granule extracts, or by lysozyme and the cationic peptide, human β defensin-2 (HBD2). Mucins also suppressed the neutrophil oxidative burst through a charge-dependent mechanism that could be reversed by the cationic aminoglycoside, tobramycin. Our data indicate that airway mucins protect Gram-negative bacteria against neutrophil killing by suppressing the oxidative burst and inhibiting the bactericidal capacity of cationic proteins and peptides. Mucin hypersecretion, dehydration, stasis and anionic charge represent key therapeutic targets for improving host defenses and airway inflammation in CF and other muco-secretory airway diseases.
Collapse
Affiliation(s)
- André M. Cantin
- Pulmonary Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|
11
|
Najder K, Rugi M, Lebel M, Schröder J, Oster L, Schimmelpfennig S, Sargin S, Pethő Z, Bulk E, Schwab A. Role of the Intracellular Sodium Homeostasis in Chemotaxis of Activated Murine Neutrophils. Front Immunol 2020; 11:2124. [PMID: 33013896 PMCID: PMC7506047 DOI: 10.3389/fimmu.2020.02124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022] Open
Abstract
The importance of the intracellular Ca2+ concentration ([Ca2+]i) in neutrophil function has been intensely studied. However, the role of the intracellular Na+ concentration ([Na+]i) which is closely linked to the intracellular Ca2+ regulation has been largely overlooked. The [Na+]i is regulated by Na+ transport proteins such as the Na+/Ca2+-exchanger (NCX1), Na+/K+-ATPase, and Na+-permeable, transient receptor potential melastatin 2 (TRPM2) channel. Stimulating with either N-formylmethionine-leucyl-phenylalanine (fMLF) or complement protein C5a causes distinct changes of the [Na+]i. fMLF induces a sustained increase of [Na+]i, surprisingly, reaching higher values in TRPM2-/- neutrophils. This outcome is unexpected and remains unexplained. In both genotypes, C5a elicits only a transient rise of the [Na+]i. The difference in [Na+]i measured at t = 10 min after stimulation is inversely related to neutrophil chemotaxis. Neutrophil chemotaxis is more efficient in C5a than in an fMLF gradient. Moreover, lowering the extracellular Na+ concentration from 140 to 72 mM improves chemotaxis of WT but not of TRPM2-/- neutrophils. Increasing the [Na+]i by inhibiting the Na+/K+-ATPase results in disrupted chemotaxis. This is most likely due to the impact of the altered Na+ homeostasis and presumably NCX1 function whose expression was shown by means of qPCR and which critically relies on proper extra- to intracellular Na+ concentration gradients. Increasing the [Na+]i by a few mmol/l may suffice to switch its transport mode from forward (Ca2+-efflux) to reverse (Ca2+-influx) mode. The role of NCX1 in neutrophil chemotaxis is corroborated by its blocker, which also causes a complete inhibition of chemotaxis.
Collapse
Affiliation(s)
- Karolina Najder
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Micol Rugi
- Institute of Physiology II, University Hospital Münster, Münster, Germany
- University of Florence, Florence, Italy
| | - Mégane Lebel
- University of Sherbrooke, Sherbrooke, QC, Canada
| | - Julia Schröder
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Leonie Oster
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | | | - Sarah Sargin
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Zoltán Pethő
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Etmar Bulk
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| |
Collapse
|
12
|
CGD: less is more. Blood 2020; 135:883-884. [PMID: 32191802 DOI: 10.1182/blood.2020005062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
13
|
Complement C5a Alters the Membrane Potential of Neutrophils during Hemorrhagic Shock. Mediators Inflamm 2018; 2018:2052356. [PMID: 30002598 PMCID: PMC5996468 DOI: 10.1155/2018/2052356] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/26/2018] [Accepted: 04/01/2018] [Indexed: 12/17/2022] Open
Abstract
Background Polymorphonuclear granulocytes (PMN) play a crucial role in host defense. Physiologically, exposure of PMN to the complement activation product C5a results in a protective response against pathogens, whereas in the case of systemic inflammation, excessive C5a substantially impairs neutrophil functions. To further elucidate the inability of PMN to properly respond to C5a, this study investigates the role of the cellular membrane potential of PMN in response to C5a. Methods Electrophysiological changes in cellular and mitochondrial membrane potential and intracellular pH of PMN from human healthy volunteers were determined by flow cytometry after exposure to C5a. Furthermore, PMN from male Bretoncelles-Meishan-Willebrand cross-bred pigs before and three hours after severe hemorrhagic shock were analyzed for their electrophysiological response. Results PMN showed a significant dose- and time-dependent depolarization in response to C5a with a strong response after one minute. The chemotactic peptide fMLP also evoked a significant shift in the membrane potential of PMN. Acidification of the cellular microenvironment significantly enhanced depolarization of PMN. In a clinically relevant model of porcine hemorrhagic shock, the C5a-induced changes in membrane potential of PMN were markedly diminished compared to healthy littermates. Overall, these membrane potential changes may contribute to PMN dysfunction in an inflammatory environment.
Collapse
|
14
|
DeCoursey TE. Voltage and pH sensing by the voltage-gated proton channel, H V1. J R Soc Interface 2018; 15:20180108. [PMID: 29643227 PMCID: PMC5938591 DOI: 10.1098/rsif.2018.0108] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 03/19/2018] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated proton channels are unique ion channels, membrane proteins that allow protons but no other ions to cross cell membranes. They are found in diverse species, from unicellular marine life to humans. In all cells, their function requires that they open and conduct current only under certain conditions, typically when the electrochemical gradient for protons is outwards. Consequently, these proteins behave like rectifiers, conducting protons out of cells. Their activity has electrical consequences and also changes the pH on both sides of the membrane. Here we summarize what is known about the way these proteins sense the membrane potential and the pH inside and outside the cell. Currently, it is hypothesized that membrane potential is sensed by permanently charged arginines (with very high pKa) within the protein, which results in parts of the protein moving to produce a conduction pathway. The mechanism of pH sensing appears to involve titratable side chains of particular amino acids. For this purpose their pKa needs to be within the operational pH range. We propose a 'counter-charge' model for pH sensing in which electrostatic interactions within the protein are selectively disrupted by protonation of internally or externally accessible groups.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, 1750 West Harrison, Chicago, IL 60612, USA
| |
Collapse
|
15
|
Buvelot H, Posfay-Barbe KM, Linder P, Schrenzel J, Krause KH. Staphylococcus aureus, phagocyte NADPH oxidase and chronic granulomatous disease. FEMS Microbiol Rev 2017; 41:139-157. [PMID: 27965320 DOI: 10.1093/femsre/fuw042] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2016] [Indexed: 11/14/2022] Open
Abstract
Dysfunction of phagocytes is a relevant risk factor for staphylococcal infection. The most common hereditary phagocyte dysfunction is chronic granulomatous disease (CGD), characterized by impaired generation of reactive oxygen species (ROS) due to loss of function mutations within the phagocyte NADPH oxidase NOX2. Phagocytes ROS generation is fundamental to eliminate pathogens and to regulate the inflammatory response to infection. CGD is characterized by recurrent and severe bacterial and fungal infections, with Staphylococcus aureus as the most frequent pathogen, and skin and lung abscesses as the most common clinical entities. Staphylococcus aureus infection may occur in virtually any human host, presumably because of the many virulence factors of the bacterium. However, in the presence of functional NOX2, staphylococcal infections remain rare and are mainly linked to breaches of the skin barrier. In contrast, in patients with CGD, S. aureus readily survives and frequently causes clinically apparent disease. Astonishingly, little is known why S. aureus, which possesses a wide range of antioxidant enzymes (e.g. catalase, SOD), is particularly sensitive to control through NOX2. In this review, we will evaluate the discovery of CGD and our present knowledge of the role of NOX2 in S. aureus infection.
Collapse
Affiliation(s)
- Helene Buvelot
- Division of General Internal Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland
| | - Klara M Posfay-Barbe
- Paediatric Infectious Diseases Unit, Department of Paediatrics, University Hospitals of Geneva, 1205 Geneva and Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Patrick Linder
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Jacques Schrenzel
- Divisions of Infectious Diseases and Laboratory Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland
| | - Karl-Heinz Krause
- Divisions of Infectious Diseases and Laboratory Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| |
Collapse
|
16
|
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
|
17
|
Vorobjeva N, Prikhodko A, Galkin I, Pletjushkina O, Zinovkin R, Sud'ina G, Chernyak B, Pinegin B. Mitochondrial reactive oxygen species are involved in chemoattractant-induced oxidative burst and degranulation of human neutrophils in vitro. Eur J Cell Biol 2017; 96:254-265. [PMID: 28325500 DOI: 10.1016/j.ejcb.2017.03.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 03/04/2017] [Accepted: 03/04/2017] [Indexed: 12/23/2022] Open
Abstract
Activation of neutrophils is accompanied by the oxidative burst, exocytosis of various granule types (degranulation) and a delay in spontaneous apoptosis. The major source of reactive oxygen species (ROS) in human neutrophils is NADPH oxidase (NOX2), however, other sources of ROS also exist. Although the function of ROS is mainly defensive, they can also play a regulatory role in cell signaling. However, the contribution of various sources of ROS in these processes is not clear. We investigated a possible role of mitochondria-derived ROS (mtROS) in the regulation of neutrophil activation induced by chemoattractant fMLP in vitro. Using the mitochondria-targeted antioxidant SkQ1, we demonstrated that mtROS are implicated in the oxidative burst caused by NOX2 activation as well as in the exocytosis of primary (azurophil) and secondary (specific) granules. Scavenging of mtROS with SkQ1 slightly accelerated spontaneous apoptosis and significantly stimulated apoptosis of fMLP-activated neutrophils. These data indicate that mtROS play a critical role in signal transduction that mediates the major neutrophil functional responses in the process of activation.
Collapse
Affiliation(s)
- Nina Vorobjeva
- Department Immunology, Biology Faculty; Lomonosov Moscow State University, 119998 Moscow, Russia; Institute of Immunology, FMBA, Kashirskoe Shosse 24/2, 115478, Moscow, Russia.
| | - Anastasia Prikhodko
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ivan Galkin
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Olga Pletjushkina
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Roman Zinovkin
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Galina Sud'ina
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Boris Chernyak
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Boris Pinegin
- Institute of Immunology, FMBA, Kashirskoe Shosse 24/2, 115478, Moscow, Russia
| |
Collapse
|
18
|
Seredenina T, Demaurex N, Krause KH. Voltage-Gated Proton Channels as Novel Drug Targets: From NADPH Oxidase Regulation to Sperm Biology. Antioxid Redox Signal 2015; 23:490-513. [PMID: 24483328 PMCID: PMC4543398 DOI: 10.1089/ars.2013.5806] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
SIGNIFICANCE Voltage-gated proton channels are increasingly implicated in cellular proton homeostasis. Proton currents were originally identified in snail neurons less than 40 years ago, and subsequently shown to play an important auxiliary role in the functioning of reactive oxygen species (ROS)-generating nicotinamide adenine dinucleotide phosphate (NADPH) oxidases. Molecular identification of voltage-gated proton channels was achieved less than 10 years ago. Interestingly, so far, only one gene coding for voltage-gated proton channels has been identified, namely hydrogen voltage-gated channel 1 (HVCN1), which codes for the HV1 proton channel protein. Over the last years, the first picture of putative physiological functions of HV1 has been emerging. RECENT ADVANCES The best-studied role remains charge and pH compensation during the respiratory burst of the phagocyte NADPH oxidase (NOX). Strong evidence for a role of HV1 is also emerging in sperm biology, but the relationship with the sperm NOX5 remains unclear. Probably in many instances, HV1 functions independently of NOX: for example in snail neurons, basophils, osteoclasts, and cancer cells. CRITICAL ISSUES Generally, ion channels are good drug targets; however, this feature has so far not been exploited for HV1, and hitherto no inhibitors compatible with clinical use exist. However, there are emerging indications for HV1 inhibitors, ranging from diseases with a strong activation of the phagocyte NOX (e.g., stroke) to infertility, osteoporosis, and cancer. FUTURE DIRECTIONS Clinically useful HV1-active drugs should be developed and might become interesting drugs of the future.
Collapse
Affiliation(s)
- Tamara Seredenina
- 1 Department of Pathology and Immunology, Geneva University Medical Faculty , Centre Médical Universitaire, Geneva, Switzerland
| | - Nicolas Demaurex
- 2 Department of Cellular Physiology and Metabolism, Geneva University Medical Faculty , Centre Médical Universitaire, Geneva, Switzerland
| | - Karl-Heinz Krause
- 1 Department of Pathology and Immunology, Geneva University Medical Faculty , Centre Médical Universitaire, Geneva, Switzerland .,3 Department of Genetic and Laboratory Medicine, Geneva University Hospitals , Centre Médical Universitaire, Geneva, Switzerland
| |
Collapse
|
19
|
Herrmann JM, Meyle J. Neutrophil activation and periodontal tissue injury. Periodontol 2000 2015; 69:111-27. [DOI: 10.1111/prd.12088] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
Wen D, Liu D, Tang J, Dong L, Liu Y, Tao Z, Wan J, Gao D, Wang L, Sun H, Fan J, Wu W. Malic enzyme 1 induces epithelial-mesenchymal transition and indicates poor prognosis in hepatocellular carcinoma. Tumour Biol 2015; 36:6211-21. [PMID: 25753478 DOI: 10.1007/s13277-015-3306-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/02/2015] [Indexed: 12/20/2022] Open
Abstract
Malic enzyme 1 (ME1) links the glycolytic and citric acid cycles and is important for NADPH production, glutamine metabolism, and lipogenesis. Recently, its deregulation has been implicated in the progression of various cancers. However, the role of ME1 in the progression of hepatocellular carcinoma (HCC) remains unclear. In this study, we utilized short hairpin RNA-mediated gene silencing to investigate the biological effects of ME1 depletion in HCC and determined its prognostic significance in HCC. ME1 expression was examined by real-time (RT)-PCR and Western blot using five HCC cell lines and one normal liver cell line. We used polyethylenimine nanoparticles to deliver a short hairpin RNA to induce cessation of ME1 expression in HCC cells. Changes in NADPH production and reactive oxygen species (ROS) production were studied. Metastatic potentials of HCC cells were evaluated in vitro. Furthermore, we evaluated the protein level of ME1 in para-tumor and cancerous tissues of 65 HCC patients with detailed clinical, pathological, and clinical follow-up data. Patients' survivals were further assessed as well. Upregulated ME1 expression was observed in HCC cell lines. Downregulation of ME1 attenuated NADPH production and stimulated ROS production. Silencing ME1 was noted to inhibit migratory and invasive properties of HCC cells by inducing the E-cadherin expression and decreasing of N-cadherin and vimentin expression in a ROS-dependent pathway. Overexpression of ME1 was observed in a major fraction of HCC samples. Higher level of ME1 in tumors was significantly associated with reduced overall survival (Kaplan-Meier analysis, P = 0.024) and reduced progression-free survival (Kaplan-Meier analysis, P = 0.011). Inhibition of ME1 expression decreases HCC metastasis via suppression of epithelial-mesenchymal transition (EMT) processes in ROS-induced pathways. ME1 overexpression associates with unfavorable prognoses in patients with HCC, suggesting that ME1 is a poor prognostic predictor of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Duo Wen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 2000032, China,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Nauseef WM, Borregaard N. Neutrophils at work. Nat Immunol 2014; 15:602-11. [PMID: 24940954 DOI: 10.1038/ni.2921] [Citation(s) in RCA: 648] [Impact Index Per Article: 58.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/13/2014] [Indexed: 12/12/2022]
Abstract
In this Review we discuss data demonstrating recently recognized aspects of neutrophil homeostasis in the steady state, granulopoiesis in 'emergency' conditions and interactions of neutrophils with the adaptive immune system. We explore in vivo observations of the recruitment of neutrophils from blood to tissues in models of blood-borne infections versus bacterial invasion through epithelial linings. We examine data on novel aspects of the activation of NADPH oxidase and the heterogeneity of phagosomes and, finally, consider the importance of two neutrophil-derived biological agents: neutrophil extracellular traps and ectosomes.
Collapse
Affiliation(s)
- William M Nauseef
- Inflammation Program, Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, and Veterans Administration Medical Center, Iowa City, Iowa, USA
| | - Niels Borregaard
- The Granulocyte Research Laboratory, Department of Hematology, National University Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Bao Y, Ledderose C, Seier T, Graf AF, Brix B, Chong E, Junger WG. Mitochondria regulate neutrophil activation by generating ATP for autocrine purinergic signaling. J Biol Chem 2014; 289:26794-26803. [PMID: 25104353 DOI: 10.1074/jbc.m114.572495] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) form the first line of defense against invading microorganisms. We have shown previously that ATP release and autocrine purinergic signaling via P2Y2 receptors are essential for PMN activation. Here we show that mitochondria provide the ATP that initiates PMN activation. Stimulation of formyl peptide receptors increases the mitochondrial membrane potential (Δψm) and triggers a rapid burst of ATP release from PMNs. This burst of ATP release can be blocked by inhibitors of mitochondrial ATP production and requires an initial formyl peptide receptor-induced Ca(2+) signal that triggers mitochondrial activation. The burst of ATP release generated by the mitochondria fuels a first phase of purinergic signaling that boosts Ca(2+) signaling, amplifies mitochondrial ATP production, and initiates functional PMN responses. Cells then switch to glycolytic ATP production, which fuels a second round of purinergic signaling that sustains Ca(2+) signaling via P2X receptor-mediated Ca(2+) influx and maintains functional PMN responses such as oxidative burst, degranulation, and phagocytosis.
Collapse
Affiliation(s)
- Yi Bao
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Thomas Seier
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,; Paracelsus Medical University, Salzburg A-5020, Austria, and
| | - Amelie F Graf
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,; Paracelsus Medical University, Salzburg A-5020, Austria, and
| | - Bianca Brix
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Eritza Chong
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,; Ludwig Boltzmann Institute for Traumatology, Vienna A-1200, Austria.
| |
Collapse
|
23
|
Kovács I, Horváth M, Kovács T, Somogyi K, Tretter L, Geiszt M, Petheő GL. Comparison of proton channel, phagocyte oxidase, and respiratory burst levels between human eosinophil and neutrophil granulocytes. Free Radic Res 2014; 48:1190-9. [DOI: 10.3109/10715762.2014.938234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
Tsutsui H, Jinno Y, Tomita A, Okamura Y. Rapid evaluation of a protein-based voltage probe using a field-induced membrane potential change. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:1730-7. [DOI: 10.1016/j.bbamem.2014.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/27/2014] [Accepted: 03/10/2014] [Indexed: 10/25/2022]
|
25
|
DeCoursey TE, Hosler J. Philosophy of voltage-gated proton channels. J R Soc Interface 2014; 11:20130799. [PMID: 24352668 PMCID: PMC3899857 DOI: 10.1098/rsif.2013.0799] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 11/22/2013] [Indexed: 02/02/2023] Open
Abstract
In this review, voltage-gated proton channels are considered from a mainly teleological perspective. Why do proton channels exist? What good are they? Why did they go to such lengths to develop several unique hallmark properties such as extreme selectivity and ΔpH-dependent gating? Why is their current so minuscule? How do they manage to be so selective? What is the basis for our belief that they conduct H(+) and not OH(-)? Why do they exist in many species as dimers when the monomeric form seems to work quite well? It is hoped that pondering these questions will provide an introduction to these channels and a way to logically organize their peculiar properties as well as to understand how they are able to carry out some of their better-established biological functions.
Collapse
Affiliation(s)
- Thomas E. DeCoursey
- Department of Molecular Biophysics and Physiology, Rush University, 1750 West Harrison, Chicago, IL 60612, USA
| | - Jonathan Hosler
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
26
|
Abstract
The past decade has seen increasing use of the patch-clamp technique on neutrophils and eosinophils. The main goal of these electrophysiological studies has been to elucidate the mechanisms underlying the phagocyte respiratory burst. NADPH oxidase activity, which defines the respiratory burst in granulocytes, is electrogenic because electrons from NADPH are transported across the cell membrane, where they reduce oxygen to form superoxide anion (O2 (-)). This passage of electrons comprises an electrical current that would rapidly depolarize the membrane if the charge movement were not balanced by proton efflux. The patch-clamp technique enables simultaneous recording of NADPH oxidase-generated electron current and H(+) flux through the closely related H(+) channel. Increasing evidence suggests that other ion channels may play crucial roles in degranulation, phagocytosis, and chemotaxis, highlighting the importance of electrophysiological studies to advance knowledge of granulocyte function. Several configurations of the patch-clamp technique exist. Each has advantages and limitations that are discussed here. Meaningful measurements of ion channels cannot be achieved without an understanding of their fundamental properties. We describe the types of measurements that are necessary to characterize a particular ion channel.
Collapse
|
27
|
Microglial voltage-gated proton channel Hv1 in ischemic stroke. Transl Stroke Res 2013; 5:99-108. [PMID: 24323712 DOI: 10.1007/s12975-013-0289-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/11/2013] [Accepted: 09/19/2013] [Indexed: 12/22/2022]
Abstract
Microglia, resident immune cells in the brain, contribute both to the damage and resolution of ischemic stroke. However, the mechanisms of microglia's detrimental or beneficial role in the disease are poorly understood. The voltage-gated proton channel, Hv1, rapidly removes protons from depolarized cytoplasm, and is highly expressed in the immune system. In the brain, Hv1 is selectively and functionally expressed in microglia but not neurons. Although the physiological function of microglial Hv1 is still not clear, Hv1 is one of major ion channels expressed in resting microglia. Under pathological conditions, microglial Hv1 is required for NADPH oxidase (NOX)-dependent generation of reactive oxygen species (ROS) by providing charge compensation for exported electrons and relieving intracellular acidosis. In a mouse model of cerebral middle artery occlusion, Hv1 knockout mice are protected from ischemic damage, showing reduced NOX-dependent ROS production, microglial activation and neuronal cell death. Therefore, microglial Hv1 aids in NOX-dependent ROS generation, which subsequently induces neuronal cell death and a significant fraction of brain damage after ischemic stroke. These studies illuminate a critical role of microglial Hv1 in ischemic brain injury, providing a rationale for Hv1 as a potential therapeutic target for the treatment of ischemic stroke. The current understanding of Hv1 in ischemic injury through NOX-dependent ROS production may serve as a common model to reveal the deleterious role of microglia in neurological diseases other than ischemic stroke, such as multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, and neuropathic pain.
Collapse
|
28
|
Zhu X, Mose E, Zimmermann N. Proton channel HVCN1 is required for effector functions of mouse eosinophils. BMC Immunol 2013; 14:24. [PMID: 23705768 PMCID: PMC3668235 DOI: 10.1186/1471-2172-14-24] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/20/2013] [Indexed: 01/21/2023] Open
Abstract
Background Proton currents are required for optimal respiratory burst in phagocytes. Recently, HVCN1 was identified as the molecule required for the voltage-gated proton channel activity associated with the respiratory burst in neutrophils. Although there are similarities between eosinophils and neutrophils regarding their mechanism for respiratory burst, the role of proton channels in eosinophil functions has not been fully understood. Results In the present study, we first identified the expression of the proton channel HVCN1 in mouse eosinophils. Furthermore, using HVCN1-deficient eosinophils, we demonstrated important cell-specific effector functions for HVCN1. Similar to HVCN1-deficient neutrophils, HVCN1-deficient eosinophils produced significantly less reactive oxygen species (ROS) upon phorbol myristate acetate (PMA) stimulation compared with WT eosinophils. In contrast to HVCN1-deficient neutrophils, HVCN1-deficient eosinophils did not show impaired calcium mobilization or migration ability compared with wild-type (WT) cells. Uniquely, HVCN1-deficient eosinophils underwent significantly increased cell death induced by PMA stimulation compared with WT eosinophils. The increased cell death was dependent on NADPH oxidase activation, and correlated with the failure of HVCN1-deficient cells to maintain membrane polarization and intracellular pH in the physiological range upon activation. Conclusions Eosinophils require proton channel HVCN1 for optimal ROS generation and prevention of activation-induced cell death.
Collapse
Affiliation(s)
- Xiang Zhu
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | | | |
Collapse
|
29
|
Membrane potential measurements of isolated neurons using a voltage-sensitive dye. PLoS One 2013; 8:e58260. [PMID: 23516458 PMCID: PMC3596405 DOI: 10.1371/journal.pone.0058260] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 02/01/2013] [Indexed: 01/31/2023] Open
Abstract
The ability to monitor changes in membrane potential is a useful tool for studying neuronal function, but there are only limited options available at present. Here, we have investigated the potential of a commercially available FLIPR membrane potential (FMP) dye, developed originally for high throughput screening using a plate reader, for imaging the membrane potential of cultured cells using an epifluorescence-based single cell imaging system. We found that the properties of the FMP dye make it highly suitable for such imaging since 1) its fluorescence displayed a high signal-to-noise ratio, 2) robust signals meant only minimal exposure times of around 5 ms were necessary, and 3) bidirectional changes in fluorescence were detectable resulting from hyper- or depolarising conditions, reaching equilibrium with a time constant of 4–8 s. Measurements were possible independently of whether membrane potential changes were induced by voltage clamping, or manipulating the ionic distribution of either Na+ or K+. Since FMP behaves as a charged molecule which accumulates in the cytosol, equations based on the Boltzmann distribution were developed determining that the apparent charge of FMP which represents a measure of the voltage sensitivity of the dye, is between −0.62 and −0.72. Finally, we demonstrated that FMP is suitable for use in a variety of neuronal cell types and detects membrane potential changes arising from spontaneous firing of action potentials and through stimulation with a variety of excitatory and inhibitory neurotransmitters.
Collapse
|
30
|
El Chemaly A, Demaurex N. Do Hv1 proton channels regulate the ionic and redox homeostasis of phagosomes? Mol Cell Endocrinol 2012; 353:82-7. [PMID: 22056415 DOI: 10.1016/j.mce.2011.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 10/06/2011] [Accepted: 10/06/2011] [Indexed: 12/28/2022]
Abstract
Recent work on animal models has revealed the important role played by the voltage-gated proton channel Hv1 during bacterial killing by innate immune cells. Studies from mice lacking Hv1 channels showed that Hv1 proton channels are required for high-level production of reactive oxygen species (ROS) by the NADPH oxidase of phagocytes (NOX2) in two ways. First, Hv1 channels maintain a physiological membrane potential during the respiratory burst of neutrophils by providing a compensating charge for the electrons transferred by NOX2 from NADPH to superoxide. Second, Hv1 channels maintain a physiological cytosolic pH by extruding the acid generated by the NOX2-dependent consumption of NADPH. The two mechanisms directly sustain the activity of the NOX2 enzyme and indirectly sustain other neutrophil functions by enhancing the driving force for the entry of calcium into cells, thereby boosting cellular calcium signals. The increased depolarization of Hv1-deficient neutrophils aborted calcium responses to chemoattractants and revealed adhesion and migration defects that were associated with an impaired depolymerization of the cortical actin cytoskeleton. Current research aims to transpose these findings to phagosomes, the phagocytic vacuoles where bacterial killing takes place. However, the mechanisms that control the phagosomal pH appear to vary greatly between phagocytes: phagosomes rapidly acidify in macrophages but remain neutral for several minutes in neutrophils following ingestion of solid particles, whereas in dendritic cells phagosomes alkalinize, a mechanism thought to promote antigen cross-presentation. In this review, we discuss how the knowledge gained on the role of Hv1 channels at the plasma membrane of neutrophils can be used to study the regulation of the phagosomal pH, ROS, membrane potential, and calcium fluxes in different phagocytic cells.
Collapse
Affiliation(s)
- Antoun El Chemaly
- Department of Cell Physiology and Metabolism, University of Geneva, 1211 Geneva 4, Switzerland
| | | |
Collapse
|
31
|
Abstract
Voltage-gated proton channels, HV1, have vaulted from the realm of the esoteric into the forefront of a central question facing ion channel biophysicists, namely, the mechanism by which voltage-dependent gating occurs. This transformation is the result of several factors. Identification of the gene in 2006 revealed that proton channels are homologues of the voltage-sensing domain of most other voltage-gated ion channels. Unique, or at least eccentric, properties of proton channels include dimeric architecture with dual conduction pathways, perfect proton selectivity, a single-channel conductance approximately 10(3) times smaller than most ion channels, voltage-dependent gating that is strongly modulated by the pH gradient, ΔpH, and potent inhibition by Zn(2+) (in many species) but an absence of other potent inhibitors. The recent identification of HV1 in three unicellular marine plankton species has dramatically expanded the phylogenetic family tree. Interest in proton channels in their own right has increased as important physiological roles have been identified in many cells. Proton channels trigger the bioluminescent flash of dinoflagellates, facilitate calcification by coccolithophores, regulate pH-dependent processes in eggs and sperm during fertilization, secrete acid to control the pH of airway fluids, facilitate histamine secretion by basophils, and play a signaling role in facilitating B-cell receptor mediated responses in B-lymphocytes. The most elaborate and best-established functions occur in phagocytes, where proton channels optimize the activity of NADPH oxidase, an important producer of reactive oxygen species. Proton efflux mediated by HV1 balances the charge translocated across the membrane by electrons through NADPH oxidase, minimizes changes in cytoplasmic and phagosomal pH, limits osmotic swelling of the phagosome, and provides substrate H(+) for the production of H2O2 and HOCl, reactive oxygen species crucial to killing pathogens.
Collapse
Affiliation(s)
- Thomas E Decoursey
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, Illinois, USA.
| |
Collapse
|
32
|
|
33
|
Amyloid-β-induced reactive oxygen species production and priming are differentially regulated by ion channels in microglia. J Cell Physiol 2011; 226:3295-302. [DOI: 10.1002/jcp.22675] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
34
|
DeCoursey TE. Voltage-gated proton channels find their dream job managing the respiratory burst in phagocytes. Physiology (Bethesda) 2010; 25:27-40. [PMID: 20134026 PMCID: PMC3023998 DOI: 10.1152/physiol.00039.2009] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The voltage-gated proton channel bears surprising resemblance to the voltage-sensing domain (S1-S4) of other voltage-gated ion channels but is a dimer with two conduction pathways. The proton channel seems designed for efficient proton extrusion from cells. In phagocytes, it facilitates the production of reactive oxygen species by NADPH oxidase.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, Illinois, USA.
| |
Collapse
|
35
|
El Chemaly A, Okochi Y, Sasaki M, Arnaudeau S, Okamura Y, Demaurex N. VSOP/Hv1 proton channels sustain calcium entry, neutrophil migration, and superoxide production by limiting cell depolarization and acidification. ACTA ACUST UNITED AC 2009; 207:129-39. [PMID: 20026664 PMCID: PMC2812533 DOI: 10.1084/jem.20091837] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Neutrophils kill microbes with reactive oxygen species generated by the NADPH oxidase, an enzyme which moves electrons across membranes. Voltage-gated proton channels (voltage-sensing domain only protein [VSOP]/Hv1) are required for high-level superoxide production by phagocytes, but the mechanism of this effect is not established. We show that neutrophils from VSOP/Hv1−/− mice lack proton currents but have normal electron currents, indicating that these cells have a fully functional oxidase that cannot conduct protons. VSOP/Hv1−/− neutrophils had a more acidic cytosol, were more depolarized, and produced less superoxide and hydrogen peroxide than neutrophils from wild-type mice. Hydrogen peroxide production was rescued by providing an artificial conductance with gramicidin. Loss of VSOP/Hv1 also aborted calcium responses to chemoattractants, increased neutrophil spreading, and decreased neutrophil migration. The migration defect was restored by the addition of a calcium ionophore. Our findings indicate that proton channels extrude the acid and compensate the charge generated by the oxidase, thereby sustaining calcium entry signals that control the adhesion and motility of neutrophils. Loss of proton channels thus aborts superoxide production and causes a severe signaling defect in neutrophils.
Collapse
Affiliation(s)
- Antoun El Chemaly
- Department of Cell Physiology and Metabolism, University of Geneva, 1211 Geneva 4, Switzerland
| | | | | | | | | | | |
Collapse
|
36
|
Lamb FS, Moreland JG, Miller FJ. Electrophysiology of reactive oxygen production in signaling endosomes. Antioxid Redox Signal 2009; 11:1335-47. [PMID: 19207039 PMCID: PMC2872256 DOI: 10.1089/ars.2008.2448] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Endosome trafficking and function require acidification by the vacuolar ATPase (V-ATPase). Electrogenic proton (H+) transport reduces the pH and creates a net positive charge in the endosomal lumen. Concomitant chloride (Cl-) influx has been proposed to occur via ClC Cl-=H+ exchangers. This maintains charge balance and drives Cl- accumulation, which may itself be critical to endosome function. Production of reactive oxygen species (ROS) in response to cytokines occurs within specialized endosomes that form in response to receptor occupation. ROS production requires an NADPH oxidase (Nox) and the ClC-3 Cl-=H+ exchanger. Like the V-ATPase, Nox activity is highly electrogenic, but separates charge with an opposite polarity (lumen negative). Here we review established paradigms of early endosomal ion transport focusing on the relation between the V-ATPase and ClC proteins. Electrophysiologic constraints on Nox-mediated vesicular ROS production are then considered. The potential for ClC-3 to participate in charge neutralization of both proton (V-ATPase) and electron (Nox) transport is discussed. It is proposed that uncompensated charge separation generated by Nox enzymatic activity could be used to drive secondary transport into negatively charged vesicles. Further experimentation will be necessary to establish firmly the biochemistry and functional implications of endosomal ROS production.
Collapse
Affiliation(s)
- Fred S Lamb
- Department of Pediatrics, University of Iowa Hospitals and Clinics, University of Iowa Children's Hospital, and Department of Veterans Affairs Medical Center, Iowa City, Iowa 52242, USA.
| | | | | |
Collapse
|
37
|
Salmon MD, Ahluwalia J. Swell activated chloride channel function in human neutrophils. Biochem Biophys Res Commun 2009; 381:462-5. [DOI: 10.1016/j.bbrc.2009.02.147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 02/26/2009] [Indexed: 11/24/2022]
|
38
|
Bréchard S, Salsmann A, Tschirhart EJ. OAG induces an additional PKC-, PI3K-, and Rac2-mediated signaling pathway up-regulating NOX2 activity, independently of Ca2+ entry. J Leukoc Biol 2009; 85:638-47. [PMID: 19118104 DOI: 10.1189/jlb.0508330] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The requirement of calcium ion (Ca(2)(+)) entry for neutrophil NADPH oxidase (NOX2) regulation is clearly established. However, its role in the signaling pathway leading to NOX2 activation is still elusive. 1-oleoyl-2-acetyl-sn-glycerol (OAG) causes an increase in NOX2 activity and has been shown to directly modulate Ca(2)(+) channels unrelated to the well-known store-operated Ca(2)(+) entry (SOCE) mechanism. In our study, we have investigated the potential role of OAG in Ca(2)(+) influx-mediated NOX2 activity in neutrophil-like-differentiated HL-60 cells to further characterize second signals involved in the regulation of NOX2. OAG inhibited fMLF- and thapsigargin-induced Ca(2)(+) entry, a phenomenon that was not restored by protein kinase C (PKC) or PI3K blockade. Addition of OAG resulted in a rapid decrease of maximal intracellular Ca(2)(+) concentration induced by thapsigargin. Both results suggest that OAG has an inhibitory effect, independent of PI3K and PKC, on the regulation of SOCE. In contrast to SOCE inhibition, OAG-induced NOX2 activation was mediated by PKC and PI3K. Our data establish that both kinases exert their effects through the regulation of Rac2 activity. In addition, OAG potentiated the effect of fMLF on the activation of NOX2 and led to a discernible activity of NOX2 upon thapsigargin stimulation. In conclusion, our results demonstrate that an additional PKC- and/or PI3K-dependent signal may act in synergy with Ca(2)(+) influx to trigger NOX2 activation.
Collapse
Affiliation(s)
- Sabrina Bréchard
- Life Sciences Research Unit, University of Luxembourg, Avenue de la Faïencerie, L-1511 Luxembourg
| | | | | |
Collapse
|
39
|
Musset B, Cherny VV, Morgan D, DeCoursey TE. The intimate and mysterious relationship between proton channels and NADPH oxidase. FEBS Lett 2009; 583:7-12. [PMID: 19084015 PMCID: PMC2630394 DOI: 10.1016/j.febslet.2008.12.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 12/03/2008] [Accepted: 12/03/2008] [Indexed: 10/21/2022]
Abstract
Voltage gated proton channels and NADPH oxidase function cooperatively in phagocytes during the respiratory burst, when reactive oxygen species are produced to kill microbial invaders. Although these molecules are distinct entities, with no proven physical interaction, their presence and activity in many cells appears to be coordinated. We describe these interactions and discuss several types of mechanisms that might explain them.
Collapse
Affiliation(s)
- Boris Musset
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
40
|
Abstract
This review is an attempt to identify and place in context some of the many questions about voltage-gated proton channels that remain unsolved. As the gene was identified only 2 years ago, the situation is very different than in fields where the gene has been known for decades. For the proton channel, most of the obvious and less obvious structure-function questions are still wide open. Remarkably, the proton channel protein strongly resembles the voltage-sensing domain of many voltage-gated ion channels, and thus offers a novel approach to study gating mechanisms. Another surprise is that the proton channel appears to function as a dimer, with two separate conduction pathways. A number of significant biological questions remain in dispute, unanswered, or in some cases, not yet asked. This latter deficit is ascribable to the intrinsic difficulty in evaluating the importance of one component in a complex system, and in addition, to the lack, until recently, of a means of performing an unambiguous lesion experiment, that is, of selectively eliminating the molecule in question. We still lack a potent, selective pharmacological inhibitor, but the identification of the gene has allowed the development of powerful new tools including proton channel antibodies, siRNA and knockout mice.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, 1750 W. Harrison, Chicago, IL 60612, USA.
| |
Collapse
|
41
|
Abstract
The history of research on voltage-gated proton channels is recounted, from their proposed existence in dinoflagellates by Hastings in 1972 and their demonstration in snail neurons by Thomas and Meech in 1982 to the discovery in 2006 (after a decade of controversy) of genes that unequivocally code for proton channels. Voltage-gated proton channels are perfectly selective for protons, conduct deuterons half as well, and the conductance is strongly temperature dependent. These properties are consistent with a conduction mechanism involving hydrogen-bonded-chain transfer, in which the selectivity filter is a titratable amino acid residue. Channel opening is regulated stringently by pH such that only outward current is normally activated. Main functions of proton channels include acid extrusion from cells and charge compensation for the electrogenic activity of the phagocyte NADPH oxidase. Genetic approaches hold the promise of rapid progress in the near future.
Collapse
Affiliation(s)
- T E DeCoursey
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, 1750 W. Harrison, Chicago, Illinois 60612, USA.
| |
Collapse
|
42
|
Role of Nox2 in elimination of microorganisms. Semin Immunopathol 2008; 30:237-53. [PMID: 18574584 DOI: 10.1007/s00281-008-0126-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Accepted: 05/23/2008] [Indexed: 12/16/2022]
Abstract
NADPH oxidase of the phagocytic cells (Nox2) transfers electrons from cytosolic NADPH to molecular oxygen in the extracellular or intraphagosomal space. The produced superoxide anion (O*2) provides the source for formation of all toxic oxygen derivatives, but continuous O*2 generation depends on adequate charge compensation. The vital role of Nox2 in efficient elimination of microorganisms is clearly indicated by human pathology as insufficient activity of the enzyme results in severe, recurrent bacterial infections, the typical symptoms of chronic granulomatous disease. The goals of this contribution are to provide critical review of the Nox2-dependent cellular processes that potentially contribute to bacterial killing and degradation and to indicate possible targets of pharmacological interventions.
Collapse
|
43
|
Morgan D, DeCoursey TE. Analysis of electrophysiological properties and responses of neutrophils. Methods Mol Biol 2008; 412:139-75. [PMID: 18453111 DOI: 10.1007/978-1-59745-467-4_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The past decade has seen increasing use of the patch clamp technique on neutrophils and eosinophils. The main goal of these electrophysiological studies has been to elucidate the mechanisms underlying the phagocyte respiratory burst. NADPH oxidase activity, which defines the respiratory burst in granulocytes, is electrogenic because electrons from NADPH are transported across the cell membrane, where they reduce oxygen to form superoxide anion (O2-). This passage of electrons comprises an electrical current that would rapidly depolarize the membrane if the charge movement were not balanced by proton efflux. The patch clamp technique enables simultaneous recording of NADPH oxidase-generated electron current and H+ flux through the closely related H+ channel. Increasing evidence suggests that other ion channels may play crucial roles in degranulation, phagocytosis, and chemotaxis, highlighting the importance of electrophysiological studies to advance knowledge of granulocyte function. Several configurations of the patch clamp technique exist. Each has advantages and limitations that are discussed here. Meaningful measurements of ion channels cannot be achieved without an understanding of their fundamental properties. We describe the types of measurements that are necessary to characterize a particular ion channel.
Collapse
Affiliation(s)
- Deri Morgan
- Department of Molecular Biophysics and Physiology, Rush University and Medical Center, Chicago, IL, USA
| | | |
Collapse
|
44
|
Bréchard S, Tschirhart EJ. Regulation of superoxide production in neutrophils: role of calcium influx. J Leukoc Biol 2008; 84:1223-37. [PMID: 18519744 PMCID: PMC2567897 DOI: 10.1189/jlb.0807553] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Upon stimulation, activation of NADPH oxidase complexes in neutrophils produces a burst of superoxide anions contributing to oxidative stress and the development of inflammatory process. Store-operated calcium entry (SOCE), whereby the depletion of intracellular stores induces extracellular calcium influx, is known to be a crucial element of NADPH oxidase regulation. However, the mechanistic basis mediating SOCE is still only partially understood, as is the signal-coupling pathway leading to modulation of store-operated channels. This review emphasizes the role of calcium influx in the control of the NADPH oxidase and summarizes the current knowledge of pathways mediating this extracellular calcium entry in neutrophils. Such investigations into the cross-talk between NADPH oxidase and calcium might allow the identification of novel pharmacological targets with clinical use, particularly in inflammatory diseases.
Collapse
Affiliation(s)
- Sabrina Bréchard
- Life Sciences Research Unit, University of Luxembourg, Luxembourg.
| | | |
Collapse
|
45
|
Abstract
Neutrophils constitute the dominant cell in the circulation that mediates the earliest innate immune human responses to infection. The morbidity and mortality from infection rise dramatically in patients with quantitative or qualitative neutrophil defects, providing clinical confirmation of the important role of normal neutrophils for human health. Neutrophil-dependent anti-microbial activity against ingested microbes represents the collaboration of multiple agents, including those prefabricated during granulocyte development in the bone marrow and those generated de novo following neutrophil activation. Furthermore, neutrophils cooperate with extracellular agents as well as other immune cells to optimally kill and degrade invading microbes. This brief review focuses attention on two examples of the integrated nature of neutrophil-mediated anti-microbial action within the phagosome. The importance and complexity of myeloperoxidase-mediated events illustrate a collaboration of anti-microbial responses that are endogenous to the neutrophil, whereas the synergy between the phagocyte NADPH (nicotinamide adenine dinucleotide phosphate) oxidase and plasma-derived group IIA phospholipase A(2) exemplifies the collective effects of the neutrophil with an exogenous factor to achieve degradation of ingested staphylococci.
Collapse
Affiliation(s)
- William M Nauseef
- Inflammation Program, Department of Medicine, University of Iowa, Iowa City, IA 52241, USA.
| |
Collapse
|
46
|
Hydrogen peroxide activates calcium influx in human neutrophils. Mol Cell Biochem 2007; 309:151-6. [PMID: 18008137 DOI: 10.1007/s11010-007-9653-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 10/31/2007] [Indexed: 01/23/2023]
Abstract
The correlation between an increased production of reactive oxygen species (ROS) and an enhanced calcium entry in primed neutrophils stimulated with fMLP suggests that endogenous ROS could serve as an agonist to reinforce calcium signaling by positive feedback. This work shows that exogenous H2O2 produced a rapid influx of Mn2+ and an increase of intracellular calcium. The H2O2 was insufficient to produce significant changes in the absence of extracellular calcium but addition of Ca2+ to H2O2-treated cells suspended in a free Ca2+/EGTA buffer resulted in a great increase in [Ca2+]i reflecting influx of Ca2+ across the cell membrane. The increase of intracellular calcium was inhibited by Ni2+, La3+, and hyperosmotic solutions of mannitol and other osmolytes. This raises the possibility that the secretion of H2O2 by activated neutrophils could act as an autocrine regulator of neutrophil function through the activation of calcium entry.
Collapse
|
47
|
Moreland JG, Davis AP, Matsuda JJ, Hook JS, Bailey G, Nauseef WM, Lamb FS. Endotoxin priming of neutrophils requires NADPH oxidase-generated oxidants and is regulated by the anion transporter ClC-3. J Biol Chem 2007; 282:33958-67. [PMID: 17908687 DOI: 10.1074/jbc.m705289200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Several soluble mediators, including endotoxin, prime neutrophils for an enhanced respiratory burst in response to subsequent stimulation. Priming of neutrophils occurs in vitro, and primed neutrophils are found in vivo. We previously localized the anion transporter ClC-3 to polymorphonuclear leukocytes (PMN) secretory vesicles and demonstrated that it is required for normal NADPH oxidase activation in response to both particulate and soluble stimuli. We now explore the contribution of the NADPH oxidase and ClC-3 to endotoxin-mediated priming. Lipooligosaccharide (LOS) from Neisseria meningitidis enhances the respiratory burst in response to formyl-Met-Leu-Phe, an effect that was impaired in PMNs lacking functional ClC-3 and under anaerobic conditions. Mobilization of receptors to the cell surface and phosphorylation of p38 MAPK by LOS were both impaired in PMN with the NADPH oxidase chemically inhibited or genetically absent and in cells lacking functional ClC-3. Furthermore, inhibition of the NADPH oxidase or ClC-3 in otherwise unstimulated cells elicited a phenotype similar to that seen after endotoxin priming, suggesting that basal oxidant production helps to maintain cellular quiescence. In summary, NADPH oxidase activation was required for LOS-mediated priming, but basal oxidants kept unstimulated cells from becoming primed. ClC-3 contributes to both of these processes.
Collapse
Affiliation(s)
- Jessica G Moreland
- Division of Critical Care, Department of Pediatrics, The Inflammation Program, University of Iowa and Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
DeCoursey TE. Electrophysiology of the phagocyte respiratory burst. Focus on "Large-conductance calcium-activated potassium channel activity is absent in human and mouse neutrophils and is not required for innate immunity". Am J Physiol Cell Physiol 2007; 293:C30-2. [PMID: 17625039 DOI: 10.1152/ajpcell.00093.2007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
49
|
Steinberg BE, Grinstein S. Unconventional roles of the NADPH oxidase: signaling, ion homeostasis, and cell death. ACTA ACUST UNITED AC 2007; 2007:pe11. [PMID: 17392241 DOI: 10.1126/stke.3792007pe11] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although the central role of the phagocytic NADPH oxidase in mediating bacterial killing has long been appreciated, this sophisticated enzyme complex serves various other important functions. This Perspective focuses on these underappreciated roles of phagocytic NADPH oxidase, highlighting recent work implicating reactive oxygen species in triggering an unconventional form of cell death.
Collapse
|
50
|
Heiner I, Eisfeld J, Warnstedt M, Radukina N, Jüngling E, Lückhoff A. Endogenous ADP-ribose enables calcium-regulated cation currents through TRPM2 channels in neutrophil granulocytes. Biochem J 2006; 398:225-32. [PMID: 16719842 PMCID: PMC1550310 DOI: 10.1042/bj20060183] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TRPM2 (transient receptor potential melastatin 2) is a Ca2+-permeable cation channel gated by ADPR (ADP-ribose) from the cytosolic side. To test whether endogenous concentrations of intracellular ADPR are sufficient for TRPM2 gating in neutrophil granulocytes, we devised an HPLC method to determine ADPR contents in HClO4 cell extracts. The reversed-phase ion-pair HPLC method with an Mg2+-containing isocratic eluent allows baseline resolution of one ADPR peak. Intracellular ADPR concentrations were approx. 5 muM in granulocytes and not significantly altered by stimulation with the chemoattractant peptide fMLP (N-formylmethionyl-leucylphenylalanine). We furthermore determined intracellular concentrations of cADPR (cyclic ADPR) with a cyclase assay involving enzymatic conversion of cADPR into NAD+ and fluorimetric determination of NAD+. Intracellular cADPR concentrations were approx. 0.2 microM and not altered by fMLP. In patch-clamp experiments, ADPR (0.1-100 microM) was dialysed into granulocytes to analyse its effects on whole-cell currents characteristic for TRPM2, in the presence of a low (<10 nM) or a high (1 microM) intracellular Ca2+ concentration. TRPM2 currents were significantly larger at high than at low [Ca2+] (e.g. -225+/-27.1 versus -7+/-2.0 pA/pF at 5 muM ADPR), but no currents at all were observed in the absence of ADPR (ADPR concentration < or =0.3 microM). cADPR (0.1, 0.3 and 10 microM) was without effect even in the presence of subthreshold ADPR (0.1 microM). We conclude that ADPR enables an effective regulation of TRPM2 by cytosolic Ca2+. Thus ADPR and Ca2+ in concert behave as a messenger system for agonist-induced influx of Ca2+ through TRPM2 in granulocytes.
Collapse
Affiliation(s)
- Inka Heiner
- Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen, Pauwelsstr. 30, D-52057 Aachen, Germany
| | - Jörg Eisfeld
- Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen, Pauwelsstr. 30, D-52057 Aachen, Germany
| | - Maike Warnstedt
- Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen, Pauwelsstr. 30, D-52057 Aachen, Germany
| | - Natalia Radukina
- Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen, Pauwelsstr. 30, D-52057 Aachen, Germany
| | - Eberhard Jüngling
- Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen, Pauwelsstr. 30, D-52057 Aachen, Germany
| | - Andreas Lückhoff
- Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen, Pauwelsstr. 30, D-52057 Aachen, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|