1
|
Bioelectric regulation of intestinal stem cells. Trends Cell Biol 2022:S0962-8924(22)00234-3. [PMID: 36396487 PMCID: PMC10183058 DOI: 10.1016/j.tcb.2022.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022]
Abstract
Proper regulation of ion balance across the intestinal epithelium is essential for physiological functions, while ion imbalance causes intestinal disorders with dire health consequences. Ion channels, pumps, and exchangers are vital for regulating ion movements (i.e., bioelectric currents) that control epithelial absorption and secretion. Recent in vivo studies used the Drosophila gut to identify conserved pathways that link regulators of Ca2+, Na+ and Cl- with intestinal stem cell (ISC) proliferation. These studies laid a foundation for using the Drosophila gut to identify conserved proliferative responses triggered by bioelectric regulators. Here, we review these studies, discuss their significance, as well as the advantages of using Drosophila to unravel conserved bioelectrically induced molecular pathways in the intestinal epithelium under physiological, pathophysiological, and regenerative conditions.
Collapse
|
2
|
Selvakumar P, Fernández-Mariño AI, Khanra N, He C, Paquette AJ, Wang B, Huang R, Smider VV, Rice WJ, Swartz KJ, Meyerson JR. Structures of the T cell potassium channel Kv1.3 with immunoglobulin modulators. Nat Commun 2022; 13:3854. [PMID: 35788586 PMCID: PMC9253088 DOI: 10.1038/s41467-022-31285-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
The Kv1.3 potassium channel is expressed abundantly on activated T cells and mediates the cellular immune response. This role has made the channel a target for therapeutic immunomodulation to block its activity and suppress T cell activation. Here, we report structures of human Kv1.3 alone, with a nanobody inhibitor, and with an antibody-toxin fusion blocker. Rather than block the channel directly, four copies of the nanobody bind the tetramer's voltage sensing domains and the pore domain to induce an inactive pore conformation. In contrast, the antibody-toxin fusion docks its toxin domain at the extracellular mouth of the channel to insert a critical lysine into the pore. The lysine stabilizes an active conformation of the pore yet blocks ion permeation. This study visualizes Kv1.3 pore dynamics, defines two distinct mechanisms to suppress Kv1.3 channel activity with exogenous inhibitors, and provides a framework to aid development of emerging T cell immunotherapies.
Collapse
Affiliation(s)
- Purushotham Selvakumar
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Ana I Fernández-Mariño
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Nandish Khanra
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Changhao He
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Alice J Paquette
- Cryo-Electron Microscopy Core, New York University School of Medicine, New York, NY, USA
| | - Bing Wang
- Cryo-Electron Microscopy Core, New York University School of Medicine, New York, NY, USA
| | - Ruiqi Huang
- Applied Biomedical Science Institute, San Diego, CA, USA
- Minotaur Therapeutics, San Diego, CA, USA
| | - Vaughn V Smider
- Applied Biomedical Science Institute, San Diego, CA, USA
- Minotaur Therapeutics, San Diego, CA, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - William J Rice
- Cryo-Electron Microscopy Core, New York University School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Kenton J Swartz
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Joel R Meyerson
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
3
|
Zeng Q, Lu W, Deng Z, Zhang B, Wu J, Chai J, Chen X, Xu X. The toxin mimic FS48 from the salivary gland of Xenopsylla cheopis functions as a Kv1.3 channel-blocking immunomodulator of T cell activation. J Biol Chem 2022; 298:101497. [PMID: 34919963 PMCID: PMC8732088 DOI: 10.1016/j.jbc.2021.101497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/14/2021] [Accepted: 12/10/2021] [Indexed: 11/29/2022] Open
Abstract
The Kv1.3 channel has been widely demonstrated to play crucial roles in the activation and proliferation of T cells, which suggests that selective blockers could serve as potential therapeutics for autoimmune diseases mediated by T cells. We previously described that the toxin mimic FS48 from salivary gland of Xenopsylla cheopis downregulates the secretion of proinflammatory factors by Raw 264.7 cells by blocking the Kv1.3 channel and the subsequent inactivation of the proinflammatory MAPK/NF-κB pathways. However, the effects of FS48 on human T cells and autoimmune diseases are unclear. Here, we described its immunomodulatory effects on human T cells derived from suppression of Kv1.3 channel. Kv1.3 currents in Jurkat T cells were recorded by whole-cell patch-clamp, and Ca2+ influx, cell proliferation, and TNF-α and IL-2 secretion were measured using Fluo-4, CCK-8, and ELISA assays, respectively. The in vivo immunosuppressive activity of FS48 was evaluated with a rat DTH model. We found that FS48 reduced Kv1.3 currents in Jurkat T cells in a concentration-dependent manner with an IC50 value of about 1.42 μM. FS48 also significantly suppressed Kv1.3 protein expression, Ca2+ influx, MAPK/NF-κB/NFATc1 pathway activation, and TNF-α and IL-2 production in activated Jurkat T cells. Finally, we show that FS48 relieved the DTH response in rats. We therefore conclude that FS48 can block the Kv1.3 channel and inhibit human T cell activation, which most likely contributes to its immunomodulatory actions and highlights the great potential of this evolutionary-guided peptide as a drug template in future studies.
Collapse
Affiliation(s)
- Qingye Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wancheng Lu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Zhenhui Deng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Bei Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Voros O, Panyi G, Hajdu P. Immune Synapse Residency of Orai1 Alters Ca 2+ Response of T Cells. Int J Mol Sci 2021; 22:ijms222111514. [PMID: 34768945 PMCID: PMC8583858 DOI: 10.3390/ijms222111514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
CRAC, which plays important role in Ca2+-dependent T-lymphocyte activation, is composed of the ER-resident STIM1 and the plasma membrane Orai1 pore-forming subunit. Both accumulate at the immunological synapse (IS) between a T cell and an antigen-presenting cell (APC). We hypothesized that adapter/interacting proteins regulate Orai1 residence in the IS. We could show that mGFP-tagged Orai1-Full channels expressed in Jurkat cells had a biphasic IS-accumulation kinetics peaked at 15 min. To understand the background of Orai1 IS-redistribution we knocked down STIM1 and SAP97 (adaptor protein with a short IS-residency (15 min) and ability to bind Orai1 N-terminus): the mGFP-Orai1-Full channels kept on accumulating in the IS up to the 60th minute in the STIM1- and SAP97-lacking Jurkat cells. Deletion of Orai1 N terminus (mGFP-Orai1-Δ72) resulted in the same time course as described for STIM1/SAP97 knock-down cells. Ca2+-imaging of IS-engaged T-cells revealed that of Orai1 residency modifies the Ca2+-response: cells expressing mGFP-Orai1-Δ72 construct or mGFP-Orai1-Full in SAP-97 knock-down cells showed higher number of Ca2+-oscillation up to the 90th minute after IS formation. Overall, these data suggest that SAP97 may contribute to the short-lived IS-residency of Orai1 and binding of STIM1 to Orai1 N-terminus is necessary for SAP97-Orai1 interaction.
Collapse
Affiliation(s)
- Orsolya Voros
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (O.V.); (G.P.)
| | - György Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (O.V.); (G.P.)
| | - Péter Hajdu
- Department of Biophysics and Cell Biology, Faculty of Dentistry, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
- Correspondence: ; Tel.: +36-52-258603
| |
Collapse
|
5
|
Unterweger AL, Jensen MØ, Giordanetto F, Jogini V, Rüschher A, Seuß M, Winkelmann P, Koletzko L, Shaw DE, Siebeck M, Gropp R, Beigel F, Aszodi A. Suppressing Kv1.3 Ion Channel Activity with a Novel Small Molecule Inhibitor Ameliorates Inflammation in a Humanised Mouse Model of Ulcerative Colitis. J Crohns Colitis 2021; 15:1943-1958. [PMID: 33891001 PMCID: PMC8575044 DOI: 10.1093/ecco-jcc/jjab078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND AIMS The potassium channel Kv1.3 is a potentially attractive therapeutic target in T cell-mediated inflammatory diseases, as the activity of antigen-activated T cells is selectively impeded by Kv1.3 inhibition. In this study, we examined Kv1.3 as a potential therapeutic intervention point for ulcerative colitis [UC], and studied the efficacy of DES1, a small-molecule inhibitor of Kv1.3, in vitro and in vivo. METHODS Kv1.3 expression on T cells in peripheral blood mononuclear cells [PBMCs] isolated from donors with and without UC was examined by flow cytometry. In biopsies from UC patients, Kv1.3-expressing CD4+ T cells were detected by flow cytometry and immunohistochemistry. In vitro, we determined the ability of DES1 to inhibit anti-CD3-driven activation of T cells. In vivo, the efficacy of DES1 was determined in a humanised mouse model of UC and compared with infliximab and tofacitinib in head-to-head studies. RESULTS Kv1.3 expression was elevated in PBMCs from UC patients and correlated with the prevalence of TH1 and TH2 T cells. Kv1.3 expression was also detected on T cells from biopsies of UC patients. In vitro, DES1 suppressed anti-CD3-driven activation of T cells in a concentration-dependent manner. In vivo, DES1 significantly ameliorated inflammation in the UC model and most effectively so when PBMCs from donors with higher levels of activated T cells were selected for reconstitution. The efficacy of DES1 was comparable to that of either infliximab or tofacitinib. CONCLUSION Inhibition of Kv1.3 [by DES1, for instance] appears to be a potential therapeutic intervention strategy for UC patients.
Collapse
Affiliation(s)
- Anna-Lena Unterweger
- Department of General, Visceral und Transplantation Surgery, University Hospital, LMU, Munich, Germany
| | | | | | | | - Alena Rüschher
- Department of General, Visceral und Transplantation Surgery, University Hospital, LMU, Munich, Germany
| | - Marietta Seuß
- Department of General, Visceral und Transplantation Surgery, University Hospital, LMU, Munich, Germany
| | - Paula Winkelmann
- Department of General, Visceral und Transplantation Surgery, University Hospital, LMU, Munich, Germany
| | - Leandra Koletzko
- Department of Medicine II, University Hospital, LMUMunich, Germany
| | - David E Shaw
- D. E. Shaw Research, New York, NY, USA,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Matthias Siebeck
- Department of General, Visceral und Transplantation Surgery, University Hospital, LMU, Munich, Germany
| | - Roswitha Gropp
- Department of General, Visceral und Transplantation Surgery, University Hospital, LMU, Munich, Germany,Corresponding author: Roswitha Gropp, Department of General, Visceral and Transplantation Surgery, Hospital of the Ludwig-Maximilian University Munich, Nussbaumstr. 20, 80336 Munich, Germany.
| | - Florian Beigel
- Department of Medicine II, University Hospital, LMUMunich, Germany
| | - Attila Aszodi
- Department of General, Trauma and Reconstructive Surgery, University Hospital, LMU Munich, Germany’
| |
Collapse
|
6
|
Abundant Monovalent Ions as Environmental Signposts for Pathogens during Host Colonization. Infect Immun 2021; 89:IAI.00641-20. [PMID: 33526568 DOI: 10.1128/iai.00641-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Host colonization by a pathogen requires proper sensing and response to local environmental cues, to ensure adaptation and continued survival within the host. The ionic milieu represents a critical potential source of environmental cues, and indeed, there has been extensive study of the interplay between host and pathogen in the context of metals such as iron, zinc, and manganese, vital ions that are actively sequestered by the host. The inherent non-uniformity of the ionic milieu also extends, however, to "abundant" ions such as chloride and potassium, whose concentrations vary greatly between tissue and cellular locations, and with the immune response. Despite this, the concept of abundant ions as environmental cues and key players in host-pathogen interactions is only just emerging. Focusing on chloride and potassium, this review brings together studies across multiple bacterial and parasitic species that have begun to define both how these abundant ions are exploited as cues during host infection, and how they can be actively manipulated by pathogens during host colonization. The close links between ion homeostasis and sensing/response to different ionic signals, and the importance of studying pathogen response to cues in combination, are also discussed, while considering the fundamental insight still to be uncovered from further studies in this nascent area of inquiry.
Collapse
|
7
|
Abstract
Kv7 channels (Kv7.1-7.5) are voltage-gated K+ channels that can be modulated by five β-subunits (KCNE1-5). Kv7.1-KCNE1 channels produce the slow-delayed rectifying K+ current, IKs, which is important during the repolarization phase of the cardiac action potential. Kv7.2-7.5 are predominantly neuronally expressed and constitute the muscarinic M-current and control the resting membrane potential in neurons. Kv7.1 produces drastically different currents as a result of modulation by KCNE subunits. This flexibility allows the Kv7.1 channel to have many roles depending on location and assembly partners. The pharmacological sensitivity of Kv7.1 channels differs from that of Kv7.2-7.5 and is largely dependent upon the number of β-subunits present in the channel complex. As a result, the development of pharmaceuticals targeting Kv7.1 is problematic. This review discusses the roles and the mechanisms by which different signaling pathways affect Kv7.1 and KCNE channels and could potentially provide different ways of targeting the channel.
Collapse
Affiliation(s)
- Emely Thompson
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| | - Jodene Eldstrom
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| | - David Fedida
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| |
Collapse
|
8
|
Barrera P, Skorka C, Boktor M, Dave N, Jimenez V. A Novel Calcium-Activated Potassium Channel Controls Membrane Potential and Intracellular pH in Trypanosoma cruzi. Front Cell Infect Microbiol 2020; 9:464. [PMID: 32010643 PMCID: PMC6974456 DOI: 10.3389/fcimb.2019.00464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Trypanosoma cruzi develops in environments where nutrient availability, osmolarity, ionic concentrations, and pH undergo significant changes. The ability to adapt and respond to such conditions determines the survival and successful transmission of T. cruzi. Ion channels play fundamental roles in controlling physiological parameters that ensure cell homeostasis by rapidly triggering compensatory mechanisms. Combining molecular, cellular and electrophysiological approaches we have identified and characterized the expression and function of a novel calcium-activated potassium channel (TcCAKC). This channel resides in the plasma membrane of all 3 life stages of T. cruzi and shares structural features with other potassium channels. We expressed TcCAKC in Xenopus laevis oocytes and established its biophysical properties by two-electrode voltage clamp. Oocytes expressing TcCAKC showed a significant increase in inward currents after addition of calcium ionophore ionomycin or thapsigargin. These responses were abolished by EGTA suggesting that TcCAKC activation is dependent of extracellular calcium. This activation causes an increase in current and a negative shift in reversal potential that is blocked by barium. As predicted, a single point mutation in the selectivity filter (Y313A) completely abolished the activity of the channels, confirming its potassium selective nature. We have generated knockout parasites deleting one or both alleles of TcCAKC. These parasite strains showed impaired growth, decreased production of trypomastigotes and slower intracellular replication, pointing to an important role of TcCAKC in regulating infectivity. To understand the cellular mechanisms underlying these phenotypic defects, we used fluorescent probes to evaluate intracellular membrane potential, pH, and intracellular calcium. Epimastigotes lacking the channel had significantly lower cytosolic calcium, hyperpolarization, changes in intracellular pH, and increased rate of proton extrusion. These results are in agreement with previous reports indicating that, in trypanosomatids, membrane potential and intracellular pH maintenance are linked. Our work shows TcCAKC is a novel potassium channel that contributes to homeostatic regulation of important physiological processes in T. cruzi and provides new avenues to explore the potential of ion channels as targets for drug development against protozoan parasites.
Collapse
Affiliation(s)
- Patricia Barrera
- Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA, United States
| | - Christopher Skorka
- Departmento de Biología, Facultad de Ciencias Exactas y Naturales, Instituto de Histologia y Embriologia IHEM-CONICET, Facultad de Medicina, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Michael Boktor
- Departmento de Biología, Facultad de Ciencias Exactas y Naturales, Instituto de Histologia y Embriologia IHEM-CONICET, Facultad de Medicina, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Noopur Dave
- Departmento de Biología, Facultad de Ciencias Exactas y Naturales, Instituto de Histologia y Embriologia IHEM-CONICET, Facultad de Medicina, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Veronica Jimenez
- Departmento de Biología, Facultad de Ciencias Exactas y Naturales, Instituto de Histologia y Embriologia IHEM-CONICET, Facultad de Medicina, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
9
|
Hodo TW, de Aquino MTP, Shimamoto A, Shanker A. Critical Neurotransmitters in the Neuroimmune Network. Front Immunol 2020; 11:1869. [PMID: 32973771 PMCID: PMC7472989 DOI: 10.3389/fimmu.2020.01869] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Immune cells rely on cell-cell communication to specify and fine-tune their responses. They express an extensive network of cell communication modes, including a vast repertoire of cell surface and transmembrane receptors and ligands, membrane vesicles, junctions, ligand and voltage-gated ion channels, and transporters. During a crosstalk between the nervous system and the immune system these modes of cellular communication and the downstream signal transduction events are influenced by neurotransmitters present in the local tissue environments in an autocrine or paracrine fashion. Neurotransmitters thus influence innate and adaptive immune responses. In addition, immune cells send signals to the brain through cytokines, and are present in the brain to influence neural responses. Altered communication between the nervous and immune systems is emerging as a common feature in neurodegenerative and immunopathological diseases. Here, we present the mechanistic frameworks of immunostimulatory and immunosuppressive effects critical neurotransmitters - dopamine (3,4-dihydroxyphenethylamine), serotonin (5-hydroxytryptamine), substance P (trifluoroacetate salt powder), and L-glutamate - exert on lymphocytes and non-lymphoid immune cells. Furthermore, we discuss the possible roles neurotransmitter-driven neuroimmune networks play in the pathogenesis of neurodegenerative disorders, autoimmune diseases, cancer, and outline potential clinical implications of balancing neuroimmune crosstalk by therapeutic modulation.
Collapse
Affiliation(s)
- Thomas Wesley Hodo
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College School of Medicine, Nashville, TN, United States.,Department of Microbiology and Immunology, Meharry Medical College School of Medicine, Nashville, TN, United States.,School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Maria Teresa Prudente de Aquino
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College School of Medicine, Nashville, TN, United States
| | - Akiko Shimamoto
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College School of Medicine, Nashville, TN, United States
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College School of Medicine, Nashville, TN, United States.,School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States.,Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
10
|
Peruzzo R, Mattarei A, Romio M, Paradisi C, Zoratti M, Szabò I, Leanza L. Regulation of Proliferation by a Mitochondrial Potassium Channel in Pancreatic Ductal Adenocarcinoma Cells. Front Oncol 2017; 7:239. [PMID: 29034212 PMCID: PMC5626813 DOI: 10.3389/fonc.2017.00239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/19/2017] [Indexed: 12/21/2022] Open
Abstract
Previous results link the mitochondrial potassium channel Kv1.3 (mitoKv1.3) to the regulation of apoptosis. By synthesizing new, mitochondria-targeted derivatives (PAPTP and PCARBTP) of PAP-1, a specific membrane-permeant Kv1.3 inhibitor, we have recently provided evidence that both drugs acting on mitoKv1.3 are able to induce apoptosis and reduce tumor growth in vivo without affecting healthy tissues and cells. In the present article, by exploiting these new drugs, we addressed the question whether mitoKv1.3 contributes to the regulation of cell proliferation as well. When used at low concentrations, which do not compromise cell survival, both drugs slightly increased the percentage of cells in S phase while decreased the population at G0/G1 stage of cells from two different pancreatic ductal adenocarcinoma lines. Our data suggest that the observed modulation is related to ROS levels within the cells, opening the way to link mitochondrial ion channel function to downstream, ROS-related signaling events that might be important for cell cycle progression.
Collapse
Affiliation(s)
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Matteo Romio
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Cristina Paradisi
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Mario Zoratti
- Institute of Neuroscience, CNR, Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padova, Padova, Italy.,Institute of Neuroscience, CNR, Padova, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
11
|
Fung-Leung WP, Edwards W, Liu Y, Ngo K, Angsana J, Castro G, Wu N, Liu X, Swanson RV, Wickenden AD. T Cell Subset and Stimulation Strength-Dependent Modulation of T Cell Activation by Kv1.3 Blockers. PLoS One 2017; 12:e0170102. [PMID: 28107393 PMCID: PMC5249144 DOI: 10.1371/journal.pone.0170102] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 12/29/2016] [Indexed: 12/28/2022] Open
Abstract
Kv1.3 is a voltage-gated potassium channel expressed on T cells that plays an important role in T cell activation. Previous studies have shown that blocking Kv1.3 channels in human T cells during activation results in reduced calcium entry, cytokine production, and proliferation. The aim of the present study was to further explore the effects of Kv1.3 blockers on the response of different human T cell subsets under various stimulation conditions. Our studies show that, unlike the immune suppressor cyclosporine A, the inhibitory effect of Kv1.3 blockers was partial and stimulation strength dependent, with reduced inhibitory efficacy on T cells under strengthened anti-CD3/CD28 stimulations. T cell responses to allergens including house dust mites and ragweed were partially reduced by Kv1.3 blockers. The effect of Kv1.3 inhibition was dependent on T cell subsets, with stronger effects on CCR7- effector memory compared to CCR7+ central memory CD4 T cells. Calcium entry studies also revealed a population of CD4 T cells resistant to Kv1.3 blockade. Activation of CD4 T cells was accompanied with an increase in Kv1.3 currents but Kv1.3 transcripts were found to be reduced, suggesting a posttranscriptional mechanism in the regulation of Kv1.3 activities. In summary, Kv1.3 blockers inhibit T cell activation in a manner that is highly dependent on the T cell identity and stimulation strength, These findings suggest that Kv1.3 blockers inhibit T cells in a unique, conditional manner, further refining our understanding of the therapeutic potential of Kv1.3 blockers.
Collapse
Affiliation(s)
- Wai-Ping Fung-Leung
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
- * E-mail:
| | - Wilson Edwards
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| | - Yi Liu
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| | - Karen Ngo
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| | - Julianty Angsana
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| | - Glenda Castro
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| | - Nancy Wu
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| | - Xuejun Liu
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| | - Ronald V. Swanson
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| | - Alan D. Wickenden
- Janssen Research & Development, L.L.C., San Diego, California, United States of America
| |
Collapse
|
12
|
Ahn SJ, Fancher IS, Bian JT, Zhang CX, Schwab S, Gaffin R, Phillips SA, Levitan I. Inwardly rectifying K + channels are major contributors to flow-induced vasodilatation in resistance arteries. J Physiol 2016; 595:2339-2364. [PMID: 27859264 PMCID: PMC5374117 DOI: 10.1113/jp273255] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/07/2016] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Endothelial inwardly rectifying K+ (Kir2.1) channels regulate flow-induced vasodilatation via nitric oxide (NO) in mouse mesenteric resistance arteries. Deficiency of Kir2.1 channels results in elevated blood pressure and increased vascular resistance. Flow-induced vasodilatation in human resistance arteries is also regulated by inwardly rectifying K+ channels. This study presents the first direct evidence that Kir channels play a critical role in physiological endothelial responses to flow. ABSTRACT Inwardly rectifying K+ (Kir) channels are known to be sensitive to flow, but their role in flow-induced endothelial responses is not known. The goal of this study is to establish the role of Kir channels in flow-induced vasodilatation and to provide first insights into the mechanisms responsible for Kir signalling in this process. First, we establish that primary endothelial cells isolated from murine mesenteric arteries express functional Kir2.1 channels sensitive to shear stress. Then, using the Kir2.1+/- heterozygous mouse model, we establish that downregulation of Kir2.1 results in significant decrease in shear-activated Kir currents and inhibition of endothelium-dependent flow-induced vasodilatation (FIV) assayed in pressurized mesenteric arteries pre-constricted with endothelin-1. Deficiency in Kir2.1 also results in the loss of flow-induced phosphorylation of eNOS and Akt, as well as inhibition of NO generation. All the effects are fully rescued by endothelial cell (EC)-specific overexpression of Kir2.1. A component of FIV that is Kir independent is abrogated by blocking Ca2+ -sensitive K+ channels. Kir2.1 has no effect on endothelium-independent and K+ -induced vasodilatation in denuded arteries. Kir2.1+/- mice also show increased mean blood pressure measured by carotid artery cannulation and increased microvascular resistance measured using a tail-cuff. Importantly, blocking Kir channels also inhibits flow-induced vasodilatation in human subcutaneous adipose microvessels. Endothelial Kir channels contribute to FIV of mouse mesenteric arteries via an NO-dependent mechanism, whereas Ca2+ -sensitive K+ channels mediate FIV via an NO-independent pathway. Kir2 channels also regulate vascular resistance and blood pressure. Finally, Kir channels also contribute to FIV in human subcutaneous microvessels.
Collapse
Affiliation(s)
- Sang Joon Ahn
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| | - Ibra S Fancher
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA.,Department of Physical Therapy, University of Illinois at Chicago, Chicago, IL, USA
| | - Jing-Tan Bian
- Department of Physical Therapy, University of Illinois at Chicago, Chicago, IL, USA
| | - Chong Xu Zhang
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| | - Sarah Schwab
- Department of Physical Therapy, University of Illinois at Chicago, Chicago, IL, USA
| | - Robert Gaffin
- Department of Physiology, Physiology Core Lab, University of Illinois at Chicago, Chicago, IL, USA
| | - Shane A Phillips
- Department of Physical Therapy, University of Illinois at Chicago, Chicago, IL, USA
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
13
|
Singh J, Shah R, Singh D. Inundation of asthma target research: Untangling asthma riddles. Pulm Pharmacol Ther 2016; 41:60-85. [PMID: 27667568 DOI: 10.1016/j.pupt.2016.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/11/2016] [Accepted: 09/20/2016] [Indexed: 12/31/2022]
Abstract
Asthma is an inveterate inflammatory disorder, delineated by the airway inflammation, bronchial hyperresponsiveness (BHR) and airway wall remodeling. Although, asthma is a vague term, and is recognized as heterogenous entity encompassing different phenotypes. Targeting single mediator or receptor did not prove much clinical significant, as asthma is complex disease involving myriad inflammatory mediators. Asthma may probably involve a large number of different types of molecular and cellular components interacting through complex pathophysiological pathways. This review covers the past, present, and future therapeutic approaches and pathophysiological mechanisms of asthma. Furthermore, review describe importance of targeting several mediators/modulators and receptor antagonists involved in the physiopathology of asthma. Novel targets for asthma research include Galectins, Immunological targets, K + Channels, Kinases and Transcription Factors, Toll-like receptors, Selectins and Transient receptor potential channels. But recent developments in asthma research are very promising, these include Bitter taste receptors (TAS2R) abated airway obstruction in mouse model of asthma and Calcium-sensing receptor obliterate inflammation and in bronchial hyperresponsiveness allergic asthma. All these progresses in asthma targets, and asthma phenotypes exploration are auspicious in untangling of asthma riddles.
Collapse
Affiliation(s)
- Jatinder Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, 147002, Punjab, India
| | - Ramanpreet Shah
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, 147002, Punjab, India
| | - Dhandeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, 147002, Punjab, India.
| |
Collapse
|
14
|
Placing ion channels into a signaling network of T cells: from maturing thymocytes to healthy T lymphocytes or leukemic T lymphoblasts. BIOMED RESEARCH INTERNATIONAL 2015; 2015:750203. [PMID: 25866806 PMCID: PMC4383400 DOI: 10.1155/2015/750203] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022]
Abstract
T leukemogenesis is a multistep process, where the genetic errors during T cell maturation cause the healthy progenitor to convert into the leukemic precursor that lost its ability to differentiate but possesses high potential for proliferation, self-renewal, and migration. A new misdirecting "leukemogenic" signaling network appears, composed by three types of participants which are encoded by (1) genes implicated in determined stages of T cell development but deregulated by translocations or mutations, (2) genes which normally do not participate in T cell development but are upregulated, and (3) nondifferentially expressed genes which become highly interconnected with genes expressed differentially. It appears that each of three groups may contain genes coding ion channels. In T cells, ion channels are implicated in regulation of cell cycle progression, differentiation, activation, migration, and cell death. In the present review we are going to reveal a relationship between different genetic defects, which drive the T cell neoplasias, with calcium signaling and ion channels. We suggest that changes in regulation of various ion channels in different types of the T leukemias may provide the intracellular ion microenvironment favorable to maintain self-renewal capacity, arrest differentiation, induce proliferation, and enhance motility.
Collapse
|
15
|
Inhibition of KCa3.1 by depolarisation and 2-aminoethoxydiphenyl borate (2-APB) during Ca2+ release activated Ca2+ (CRAC) entry in human erythroleukemia (HEL) cells: Implications for the interpretation of 2-APB inhibition of CRAC entry. Cell Calcium 2015; 57:76-88. [DOI: 10.1016/j.ceca.2014.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 11/12/2014] [Accepted: 12/12/2014] [Indexed: 01/05/2023]
|
16
|
Edwards W, Fung-Leung WP, Huang C, Chi E, Wu N, Liu Y, Maher MP, Bonesteel R, Connor J, Fellows R, Garcia E, Lee J, Lu L, Ngo K, Scott B, Zhou H, Swanson RV, Wickenden AD. Targeting the ion channel Kv1.3 with scorpion venom peptides engineered for potency, selectivity, and half-life. J Biol Chem 2014; 289:22704-22714. [PMID: 24939846 DOI: 10.1074/jbc.m114.568642] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Ion channels are an attractive class of drug targets, but progress in developing inhibitors for therapeutic use has been limited largely due to challenges in identifying subtype selective small molecules. Animal venoms provide an alternative source of ion channel modulators, and the venoms of several species, such as scorpions, spiders and snails, are known to be rich sources of ion channel modulating peptides. Importantly, these peptides often bind to hyper-variable extracellular loops, creating the potential for subtype selectivity rarely achieved with small molecules. We have engineered scorpion venom peptides and incorporated them in fusion proteins to generate highly potent and selective Kv1.3 inhibitors with long in vivo half-lives. Kv1.3 has been reported to play a role in human T cell activation, and therefore, these Kv1.3 inhibitor fusion proteins may have potential for the treatment of autoimmune diseases. Our results support an emerging approach to generating subtype selective therapeutic ion channel inhibitors.
Collapse
Affiliation(s)
- Wilson Edwards
- Janssen Research and Development, LLC, San Diego, California 92121.
| | | | - Chichi Huang
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Ellen Chi
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Nancy Wu
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Yi Liu
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Michael P Maher
- Janssen Research and Development, LLC, San Diego, California 92121
| | | | - Judith Connor
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Ross Fellows
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Elena Garcia
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Jerry Lee
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Lu Lu
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Karen Ngo
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Brian Scott
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Hong Zhou
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Ronald V Swanson
- Janssen Research and Development, LLC, San Diego, California 92121
| | - Alan D Wickenden
- Janssen Research and Development, LLC, San Diego, California 92121
| |
Collapse
|
17
|
The neurotransmitter glutamate and human T cells: glutamate receptors and glutamate-induced direct and potent effects on normal human T cells, cancerous human leukemia and lymphoma T cells, and autoimmune human T cells. J Neural Transm (Vienna) 2014; 121:983-1006. [DOI: 10.1007/s00702-014-1167-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/19/2014] [Indexed: 12/26/2022]
|
18
|
Electrophysiological characterization of Ts6 and Ts7, K⁺ channel toxins isolated through an improved Tityus serrulatus venom purification procedure. Toxins (Basel) 2014; 6:892-913. [PMID: 24590385 PMCID: PMC3968367 DOI: 10.3390/toxins6030892] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/24/2014] [Accepted: 02/17/2014] [Indexed: 01/09/2023] Open
Abstract
In Brazil, Tityus serrulatus (Ts) is the species responsible for most of the scorpion related accidents. Among the Ts toxins, the neurotoxins with action on potassium channels (α-KTx) present high interest, due to their effect in the envenoming process and the ion channel specificity they display. The α-KTx toxins family is the most relevant because its toxins can be used as therapeutic tools for specific target cells. The improved isolation method provided toxins with high resolution, obtaining pure Ts6 and Ts7 in two chromatographic steps. The effects of Ts6 and Ts7 toxins were evaluated in 14 different types of potassium channels using the voltage-clamp technique with two-microelectrodes. Ts6 toxin shows high affinity for Kv1.2, Kv1.3 and Shaker IR, blocking these channels in low concentrations. Moreover, Ts6 blocks the Kv1.3 channel in picomolar concentrations with an IC50 of 0.55 nM and therefore could be of valuable assistance to further designing immunosuppressive therapeutics. Ts7 toxin blocks multiple subtypes channels, showing low selectivity among the channels analyzed. This work also stands out in its attempt to elucidate the residues important for interacting with each channel and, in the near future, to model a desired drug.
Collapse
|
19
|
Urrego D, Tomczak AP, Zahed F, Stühmer W, Pardo LA. Potassium channels in cell cycle and cell proliferation. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130094. [PMID: 24493742 PMCID: PMC3917348 DOI: 10.1098/rstb.2013.0094] [Citation(s) in RCA: 288] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Normal cell-cycle progression is a crucial task for every multicellular organism, as it determines body size and shape, tissue renewal and senescence, and is also crucial for reproduction. On the other hand, dysregulation of the cell-cycle progression leading to uncontrolled cell proliferation is the hallmark of cancer. Therefore, it is not surprising that it is a tightly regulated process, with multifaceted and very complex control mechanisms. It is now well established that one of those mechanisms relies on ion channels, and in many cases specifically on potassium channels. Here, we summarize the possible mechanisms underlying the importance of potassium channels in cell-cycle control and briefly review some of the identified channels that illustrate the multiple ways in which this group of proteins can influence cell proliferation and modulate cell-cycle progression.
Collapse
Affiliation(s)
- Diana Urrego
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, , Hermann-Rein-Strasse 3, Göttingen 37075, Germany
| | | | | | | | | |
Collapse
|
20
|
Moreno C, Prieto P, Macías Á, Pimentel-Santillana M, de la Cruz A, Través PG, Boscá L, Valenzuela C. Modulation of voltage-dependent and inward rectifier potassium channels by 15-epi-lipoxin-A4 in activated murine macrophages: implications in innate immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:6136-6146. [PMID: 24249731 DOI: 10.4049/jimmunol.1300235] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Potassium channels modulate macrophage physiology. Blockade of voltage-dependent potassium channels (Kv) by specific antagonists decreases macrophage cytokine production and inhibits proliferation. In the presence of aspirin, acetylated cyclooxygenase-2 loses the activity required to synthesize PGs but maintains the oxygenase activity to produce 15R-HETE from arachidonate. This intermediate product is transformed via 5-LOX into epimeric lipoxins, termed 15-epi-lipoxins (15-epi-lipoxin A4 [e-LXA4]). Kv have been proposed as anti-inflammatory targets. Therefore, we studied the effects of e-LXA4 on signaling and on Kv and inward rectifier potassium channels (Kir) in mice bone marrow-derived macrophages (BMDM). Electrophysiological recordings were performed in these cells by the whole-cell patch-clamp technique. Treatment of BMDM with e-LXA4 inhibited LPS-dependent activation of NF-κB and IκB kinase β activity, protected against LPS activation-dependent apoptosis, and enhanced the accumulation of the Nrf-2 transcription factor. Moreover, treatment of LPS-stimulated BMDM with e-LXA4 resulted in a rapid decrease of Kv currents, compatible with attenuation of the inflammatory response. Long-term treatment of LPS-stimulated BMDM with e-LXA4 significantly reverted LPS effects on Kv and Kir currents. Under these conditions, e-LXA4 decreased the calcium influx versus that observed in LPS-stimulated BMDM. These effects were partially mediated via the lipoxin receptor (ALX), because they were significantly reverted by a selective ALX receptor antagonist. We provide evidence for a new mechanism by which e-LXA4 contributes to inflammation resolution, consisting of the reversion of LPS effects on Kv and Kir currents in macrophages.
Collapse
Affiliation(s)
- Cristina Moreno
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Cientificas, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
A recent patent application from Bionomics/Merck Serono describes novel compounds as blockers of the voltage-gated Kv1.3 ion channel. The blockade of this channel shows great promise as a new therapeutic target for the treatment of autoimmune disorders such as multiple sclerosis, psoriasis, diabetes and rheumatoid arthritis. The generic claim of this patent refers to a new chemotype of Kv1.3 blockers based on an amide core with potent IC50's which are potentially within the nanomolar range. This article briefly reviews the chemistry and biology found in the patent and compares it with previous discoveries in the field.
Collapse
Affiliation(s)
- William Nguyen
- The Scripps Research Institute, Department of Molecular Therapeutics , 130 Scripps Way, Jupiter, FL , USA +1 561 228 2210 ; +1 561 228 3092 ;
| |
Collapse
|
22
|
Sack JT, Stephanopoulos N, Austin DC, Francis MB, Trimmer JS. Antibody-guided photoablation of voltage-gated potassium currents. ACTA ACUST UNITED AC 2013; 142:315-24. [PMID: 23940262 PMCID: PMC3753605 DOI: 10.1085/jgp.201311023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A family of 40 mammalian voltage-gated potassium (Kv) channels control membrane excitability in electrically excitable cells. The contribution of individual Kv channel types to electrophysiological signaling has been difficult to assign, as few selective inhibitors exist for individual Kv subunits. Guided by the exquisite selectivity of immune system interactions, we find potential for antibody conjugates as selective Kv inhibitors. Here, functionally benign anti-Kv channel monoclonal antibodies (mAbs) were chemically modified to facilitate photoablation of K currents. Antibodies were conjugated to porphyrin compounds that upon photostimulation inflict localized oxidative damage. Anti-Kv4.2 mAb–porphyrin conjugates facilitated photoablation of Kv4.2 currents. The degree of K current ablation was dependent on photon dose and conjugate concentration. Kv channel photoablation was selective for Kv4.2 over Kv4.3 or Kv2.1, yielding specificity not present in existing neurotoxins or other Kv channel inhibitors. We conclude that antibody–porphyrin conjugates are capable of selective photoablation of Kv currents. These findings demonstrate that subtype-specific mAbs that in themselves do not modulate ion channel function are capable of delivering functional payloads to specific ion channel targets.
Collapse
Affiliation(s)
- Jon T Sack
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA.
| | | | | | | | | |
Collapse
|
23
|
The role of PSD-95 in the rearrangement of Kv1.3 channels to the immunological synapse. Pflugers Arch 2013; 465:1341-53. [PMID: 23553419 DOI: 10.1007/s00424-013-1256-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 02/08/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
Establishment of the immunological synapse (IS) between T lymphocytes and antigen-presenting cells is a key step in the adaptive immune response. Several proteins accumulate in the IS, such as the Kv1.3 potassium channel; however, the mechanism of this translocation is unknown. PSD-95 and SAP97 are adaptor proteins that regulate the polarized cell surface expression and localization of Kv1 channels in neurons. We investigated whether these proteins affect the redistribution of Kv1.3 into the IS in non-excitable human T cells. We show here that PSD-95 and SAP97 are expressed in Jurkat and interact with the C terminus of Kv1.3. Disruption of the interaction between PSD-95 or SAP97 and Kv1.3 in Jurkat was realized by the expression of a C-terminal truncated Kv1.3, which lacks the binding domain for these proteins, or by the knockdown of the expression of PSD-95 or SAP97 using specific shRNA. Expression of the truncated Kv1.3 or knockdown of PSD-95, but not the knockdown of SAP97, inhibited the recruitment of Kv1.3 into the IS; the fraction of cells showing polarized Kv1.3 expression upon engagement in an IS was significantly lower than in control cells expressing the full-length Kv1.3, and the rearrangement of Kv1.3 did not show time dependence. In contrast, Jurkat cells expressing the full-length channel showed marked time dependence in the recruitment into the IS peaking at 1 min after the conjugation of the cells. These results demonstrate that PSD-95 participates in the targeting of Kv1.3 into the IS, implying its important role in human T-cell activation.
Collapse
|
24
|
Decher N, Netter MF, Streit AK. Putative Impact of RNA Editing on Drug Discovery. Chem Biol Drug Des 2012; 81:13-21. [DOI: 10.1111/cbdd.12045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
25
|
Nguyen W, Howard BL, Jenkins DP, Wulff H, Thompson PE, Manallack DT. Structure-activity relationship exploration of Kv1.3 blockers based on diphenoxylate. Bioorg Med Chem Lett 2012; 22:7106-9. [PMID: 23084278 PMCID: PMC3664202 DOI: 10.1016/j.bmcl.2012.09.080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 09/18/2012] [Accepted: 09/21/2012] [Indexed: 01/27/2023]
Abstract
Diphenoxylate, a well-known opioid agonist and anti-diarrhoeal agent, was recently found to block Kv1.3 potassium channels, which have been proposed as potential therapeutic targets for a range of autoimmune diseases. The molecular basis for this Kv1.3 blockade was assessed by the selective removal of functional groups from the structure of diphenoxylate as well as a number of other structural variations. Removal of the nitrile functional group and replacement of the C-4 piperidinyl substituents resulted in several compounds with submicromolar IC(50) values.
Collapse
Affiliation(s)
- William Nguyen
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Vic. 3052, Australia
| | - Brittany L. Howard
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Vic. 3052, Australia
| | - David P. Jenkins
- Department of Pharmacology, University of California, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Heike Wulff
- Department of Pharmacology, University of California, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | - Philip E. Thompson
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Vic. 3052, Australia
| | - David T. Manallack
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Vic. 3052, Australia
| |
Collapse
|
26
|
Varga Z, Gurrola-Briones G, Papp F, Rodríguez de la Vega RC, Pedraza-Alva G, Tajhya RB, Gaspar R, Cardenas L, Rosenstein Y, Beeton C, Possani LD, Panyi G. Vm24, a natural immunosuppressive peptide, potently and selectively blocks Kv1.3 potassium channels of human T cells. Mol Pharmacol 2012; 82:372-82. [PMID: 22622363 PMCID: PMC3422703 DOI: 10.1124/mol.112.078006] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 05/23/2012] [Indexed: 02/04/2023] Open
Abstract
Blockade of Kv1.3 K(+) channels in T cells is a promising therapeutic approach for the treatment of autoimmune diseases such as multiple sclerosis and type 1 diabetes mellitus. Vm24 (α-KTx 23.1) is a novel 36-residue Kv1.3-specific peptide isolated from the venom of the scorpion Vaejovis mexicanus smithi. Vm24 inhibits Kv1.3 channels of human lymphocytes with high affinity (K(d) = 2.9 pM) and exhibits >1500-fold selectivity over other ion channels assayed. It inhibits the proliferation and Ca(2+) signaling of human T cells in vitro and reduces delayed-type hypersensitivity reactions in rats in vivo. Our results indicate that Vm24 has exceptional pharmacological properties that make it an excellent candidate for treatment of certain autoimmune diseases.
Collapse
Affiliation(s)
- Zoltan Varga
- Department of Biophysics and Cell Biology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhu J, Yan J, Thornhill WB. N-glycosylation promotes the cell surface expression of Kv1.3 potassium channels. FEBS J 2012; 279:2632-44. [PMID: 22613618 DOI: 10.1111/j.1742-4658.2012.08642.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The voltage-gated potassium channel Kv1.3 plays an essential role in modulating membrane excitability in many cell types. Kv1.3 is a heavily glycosylated membrane protein. Two successive N-glycosylation consensus sites, N228NS and N229ST, are present on the S1-S2 linker of rat Kv1.3. Our data suggest that Kv1.3 contains only one N-glycan and it is predominantly attached to N229 in the S1-S2 extracellular linker. Preventing N-glycosylation of Kv1.3 significantly decreased its surface protein level and surface conductance density level, which were ∼ 49% and ∼ 46% respectively of the level of wild type. Supplementation of N-acetylglucosamine (GlcNAc), l-fucose or N-acetylneuraminic acid to the culture medium promoted Kv1.3 surface protein expression, whereas supplementation of d-glucose, d-mannose or d-galactose did not. Among the three effective monosaccharides/derivatives, adding GlcNAc appeared to reduce sialic acid content and increase the degree of branching in the N-glycan of Kv1.3, suggesting that the N-glycan structure and composition had changed. Furthermore, the cell surface half-life of the Kv1.3 surface protein was increased upon GlcNAc supplementation, indicating that it had decreased internalization. The GlcNAc effect appears to apply mainly to membrane proteins containing complex type N-glycans. Thus, N-glycosylation promotes Kv1.3 cell surface expression; supplementation of GlcNAc increased Kv1.3 surface protein level and decreased its internalization, presumably by a combined effect of decreased branch size and increased branching of the N-glycan.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Biological Sciences and Center for Cancer, Genetic Diseases and Gene Regulation, Fordham University, Bronx, NY, USA
| | | | | |
Collapse
|
28
|
Gocke AR, Lebson LA, Grishkan IV, Hu L, Nguyen HM, Whartenby KA, Chandy KG, Calabresi PA. Kv1.3 deletion biases T cells toward an immunoregulatory phenotype and renders mice resistant to autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2012; 188:5877-86. [PMID: 22581856 DOI: 10.4049/jimmunol.1103095] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increasing evidence suggests ion channels have critical functions in the differentiation and plasticity of T cells. Kv1.3, a voltage-gated K(+) channel, is a functional marker and a pharmacological target for activated effector memory T cells. Selective Kv1.3 blockers have been shown to inhibit proliferation and cytokine production by human and rat effector memory T cells. We used Kv1.3 knockout (KO) mice to investigate the mechanism by which Kv1.3 blockade affects CD4(+) T cell differentiation during an inflammatory immune-mediated disease. Kv1.3 KO animals displayed significantly lower incidence and severity of myelin oligodendrocyte glycoprotein (MOG) peptide-induced experimental autoimmune encephalomyelitis. Kv1.3 was the only K(V) channel expressed in MOG 35-55-specific CD4(+) T cell blasts, and no K(V) current was present in MOG-specific CD4(+) T cell-blasts from Kv1.3 KO mice. Fewer CD4(+) T cells migrated to the CNS in Kv1.3 KO mice following disease induction, and Ag-specific proliferation of CD4(+) T cells from these mice was impaired with a corresponding cell-cycle delay. Kv1.3 was required for optimal expression of IFN-γ and IL-17, whereas its absence led to increased IL-10 production. Dendritic cells from Kv1.3 KO mice fully activated wild-type CD4(+) T cells, indicating a T cell-intrinsic defect in Kv1.3 KO mice. The loss of Kv1.3 led to a suppressive phenotype, which may contribute to the mechanism by which deletion of Kv1.3 produces an immunotherapeutic effect. Skewing of CD4(+) T cell differentiation toward Ag-specific regulatory T cells by pharmacological blockade or genetic suppression of Kv1.3 might be beneficial for therapy of immune-mediated diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Anne R Gocke
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Lee WG, Kim WS, Park SG, Kim H, Hong J, Ko H, Kim YC. Immunosuppressive effects of subglutinol derivatives. ChemMedChem 2011; 7:218-22. [PMID: 22114006 DOI: 10.1002/cmdc.201100409] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 10/28/2011] [Indexed: 11/08/2022]
Affiliation(s)
- Won-Gil Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), 261 Cheomdan-gwagiro(Oryong- dong), Buk-gu, Gwangju 500-712, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
30
|
Valverde MA, Cantero-Recasens G, Garcia-Elias A, Jung C, Carreras-Sureda A, Vicente R. Ion channels in asthma. J Biol Chem 2011; 286:32877-82. [PMID: 21799020 DOI: 10.1074/jbc.r110.215491] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ion channels are specialized transmembrane proteins that permit the passive flow of ions following their electrochemical gradients. In the airways, ion channels participate in the production of epithelium-based hydroelectrolytic secretions and in the control of intracellular Ca(2+) levels that will ultimately activate almost all lung cells, either resident or circulating. Thus, ion channels have been the center of many studies aiming to understand asthma pathophysiological mechanisms or to identify therapeutic targets for better control of the disease. In this minireview, we focus on molecular, genetic, and animal model studies associating ion channels with asthma.
Collapse
Affiliation(s)
- Miguel A Valverde
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
31
|
Anti-proliferative effect of Kv1.3 blockers in A549 human lung adenocarcinoma in vitro and in vivo. Eur J Pharmacol 2011; 651:26-32. [DOI: 10.1016/j.ejphar.2010.10.066] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 10/07/2010] [Accepted: 10/29/2010] [Indexed: 01/05/2023]
|
32
|
Bittner S, Budde T, Wiendl H, Meuth SG. From the background to the spotlight: TASK channels in pathological conditions. Brain Pathol 2010; 20:999-1009. [PMID: 20529081 PMCID: PMC8094868 DOI: 10.1111/j.1750-3639.2010.00407.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 04/13/2010] [Indexed: 01/10/2023] Open
Abstract
TWIK-related acid-sensitive potassium channels (TASK1-3) belong to the family of two-pore domain (K(2P) ) potassium channels. Emerging knowledge about an involvement of TASK channels in cancer development, inflammation, ischemia and epilepsy puts the spotlight on a leading role of TASK channels under these conditions. TASK3 has been especially linked to cancer development. The pro-oncogenic potential of TASK3 could be shown in cell lines and in various tumor entities. Pathophysiological hallmarks in solid tumors (e.g. low pH and oxygen deprivation) regulate TASK3 channels. These conditions can also be found in (autoimmune) inflammation. Inhibition of TASK1,2,3 leads to a reduction of T cell effector function. It could be demonstrated that TASK1(-/-) mice are protected from experimental autoimmune inflammation while the same animals display increased infarct volumes after cerebral ischemia. Furthermore, TASK channels have both an anti-epileptic as well as a pro-epileptic potential. The relative contribution of these opposing influences depends on their cell type-specific expression and the conditions of the cellular environment. This indicates that TASK channels are per se neither protective nor detrimental but their functional impact depends on the "pathophysiological" scenario. Based on these findings TASK channels have evolved from "mere background" channels to key modulators in pathophysiological conditions.
Collapse
Affiliation(s)
- Stefan Bittner
- Department of Neurology, University of Wuerzburg, Wuerzburg, Germany
| | - Thomas Budde
- Institute of Physiology I, Westfaelische Wilhelms‐University Muenster, Muenster, Germany
| | - Heinz Wiendl
- Department of Neurology—Inflammatory disorders of the nervous system and neurooncology, University of Muenster, Muenster, Germany
| | - Sven G. Meuth
- Department of Neurology, University of Wuerzburg, Wuerzburg, Germany
- Department of Neurology—Inflammatory disorders of the nervous system and neurooncology, University of Muenster, Muenster, Germany
| |
Collapse
|
33
|
Wulff H. Spiro azepane-oxazolidinones as Kv1.3 potassium channel blockers: WO2010066840. Expert Opin Ther Pat 2010; 20:1759-65. [PMID: 20954790 DOI: 10.1517/13543776.2010.528392] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This article evaluates a patent application from Solvay Pharmaceuticals, which claims spiro azepane-oxazolidinones as novel blockers of the voltage-gated potassium channel Kv1.3 for the treatment of diabetes, psoriasis, obesity, transplant rejection and T-cell mediated autoimmune diseases such as rheumatoid arthritis and MS. The patent describes a new chemotype of Kv1.3 blockers and thus illustrates the growing interest of the pharmaceutical industry in Kv1.3 as a target of immunosuppression and metabolic disorders. This article briefly summarizes the chemistry and biological data provided in the patent and then compares the new compounds to Kv1.3 blockers previously disclosed by both academia and pharmaceutical companies.
Collapse
Affiliation(s)
- Heike Wulff
- University of California, Department of Pharmacology, Davis, Davis, CA 95616, USA.
| |
Collapse
|
34
|
Cheong A, Li J, Sukumar P, Kumar B, Zeng F, Riches K, Munsch C, Wood IC, Porter KE, Beech DJ. Potent suppression of vascular smooth muscle cell migration and human neointimal hyperplasia by KV1.3 channel blockers. Cardiovasc Res 2010; 89:282-9. [PMID: 20884640 PMCID: PMC3020133 DOI: 10.1093/cvr/cvq305] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIM The aim of the study was to determine the potential for K(V)1 potassium channel blockers as inhibitors of human neoinitimal hyperplasia. METHODS AND RESULTS Blood vessels were obtained from patients or mice and studied in culture. Reverse transcriptase-polymerase chain reaction and immunocytochemistry were used to detect gene expression. Whole-cell patch-clamp, intracellular calcium measurement, cell migration assays, and organ culture were used to assess channel function. K(V)1.3 was unique among the K(V)1 channels in showing preserved and up-regulated expression when the vascular smooth muscle cells switched to the proliferating phenotype. There was strong expression in neointimal formations. Voltage-dependent potassium current in proliferating cells was sensitive to three different blockers of K(V)1.3 channels. Calcium entry was also inhibited. All three blockers reduced vascular smooth muscle cell migration and the effects were non-additive. One of the blockers (margatoxin) was highly potent, suppressing cell migration with an IC(50) of 85 pM. Two of the blockers were tested in organ-cultured human vein samples and both inhibited neointimal hyperplasia. CONCLUSION K(V)1.3 potassium channels are functional in proliferating mouse and human vascular smooth muscle cells and have positive effects on cell migration. Blockers of the channels may be useful as inhibitors of neointimal hyperplasia and other unwanted vascular remodelling events.
Collapse
Affiliation(s)
- Alex Cheong
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Herrmann M, Ruprecht K, Sauter M, Martinez J, van Heteren P, Glas M, Best B, Meyerhans A, Roemer K, Mueller-Lantzsch N. Interaction of human immunodeficiency virus gp120 with the voltage-gated potassium channel BEC1. FEBS Lett 2010; 584:3513-8. [DOI: 10.1016/j.febslet.2010.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 06/18/2010] [Accepted: 07/08/2010] [Indexed: 01/04/2023]
|
36
|
Marek N, Myśliwska J, Raczyńska K, Trzonkowski P. Membrane potential of CD4+ T cells is a subset specific feature that depends on the direct cell-to-cell contacts with monocytes. Hum Immunol 2010; 71:666-75. [PMID: 20457202 DOI: 10.1016/j.humimm.2010.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 04/17/2010] [Accepted: 05/01/2010] [Indexed: 10/19/2022]
Abstract
CD4(+) T cells can be divided into three subsets: naive (Tn), central memory (Tcm), and effector memory (Tem) lymphocytes. These subpopulations differ in phenotype, migratory capacity, pattern of secreted cytokines, and activation threshold. T-cell activation is associated with changes in membrane potential, which provide an electrical driving force for calcium entry into the cytosol. These phenomena were shown to precede lymphocyte proliferation, cytokine synthesis, migration, and apoptosis. Hence the aim of the study was the analysis of these early activation events in the subsets of CD4(+) T cells. We measured the membrane potential and intracellular calcium concentration ([Ca(2+)](i)) in CD4(+) Tn, Tcm, and Tem cells as well as the dependency of these parameters in CD4(+) T cells on their cell-to-cell contacts with other leukocyte subsets. The data indicate that membrane potential of CD4(+) T cells is a subset specific feature maintained by direct contact with monocytes. In addition, monocytes were found to control Ca(2+) influx in CD4(+) T cells.
Collapse
Affiliation(s)
- Natalia Marek
- Department of Clinical Immunology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland.
| | | | | | | |
Collapse
|
37
|
Yeung CK, Law JKY, Sam SW, Ingebrandt S, Lau HYA, Rudd JA, Chan M. Modulatory action of potassium channel openers on field potential and histamine release from rat peritoneal mast cells. Can J Physiol Pharmacol 2010; 87:624-32. [PMID: 19767887 DOI: 10.1139/y09-047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To determine whether changes in membrane potential affect the extent of mast cell degranulation, compound 48/80 was added to rat peritoneal mast cell suspensions in the absence or presence of potassium channel openers (KCOs). Changes were compared between the field potential (FP) and the amount of histamine released. The results demonstrated that (i) the onset and duration of FP, which reflects the hyperpolarizing nature of the response, increased as the concentration of compound 48/80 increased; (ii) both FP and the amount of histamine released increased as the concentration of compound 48/80 increased; (iii) although both KCOs (SDZ PCO400, a benzopyran derivative, and P1060, a cyanoguanidine derivative) potentiated compound 48/80-induced increases in FP and histamine release, without compound 48/80, they had no effect on either parameter; (iv) both glibenclamide and charybdotoxin significantly attenuated the compound 48/80-induced increase in FP; and (v) glibenclamide was able to attenuate the KCO-induced potentiation of FP. The results show that drugs presumably causing hyperpolarization can affect histamine release from rat peritoneal mast cells. The effect of KCOs on compound 48/80-induced response appears to be potentiation in nature rather than synergism. It is possible that KCO hyperpolarizes the cell membrane, enhances Ca2+ influx, and thus increases histamine release. As such, selective blockers of K+ channels may be useful for the treatment of immunological disorders.
Collapse
Affiliation(s)
- Chi-Kong Yeung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | | | | | | | | | | | | |
Collapse
|
38
|
Rangaraju S, Chi V, Pennington MW, Chandy KG. Kv1.3 potassium channels as a therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2010; 13:909-24. [PMID: 19538097 DOI: 10.1517/14728220903018957] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We discuss the potential use of inhibitors of Kv1.3 potassium channels in T lymphocytes as therapeutics for multiple sclerosis. Current treatment strategies target the immune system in a non-selective manner. The resulting general immunosuppression, toxic side-effects and increased risk of opportunistic infections create the need for more selective therapeutics. Autoreactive effector-memory T (T(EM)) cells, considered to be major mediators of autoimmunity, express large numbers of Kv1.3 channels. Selective blockers of Kv1.3 inhibit calcium signaling, cytokine production and proliferation of T(EM) cells in vitro, and T(EM) cell-motility in vivo. Kv1.3 blockers ameliorate disease in animal models of multiple sclerosis, rheumatoid arthritis, type 1 diabetes mellitus and contact dermatitis without compromising the protective immune response to acute infections. Kv1.3 blockers have a good safety profile in rodents and primates.
Collapse
Affiliation(s)
- Srikant Rangaraju
- University of California, Department of Physiology and Biophysics, Irvine, California 92697, USA
| | | | | | | |
Collapse
|
39
|
TNF-α inhibits the CD3-mediated upregulation of voltage-gated K+ channel (Kv1.3) in human T cells. Biochem Biophys Res Commun 2010; 391:909-14. [DOI: 10.1016/j.bbrc.2009.11.162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 11/25/2009] [Indexed: 01/15/2023]
|
40
|
Abstract
The human genome encodes 40 voltage-gated K(+) channels (K(V)), which are involved in diverse physiological processes ranging from repolarization of neuronal and cardiac action potentials, to regulating Ca(2+) signalling and cell volume, to driving cellular proliferation and migration. K(V) channels offer tremendous opportunities for the development of new drugs to treat cancer, autoimmune diseases and metabolic, neurological and cardiovascular disorders. This Review discusses pharmacological strategies for targeting K(V) channels with venom peptides, antibodies and small molecules, and highlights recent progress in the preclinical and clinical development of drugs targeting the K(V)1 subfamily, the K(V)7 subfamily (also known as KCNQ), K(V)10.1 (also known as EAG1 and KCNH1) and K(V)11.1 (also known as HERG and KCNH2) channels.
Collapse
|
41
|
Blackiston DJ, McLaughlin KA, Levin M. Bioelectric controls of cell proliferation: ion channels, membrane voltage and the cell cycle. Cell Cycle 2009; 8:3527-36. [PMID: 19823012 DOI: 10.4161/cc.8.21.9888] [Citation(s) in RCA: 311] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
All cells possess long-term, steady-state voltage gradients across the plasma membrane. These transmembrane potentials arise from the combined activity of numerous ion channels, pumps and gap junction complexes. Increasing data from molecular physiology now reveal that the role of changes in membrane voltage controls, and is in turn controlled by, progression through the cell cycle. We review recent functional data on the regulation of mitosis by bioelectric signals, and the function of membrane voltage and specific potassium, sodium and chloride ion channels in the proliferation of embryonic, somatic and neoplastic cells. Its unique properties place this powerful, well-conserved, but still poorly-understood signaling system at the center of the coordinated cellular interactions required for complex pattern formation. Moreover, disregulation of ion channel expression and function is increasingly observed to be not only a useful marker but likely a functional element in oncogenesis. New advances in genomics and the development of in vivo biophysical techniques suggest exciting opportunities for molecular medicine, bioengineering and regenerative approaches to human health.
Collapse
Affiliation(s)
- Douglas J Blackiston
- Biology Department, and Center for Regenerative and Developmental Biology, Tufts University, Medford, MA, USA
| | | | | |
Collapse
|
42
|
Abstract
Dynamic changes in cytoplasmic calcium concentration dictate the immunological fate and functions of lymphocytes. During the past few years, important details have been revealed about the mechanism of store-operated calcium entry in lymphocytes, including the molecular identity of calcium release-activated calcium (CRAC) channels and the endoplasmic reticulum (ER) calcium sensor (STIM1) responsible for CRAC channel activation following calcium depletion of stores. However, details of the potential fine regulation of CRAC channel activation that may be imposed on lymphocytes following physiologic stimulation within an inflammatory environment have not been fully addressed. In this review, we discuss several underexplored aspects of store-operated (CRAC-mediated) and store-independent calcium signaling in B lymphocytes. First, we discuss results suggesting that coupling between stores and CRAC channels may be regulated, allowing for fine tuning of CRAC channel activation following depletion of ER stores. Second, we discuss mechanisms that sustain the duration of calcium entry via CRAC channels. Finally, we discuss distinct calcium permeant non-selective cation channels (NSCCs) that are activated by innate stimuli in B cells, the potential means by which these innate calcium signaling pathways and CRAC channels crossregulate one another, and the mechanistic basis and physiologic consequences of innate calcium signaling.
Collapse
Affiliation(s)
- Leslie B King
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
43
|
Abstract
For more than 25 years, it has been widely appreciated that Ca2+ influx is essential to trigger T-lymphocyte activation. Patch clamp analysis, molecular identification, and functional studies using blockers and genetic manipulation have shown that a unique contingent of ion channels orchestrates the initiation, intensity, and duration of the Ca2+ signal. Five distinct types of ion channels--Kv1.3, KCa3.1, Orai1+ stromal interacting molecule 1 (STIM1) [Ca2+-release activating Ca2+ (CRAC) channel], TRPM7, and Cl(swell)--comprise a network that performs functions vital for ongoing cellular homeostasis and for T-cell activation, offering potential targets for immunomodulation. Most recently, the roles of STIM1 and Orai1 have been revealed in triggering and forming the CRAC channel following T-cell receptor engagement. Kv1.3, KCa3.1, STIM1, and Orai1 have been found to cluster at the immunological synapse following contact with an antigen-presenting cell; we discuss how channels at the synapse might function to modulate local signaling. Immuno-imaging approaches are beginning to shed light on ion channel function in vivo. Importantly, the expression pattern of Ca2+ and K+ channels and hence the functional network can adapt depending upon the state of differentiation and activation, and this allows for different stages of an immune response to be targeted specifically.
Collapse
Affiliation(s)
- Michael D Cahalan
- Department of Physiology and Biophysics, and the Institute for Immunology, University of California, Irvine, Irvine, CA 92697-4561, USA.
| | | |
Collapse
|
44
|
Sundelacruz S, Levin M, Kaplan DL. Role of membrane potential in the regulation of cell proliferation and differentiation. Stem Cell Rev Rep 2009; 5:231-46. [PMID: 19562527 PMCID: PMC10467564 DOI: 10.1007/s12015-009-9080-2] [Citation(s) in RCA: 339] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 06/07/2009] [Indexed: 12/11/2022]
Abstract
Biophysical signaling, an integral regulator of long-term cell behavior in both excitable and non-excitable cell types, offers enormous potential for modulation of important cell functions. Of particular interest to current regenerative medicine efforts, we review several examples that support the functional role of transmembrane potential (V(mem)) in the regulation of proliferation and differentiation. Interestingly, distinct V(mem) controls are found in many cancer cell and precursor cell systems, which are known for their proliferative and differentiation capacities, respectively. Collectively, the data demonstrate that bioelectric properties can serve as markers for cell characterization and can control cell mitotic activity, cell cycle progression, and differentiation. The ability to control cell functions by modulating bioelectric properties such as V(mem) would be an invaluable tool for directing stem cell behavior toward therapeutic goals. Biophysical properties of stem cells have only recently begun to be studied and are thus in need of further characterization. Understanding the molecular and mechanistic basis of biophysical regulation will point the way toward novel ways to rationally direct cell functions, allowing us to capitalize upon the potential of biophysical signaling for regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA 02155, USA
| | | | | |
Collapse
|
45
|
Bradding P, Wulff H. The K+ channels K(Ca)3.1 and K(v)1.3 as novel targets for asthma therapy. Br J Pharmacol 2009; 157:1330-9. [PMID: 19681865 PMCID: PMC2765317 DOI: 10.1111/j.1476-5381.2009.00362.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 05/06/2009] [Accepted: 05/11/2009] [Indexed: 01/22/2023] Open
Abstract
Asthma affects 10% of the UK population and is an important cause of morbidity and mortality at all ages. Current treatments are either ineffective or carry unacceptable side effects for a number of patients; in consequence, development of new approaches to therapy are important. Ion channels are emerging as attractive therapeutic targets in a variety of non-excitable cells. Ion channels conducting K(+) modulate the activity of several structural and inflammatory cells which play important roles in the pathophysiology of asthma. Two channels of particular interest are the voltage-gated K(+) channel K(v)1.3 and the intermediate conductance Ca(2+)-activated K(+) channel K(Ca)3.1 (also known as IK(Ca)1 or SK4). K(v)1.3 is expressed in IFNgamma-producing T cells while K(Ca)3.1 is expressed in T cells, mast cells, macrophages, airway smooth muscle cells, fibroblasts and epithelial cells. Both channels play important roles in cell activation, migration, and proliferation through the regulation of membrane potential and calcium signalling. We hypothesize that K(Ca)3.1- and/or K(v)1.3-dependent cell processes are one of the common denominators in asthma pathophysiology. If true, these channels might serve as novel targets for the treatment of asthma. Emerging evidence lends support to this hypothesis. Further validation through the study of the role that these channels play in normal and asthmatic airway cell (patho)physiology and in vivo models will provide further justification for the assessment of small molecule blockers of K(v)1.3 and K(Ca)3.1 in the treatment of asthma.
Collapse
Affiliation(s)
- Peter Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, UK.
| | | |
Collapse
|
46
|
Schneider G, Hartenfeller M, Reutlinger M, Tanrikulu Y, Proschak E, Schneider P. Voyages to the (un)known: adaptive design of bioactive compounds. Trends Biotechnol 2009; 27:18-26. [DOI: 10.1016/j.tibtech.2008.09.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 09/14/2008] [Accepted: 09/17/2008] [Indexed: 11/30/2022]
|
47
|
Mello de Queiroz F, Ponte CG, Bonomo A, Vianna-Jorge R, Suarez-Kurtz G. Study of membrane potential in T lymphocytes subpopulations using flow cytometry. BMC Immunol 2008; 9:63. [PMID: 18980671 PMCID: PMC2584624 DOI: 10.1186/1471-2172-9-63] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 11/03/2008] [Indexed: 11/10/2022] Open
Abstract
Background Ion channels are involved in the control of membrane potential (ψ) in a variety of cells. The maintenance of ψ in human T lymphocytes is essential for T-cell activation and was suggested to depend mostly on the voltage-gated Kv1.3 channel. Blockage of Kv1.3 inhibits cytokine production and lymphocyte proliferation in vitro and suppresses immune response in vivo. T lymphocytes are a heterogeneous cell population and the expression of Kv1.3 varies among cell subsets. Oxonol diBA-C4-(3) was used to determine ψ by flow cytometry. The presence of distinct T cell subsets was evaluated by immunophenotyping techniques and the contribution of Kv1.3 channels for the maintenance of ψ was investigated using selective blockers. Results The distribution of ψ in T lymphocytes varied among blood donors and did not always follow a unimodal pattern. T lymphocytes were divided into CD3+/CD45RO- and CD3+/CD45RO+ subsets, whose peak channel values of ψ were -58 ± 3.6 mV and -37 ± 4.1 mV, respectively. MgTX (specific inhibitor of Kv1.3 channels) had no significant effect in the ψ of CD3+/CD45RO- subsets but depolarized CD3+/CD45RO+ cells to -27 ± 5.1 mV. Conclusion Combination of optical methods for determination of ψ by flow cytometry with immuophenotyping techniques opens new possibilities for the study of ion channels in the biology of heterogeneous cell populations such as T lymphocyte subsets.
Collapse
Affiliation(s)
- Fernanda Mello de Queiroz
- Molekulare Biologie Neuronaler Signale, Max-Planck-Institut für Experimentelle Medizin, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany.
| | | | | | | | | |
Collapse
|
48
|
Saito H. Progress in allergy signal research on mast cells: systemic approach to mast cell biology in allergic diseases. J Pharmacol Sci 2008; 106:341-6. [PMID: 18360090 DOI: 10.1254/jphs.fm0070192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
At the end of the last century, microarray technology that examines the total genes and transcripts present in a cell became available as a laboratory tool. Mast cells are known to play a pivotal role in initiating allergic inflammation by releasing various mediators and cytokines. According to the recent microarray-based studies, mast cells have been found to be much more versatile functional molecules than we ever thought. Also, genes that are exclusively expressed in mast cells have been identified in comparison with other cell types. In this article, the outcome of microarray-based analyses on the role of mast cells in allergic inflammation will be reviewed by focusing on the mast cell-specific genes as drug targets.
Collapse
Affiliation(s)
- Hirohisa Saito
- Department of Allergy and Immunology, National Research Institute for Child Health & Development, Japan.
| |
Collapse
|
49
|
Meuth SG, Herrmann AM, Ip CW, Kanyshkova T, Bittner S, Weishaupt A, Budde T, Wiendl H. The two-pore domain potassium channel TASK3 functionally impacts glioma cell death. J Neurooncol 2008; 87:263-70. [PMID: 18217213 DOI: 10.1007/s11060-008-9517-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Accepted: 01/02/2008] [Indexed: 01/21/2023]
Abstract
Two-pore domain K(+) channels, a recently discovered family of ion channels with a unique membrane topology, have been shown to be critically involved in cell death. We here address the functional role of TASK3 (TWIK-related acid-sensitive K(+) channel, KCNK9) in human glioblastoma in vitro and in vivo. Human glioma cell lines (n = 5) as well as glioma specimens (n = 5) constitutively express TASK3 mRNA and protein. The functional impact of the potassium channel on cell survival was investigated using a medium with high (25 mM) extracellular potassium over 7 days. Using flow cytometric assessment, we show that under these culture conditions 97 +/- 0.76% of all glioma cells survived. Application of the TASK channel opener isoflurane (1 vol%) resulted in a 30 +/- 4% reduction of cell survival in different glioma cell lines. Simultaneous application of isoflurane and the TASK channel blockers bupivacaine (20 microM) and spermine (500 microM) completely reversed this effect. Our results demonstrate the expression of TASK3 in glioma cells in vitro and in vivo and provide a direct link between the TASK3 channel function and glioma cell survival. This implies that TASK3 channels may possibly represent a novel molecular target for the treatment of this type of cancer.
Collapse
Affiliation(s)
- Sven G Meuth
- Department of Neurology, University of Würzburg, Josef-Schneider Str. 11, 97080 Wurzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Beeton C, Smith BJ, Sabo JK, Crossley G, Nugent D, Khaytin I, Chi V, Chandy KG, Pennington MW, Norton RS. The D-Diastereomer of ShK Toxin Selectively Blocks Voltage-gated K+ Channels and Inhibits T Lymphocyte Proliferation. J Biol Chem 2008; 283:988-97. [DOI: 10.1074/jbc.m706008200] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|