1
|
Varanita T, Angi B, Scattolini V, Szabo I. Kv1.3 K + Channel Physiology Assessed by Genetic and Pharmacological Modulation. Physiology (Bethesda) 2023; 38:0. [PMID: 35998249 DOI: 10.1152/physiol.00010.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Potassium channels are widespread over all kingdoms and play an important role in the maintenance of cellular ionic homeostasis. Kv1.3 is a voltage-gated potassium channel of the Shaker family with a wide tissue expression and a well-defined pharmacology. In recent decades, experiments mainly based on pharmacological modulation of Kv1.3 have highlighted its crucial contribution to different fundamental processes such as regulation of proliferation, apoptosis, and metabolism. These findings link channel function to various pathologies ranging from autoimmune diseases to obesity and cancer. In the present review, we briefly summarize studies employing Kv1.3 knockout animal models to confirm such roles and discuss the findings in comparison to the results obtained by pharmacological modulation of Kv1.3 in various pathophysiological settings. We also underline how these studies contributed to our understanding of channel function in vivo and propose possible future directions.
Collapse
Affiliation(s)
| | - Beatrice Angi
- Department of Biology, University of Padova, Padova, Italy
| | | | - Ildiko Szabo
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
2
|
Wright JR, Jones S, Parvathy S, Kaczmarek LK, Forsythe I, Farndale RW, Gibbins JM, Mahaut-Smith MP. The voltage-gated K + channel Kv1.3 modulates platelet motility and α 2β 1 integrin-dependent adhesion to collagen. Platelets 2022; 33:451-461. [PMID: 34348571 PMCID: PMC8935947 DOI: 10.1080/09537104.2021.1942818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022]
Abstract
Kv1.3 is a voltage-gated K+-selective channel with roles in immunity, insulin-sensitivity, neuronal excitability and olfaction. Despite being one of the largest ionic conductances of the platelet surface membrane, its contribution to platelet function is poorly understood. Here we show that Kv1.3-deficient platelets display enhanced ADP-evoked platelet aggregation and secretion, and an increased surface expression of platelet integrin αIIb. In contrast, platelet adhesion and thrombus formation in vitro under arterial shear conditions on surfaces coated with collagen were reduced for samples from Kv1.3-/- compared to wild type mice. Use of collagen-mimetic peptides revealed a specific defect in the engagement with α2β1. Kv1.3-/- platelets developed significantly fewer, and shorter, filopodia than wild type platelets during adhesion to collagen fibrils. Kv1.3-/- mice displayed no significant difference in thrombus formation within cremaster muscle arterioles using a laser-induced injury model, thus other pro-thrombotic pathways compensate in vivo for the adhesion defect observed in vitro. This may include the increased platelet counts of Kv1.3-/- mice, due in part to a prolonged lifespan. The ability of Kv1.3 to modulate integrin-dependent platelet adhesion has important implications for understanding its contribution to normal physiological platelet function in addition to its reported roles in auto-immune diseases and thromboinflammatory models of stroke.
Collapse
Affiliation(s)
- Joy R Wright
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Sarah Jones
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Sasikumar Parvathy
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Leonard K Kaczmarek
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, USA
| | - Ian Forsythe
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | | | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | | |
Collapse
|
3
|
Veuthey L, Aliotta A, Bertaggia Calderara D, Pereira Portela C, Alberio L. Mechanisms Underlying Dichotomous Procoagulant COAT Platelet Generation-A Conceptual Review Summarizing Current Knowledge. Int J Mol Sci 2022; 23:2536. [PMID: 35269679 PMCID: PMC8910683 DOI: 10.3390/ijms23052536] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 12/23/2022] Open
Abstract
Procoagulant platelets are a subtype of activated platelets that sustains thrombin generation in order to consolidate the clot and stop bleeding. This aspect of platelet activation is gaining more and more recognition and interest. In fact, next to aggregating platelets, procoagulant platelets are key regulators of thrombus formation. Imbalance of both subpopulations can lead to undesired thrombotic or bleeding events. COAT platelets derive from a common pro-aggregatory phenotype in cells capable of accumulating enough cytosolic calcium to trigger specific pathways that mediate the loss of their aggregating properties and the development of new adhesive and procoagulant characteristics. Complex cascades of signaling events are involved and this may explain why an inter-individual variability exists in procoagulant potential. Nowadays, we know the key agonists and mediators underlying the generation of a procoagulant platelet response. However, we still lack insight into the actual mechanisms controlling this dichotomous pattern (i.e., procoagulant versus aggregating phenotype). In this review, we describe the phenotypic characteristics of procoagulant COAT platelets, we detail the current knowledge on the mechanisms of the procoagulant response, and discuss possible drivers of this dichotomous diversification, in particular addressing the impact of the platelet environment during in vivo thrombus formation.
Collapse
Affiliation(s)
| | | | | | | | - Lorenzo Alberio
- Hemostasis and Platelet Research Laboratory, Division of Hematology and Central Hematology Laboratory, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), CH-1010 Lausanne, Switzerland; (L.V.); (A.A.); (D.B.C.); (C.P.P.)
| |
Collapse
|
4
|
Wright JR, Mahaut-Smith MP. Why do platelets express K + channels? Platelets 2021; 32:872-879. [PMID: 33872124 PMCID: PMC8437091 DOI: 10.1080/09537104.2021.1904135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 11/02/2022]
Abstract
Potassium ions have widespread roles in cellular homeostasis and activation as a consequence of their large outward concentration gradient across the surface membrane and ability to rapidly move through K+-selective ion channels. In platelets, the predominant K+ channels include the voltage-gated K+ channel Kv1.3, and the intermediate conductance Ca2+-activated K+ channel KCa3.1, also known as the Gardos channel. Inwardly rectifying potassium GIRK channels and KCa1.1 large conductance Ca2+-activated K+ channels have also been reported in the platelet, although they remain to be demonstrated using electrophysiological techniques. Whole-cell patch clamp and fluorescent indicator measurements in the platelet or their precursor cell reveal that Kv1.3 sets the resting membrane potential and KCa3.1 can further hyperpolarize the cell during activation, thereby controlling Ca2+ influx. Kv1.3-/- mice exhibit an increased platelet count, which may result from an increased splenic megakaryocyte development and longer platelet lifespan. This review discusses the evidence in the literature that Kv1.3, KCa3.1. GIRK and KCa1.1 channels contribute to a number of platelet functional responses, particularly collagen-evoked adhesion, procoagulant activity and GPCR function. Putative roles for other K+ channels and known accessory proteins which to date have only been detected in transcriptomic or proteomic studies, are also discussed.
Collapse
Affiliation(s)
- Joy R Wright
- Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Leicester, UK
| | | |
Collapse
|
5
|
Li M, Yu H. Identification of WP1066, an inhibitor of JAK2 and STAT3, as a K V 1.3 potassium channel blocker. Br J Pharmacol 2021; 178:2617-2631. [PMID: 33689167 DOI: 10.1111/bph.15441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 01/15/2021] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE KV 1.3 potassium channels play a predominant role in regulating calcium signalling that is essential for the activation and proliferation of effector memory T (TEM ) cells. This ion channel has been recognized as a promising therapeutic target against various autoimmune diseases. EXPERIMENTAL APPROACH In a high-throughput screening programme, WP1066 was identified as a KV 1.3 channel inhibitor. Using molecular biology and electrophysiological methods, the mechanism(s) underlying WP1066 blockade of Kv1.3 channels was investigated. Using TEM cell proliferation assay and mouse delayed-type hypersensitivity (DTH) model, the effects of WP1066 were examined. KEY RESULTS WP1066 blocked KV 1.3 channels in a dose-dependent manner with an IC50 of 3.2 μM and induced a hyperpolarizing shift of the steady-state inactivation curve. This blockade was use-dependent, as WP1066 interacted preferentially with channels in their open state, rather than the closed state or inactivated state. When the residues located in the S6 domain scaffolding the inner vestibule, were sequentially mutated, the potency of WP1066 was significantly impaired, especially by mutations A413C and I420C, indicating a higher affinity of interacting sites for WP1066. Moreover, WP1066 effectively suppressed mouse TEM cell proliferation in vitro and mouse DTH reaction in vivo. CONCLUSIONS AND IMPLICATIONS The results presented here have identified WP1066 as a KV 1.3 channel blocker with an open-state-dependent property, providing fundamental evidence for the application of WP1066 in further immunomodulatory studies targeting KV 1.3 channels.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haibo Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Abstract
Monoclonal antibodies combine specificity and high affinity binding with excellent pharmacokinetic properties and are rapidly being developed for a wide range of drug targets including clinically important potassium ion channels. Nonetheless, while therapeutic antibodies come with great promise, K+ channels represent particularly difficult targets for biologics development for a variety of reasons that include their dynamic structures and relatively small extracellular loops, their high degree of sequence conservation (leading to immune tolerance), and their generally low-level expression in vivo. The process is made all the more difficult when large numbers of antibody candidates must be screened for a given target, or when lead candidates fail to cross-react with orthologous channels in animal disease models due to their highly selective binding properties. While the number of antibodies targeting potassium channels in preclinical or clinical development is still modest, significant advances in the areas of protein expression and antibody screening are converging to open the field to an avalanche of new drugs. Here, the opportunities and constraints associated with the discovery of antibodies against K+ channels are discussed, with an emphasis on novel technologies that are opening the field to exciting new possibilities for biologics development.
Collapse
|
7
|
Fan C, Yang X, Wang WW, Wang J, Li W, Guo M, Huang S, Wang Z, Liu K. Role of Kv1.3 Channels in Platelet Functions and Thrombus Formation. Arterioscler Thromb Vasc Biol 2020; 40:2360-2375. [PMID: 32787516 DOI: 10.1161/atvbaha.120.314278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective:
Platelet activation by stimulatory factors leads to an increase in intracellular calcium concentration ([Ca
2+
]
i
), which is essential for almost all platelet functions. Modulation of Ca
2+
influx and [Ca
2+
]
i
in platelets has been emerging as a possible strategy for preventing and treating platelet-dependent thrombosis. Voltage-gated potassium 1.3 channels (Kv1.3) are highly expressed in platelets and able to regulate agonist-evoked [Ca
2+
]
i
increase. However, the role of Kv1.3 channels in regulating platelet functions and thrombosis has not yet been elucidated. In addition, it is difficult to obtain a specific blocker for this channel, since Kv1.3 shares identical drug-binding sites with other K
+
channels. Here, we investigate whether specific blockade of Kv1.3 channels by monoclonal antibodies affects platelet functions and thrombosis.
Approach and Results:
In this study, we produced the anti-Kv1.3 monoclonal antibody 6E12#15, which could specifically recognize both human and mouse Kv1.3 proteins and sufficiently block Kv1.3 channel currents. We found Kv1.3 blockade by 6E12#15 inhibited platelet aggregation, adhesion, and activation upon agonist stimulation. In vivo treatment with 6E12#15 alleviated thrombus formation in a mesenteric arteriole thrombosis mouse model and protected mice from collagen/epinephrine-induced pulmonary thromboembolism. Furthermore, we observed Kv1.3 regulated platelet functions by modulating Ca
2+
influx and [Ca
2+
]
i
elevation, and that this is mediated in part by P2X
1
. Interestingly,
Kv1.3
−/−
mice showed impaired platelet aggregation while displayed no abnormalities in in vivo thrombus formation. This phenomenon was related to more megakaryocytes and platelets produced in
Kv1.3
−/−
mice compared with wild-type mice.
Conclusions:
We showed specific inhibition of Kv1.3 by the novel monoclonal antibody 6E12#15 suppressed platelet functions and platelet-dependent thrombosis through modulating platelet [Ca
2+
]
i
elevation. These results indicate that Kv1.3 could act as a promising therapeutic target for antiplatelet therapies.
Collapse
Affiliation(s)
- Cheng Fan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.F., M.G., S.H., Z.W.)
| | - Xiaofang Yang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, China (X.Y.)
| | | | - Jue Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.W.)
| | - Wenzhu Li
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (W.L.)
| | - Mengyuan Guo
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.F., M.G., S.H., Z.W.)
| | - Shiyuan Huang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.F., M.G., S.H., Z.W.)
| | - Zhaohui Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.F., M.G., S.H., Z.W.)
| | - Kun Liu
- Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (K.L.)
| |
Collapse
|
8
|
Moreno-Estar S, Serrano S, Arévalo-Martínez M, Cidad P, López-López JR, Santos M, Pérez-Garcia MT, Arias FJ. Elastin-like recombinamer-based devices releasing Kv1.3 blockers for the prevention of intimal hyperplasia: An in vitro and in vivo study. Acta Biomater 2020; 115:264-274. [PMID: 32771595 DOI: 10.1016/j.actbio.2020.07.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 12/16/2022]
Abstract
Coronary artery disease (CAD) is the most common cardiovascular disorder. Vascular surgery strategies for coronary revascularization (either percutaneous or open) show a high rate of failure because of restenosis of the vessel, due to phenotypic switch of vascular smooth muscle cells (VSMCs) leading to proliferation and migration. We have previously reported that the inhibition of Kv1.3 channel function with selective blockers represents an effective strategy for the prevention of restenosis in human vessels used for coronary angioplasty procedures. However, delivery systems for controlled release of these drugs have not been investigated. Here we tested the efficacy of several formulations of elastin like recombinamers (ELRs) hydrogels to deliver the Kv1.3 blocker PAP-1 in various restenosis models. The dose and time course of PAP-1 release from ELRs click hydrogels was able to inhibit human VSMC proliferation in vitro as well as remodeling of human vessels in organ culture and restenosis in in vivo models. We conclude that this combination of active compound and advanced delivery method could improve the outcomes of vascular surgery in patients. STATEMENT OF SIGNIFICANCE: Vascular surgery strategies for coronary revascularization show a high rate of failure, because of occlusion (restenosis) of the vessel, due to vascular smooth muscle cells proliferation and migration. We have previously reported that blockers of Kv1.3 channels represent an effective anti-restenosis therapy, but delivery systems for their controlled release have not being explored. Here we tested the efficacy of several formulations of elastin like recombinamers (ELRs) hydrogels to deliver the Kv1.3 blocker PAP-1 in various restenosis models, both in vivo and in vitro, and also in human vessels. We demonstrated that combination of active compound and advanced delivery method could improve the outcomes of vascular surgery in patients.
Collapse
|
9
|
Chow LWC, Leung YM. The versatile Kv channels in the nervous system: actions beyond action potentials. Cell Mol Life Sci 2020; 77:2473-2482. [PMID: 31894358 PMCID: PMC11104815 DOI: 10.1007/s00018-019-03415-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/16/2019] [Accepted: 12/09/2019] [Indexed: 12/22/2022]
Abstract
Voltage-gated K+ (Kv) channel opening repolarizes excitable cells by allowing K+ efflux. Over the last two decades, multiple Kv functions in the nervous system have been found to be unrelated to or beyond the immediate control of excitability, such as shaping action potential contours or regulation of inter-spike frequency. These functions include neuronal exocytosis and neurite formation, neuronal cell death, regulation of astrocyte Ca2+, glial cell and glioma proliferation. Some of these functions have been shown to be independent of K+ conduction, that is, they suggest the non-canonical functions of Kv channels. In this review, we focus on neuronal or glial plasmalemmal Kv channel functions which are unrelated to shaping action potentials or immediate control of excitability. Similar functions in other cell types will be discussed to some extent in appropriate contexts.
Collapse
Affiliation(s)
- Louis W C Chow
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
- UNIMED Medical Institute, Hong Kong, China
- Organisation for Oncology and Translational Research, Hong Kong, China
| | - Yuk- Man Leung
- Department of Physiology, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
10
|
Arévalo-Martínez M, Cidad P, García-Mateo N, Moreno-Estar S, Serna J, Fernández M, Swärd K, Simarro M, de la Fuente MA, López-López JR, Pérez-García MT. Myocardin-Dependent Kv1.5 Channel Expression Prevents Phenotypic Modulation of Human Vessels in Organ Culture. Arterioscler Thromb Vasc Biol 2019; 39:e273-e286. [DOI: 10.1161/atvbaha.119.313492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective:
We have previously described that changes in the expression of Kv channels associate to phenotypic modulation (PM), so that Kv1.3/Kv1.5 ratio is a landmark of vascular smooth muscle cells phenotype. Moreover, we demonstrated that the Kv1.3 functional expression is relevant for PM in several types of vascular lesions. Here, we explore the efficacy of Kv1.3 inhibition for the prevention of remodeling in human vessels, and the mechanisms linking the switch in Kv1.3 /Kv1.5 ratio to PM.
Approach and Results:
Vascular remodeling was explored using organ culture and primary cultures of vascular smooth muscle cells obtained from human vessels. We studied the effects of Kv1.3 inhibition on serum-induced remodeling, as well as the impact of viral vector-mediated overexpression of Kv channels or myocardin knock-down. Kv1.3 blockade prevented remodeling by inhibiting proliferation, migration, and extracellular matrix secretion. PM activated Kv1.3 via downregulation of Kv1.5. Hence, both Kv1.3 blockers and Kv1.5 overexpression inhibited remodeling in a nonadditive fashion. Finally, myocardin knock-down induced vessel remodeling and Kv1.5 downregulation and myocardin overexpression increased Kv1.5, while Kv1.5 overexpression inhibited PM without changing myocardin expression.
Conclusions:
We demonstrate that Kv1.5 channel gene is a myocardin-regulated, vascular smooth muscle cells contractile marker. Kv1.5 downregulation upon PM leaves Kv1.3 as the dominant Kv1 channel expressed in dedifferentiated cells. We demonstrated that the inhibition of Kv1.3 channel function with selective blockers or by preventing Kv1.5 downregulation can represent an effective, novel strategy for the prevention of intimal hyperplasia and restenosis of the human vessels used for coronary angioplasty procedures.
Collapse
Affiliation(s)
- Marycarmen Arévalo-Martínez
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Pilar Cidad
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Nadia García-Mateo
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Sara Moreno-Estar
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Julia Serna
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Mirella Fernández
- Cardiovascular Surgery Department, Hospital Clínico Universitario de Valladolid, Spain (M.F.)
| | - Karl Swärd
- Department of Experimental Medical Science, University of Lund, Sweden (K.S.)
| | - María Simarro
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
- Departamento de Enfermería, Universidad de Valladolid, Spain (M.S.)
| | - Miguel A. de la Fuente
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
- Departamento de Biología Celular, Universidad de Valladolid, Spain (M.A.d.l.F.)
| | - José R. López-López
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
| | - M. Teresa Pérez-García
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
| |
Collapse
|
11
|
Taylor KA, Mahaut-Smith MP. A major interspecies difference in the ionic selectivity of megakaryocyte Ca 2+-activated channels sensitive to the TMEM16F inhibitor CaCCinh-A01. Platelets 2019; 30:962-966. [PMID: 31008669 PMCID: PMC6816474 DOI: 10.1080/09537104.2019.1595560] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/12/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022]
Abstract
TMEM16F is a surface membrane protein critical for platelet procoagulant activity, which exhibits both phospholipid scramblase and ion channel activities following sustained elevation of cytosolic Ca2+. The extent to which the ionic permeability of TMEM16F is important for platelet scramblase responses remains controversial. To date, only one study has reported the electrophysiological properties of TMEM16F in cells of platelet/megakaryocyte lineage, which observed cation-selectivity within excised patch recordings from murine marrow-derived megakaryocytes. This contrasts with reports using whole-cell recordings that describe this channel as displaying either selectivity for anions or being relatively non-selective amongst the major physiological monovalent ions. We have studied TMEM16F expression and channel activity in primary rat and mouse megakaryocytes and the human erythroleukemic (HEL) cell line that exhibits megakaryocytic surface markers. Immunocytochemical analysis was consistent with surface TMEM16F expression in cells from all three species. Whole-cell recordings in the absence of K+-selective currents revealed an outwardly rectifying conductance activated by a high intracellular Ca2+ concentration in all three species. These currents appeared after 5-6 minutes and were blocked by CaCCinh-A01, properties typical of TMEM16F. Ion substitution experiments showed that the underlying conductance was predominantly Cl--permeable in rat megakaryocytes and HEL cells, yet non-selective between monovalent anions and cations in mouse megakaryocytes. In conclusion, the present study further highlights the difference in ionic selectivity of TMEM16F in platelet lineage cells of the mouse compared to other mammalian species. This provides additional support for the ionic "leak" hypothesis that the scramblase activity of TMEM16F does not rely upon its ability to conduct ions of a specific type.
Collapse
Affiliation(s)
- Kirk A. Taylor
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- National Heart and Lung Institute, Cardio-respiratory Section, Imperial College London, London, UK
| | | |
Collapse
|
12
|
Janicki PK, Eyileten C, Ruiz-Velasco V, Pordzik J, Czlonkowska A, Kurkowska-Jastrzebska I, Sugino S, Imamura Kawasawa Y, Mirowska-Guzel D, Postula M. Increased burden of rare deleterious variants of the KCNQ1 gene in patients with large‑vessel ischemic stroke. Mol Med Rep 2019; 19:3263-3272. [PMID: 30816480 DOI: 10.3892/mmr.2019.9987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/30/2019] [Indexed: 11/06/2022] Open
Abstract
The impact of rare and damaging variants in genes associated with platelet function in large‑vessel ischemic stroke (LVIS) remains unknown. The aim of this study was to investigate the contribution of some of these variants to the genetic susceptibility to LVIS in Polish patients using a deep re‑sequencing of 54 selected genes, coding for proteins associated with altered platelet function. Targeted pooled re‑sequencing (Illumina HiSeq 2500) was performed on genomic DNA of 500 cases (patients with history of clinically proven diagnosis of LVIS) and 500 age‑, smoking status‑, and sex‑matched controls (no history of any type of stroke), and from the same population as patients with LVIS. After quality control and prioritization based on allele frequency and damaging probability, individual genotyping of all deleterious rare variants was performed in patients from the original cohort, and stratified to concomitant cardiac conditions differing between the study and stroke groups. We demonstrated a statistically significant increase in the number of rare and potentially damaging variants in some of the investigated genes in the LVIS pool (an increase in the genomic variants burden). Furthermore, we identified an association between LVIS and 6 rare functional and damaging variants in the Kv7.1 potassium channel gene (KCNQ1). The predicted functional properties (partial loss‑of function) for the three most damaging variants in KCNQ1 coding locus were further confirmed in vitro by analyzing the membrane potential changes in cell lines co‑transfected heterogeneously with human muscarinic type 1 receptor and wild‑type or mutated KCNQ1 cDNA constructs using fluorescence imaging plate reader. The study demonstrated an increased rare variants burden for 54 genes associated with platelet function, and identified a putative role for rare damaging variants in the KCNQ1 gene on LVIS susceptibility in the Polish population.
Collapse
Affiliation(s)
- Piotr K Janicki
- Perioperative Genomics Laboratory, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw 02‑097, Poland
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Justyna Pordzik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw 02‑097, Poland
| | - Anna Czlonkowska
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw 02‑097, Poland
| | | | - Shigekazu Sugino
- Perioperative Genomics Laboratory, Penn State College of Medicine, Hershey, PA 17033, USA
| | | | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw 02‑097, Poland
| | - Marek Postula
- Perioperative Genomics Laboratory, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
13
|
Ding L, Hao J, Luo X, Zhu W, Wu Z, Qian Y, Hu F, Liu T, Ruan X, Li S, Li J, Chen Z. The Kv1.3 channel-inhibitory toxin BF9 also displays anticoagulant activity via inhibition of factor XIa. Toxicon 2018; 152:9-15. [PMID: 30012473 DOI: 10.1016/j.toxicon.2018.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 01/13/2023]
Abstract
The Kv1.3 channel plays potential roles in immune, inflammation and coagulation system. Many studies showed that Kv1.3 channel inhibitors have immunosuppressive and anti-inflammatory activities, but no Kv1.3 channel inhibitors have been found to have anticoagulation activities. Here, based on our previous work about Kv1.3 channel toxin peptide inhibitors, we first attempt to test anticoagulation activities of four known venom-derived Kv1.3 channel inhibitors with different structural folds: BmKTX with CSα/β structural fold, OmTx3 with CSα/α structural fold, BF9 with Kuntz-type structural fold, and SjAPI-2 with Ascaris-type structural fold. Our results showed that BmKTX and OmTx3 have no activities towards both intrinsic and extrinsic coagulation pathway, SjAPI-2 just has weak activity towards intrinsic coagulation pathway, and BF9 has potent activity towards intrinsic coagulation pathway with no apparent effect on extrinsic coagulation pathway. Enzyme and inhibitor reaction kinetics experiments further showed that BF9 inhibited intrinsic coagulation pathway-associated coagulation factor XIa, but have no apparent effects on common coagulation pathway coagulation factor IIa. Structure-activity relationship showed that Gly14, Asn17, Ala18 and Ile20 of BF9 are main residues involved in the inhibiting effect on factor XIa. To the best of our knowledge, BF9 is the first anticoagulant with Kv1.3 channel inhibitory activity. Together, our present studies found the first dual functional peptides with Kv1.3 channel and coagulation factor XIa inhibitory activities, and provided a new molecular template for the lead drug discovery towards immune and thrombosis-associated human diseases.
Collapse
Affiliation(s)
- Li Ding
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China; Department of Clinical Laboratory, Dongfeng Hospital, Hubei University of Medicine, Hubei, China
| | - Jinbo Hao
- Department of Clinical Laboratory, Shiyan Occupational Disease Hospital, Hubei, China
| | - Xudong Luo
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Wen Zhu
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Zheng Wu
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Yi Qian
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Fangfang Hu
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Tianli Liu
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Xuzhi Ruan
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Shan Li
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Jian Li
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China
| | - Zongyun Chen
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Hubei, China.
| |
Collapse
|
14
|
Leiva O, Leon C, Kah Ng S, Mangin P, Gachet C, Ravid K. The role of extracellular matrix stiffness in megakaryocyte and platelet development and function. Am J Hematol 2018; 93:430-441. [PMID: 29247535 DOI: 10.1002/ajh.25008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/16/2022]
Abstract
The extracellular matrix (ECM) is a key acellular structure in constant remodeling to provide tissue cohesion and rigidity. Deregulation of the balance between matrix deposition, degradation, and crosslinking results in fibrosis. Bone marrow fibrosis (BMF) is associated with several malignant and nonmalignant pathologies severely affecting blood cell production. BMF results from abnormal deposition of collagen fibers and enhanced lysyl oxidase-mediated ECM crosslinking within the marrow, thereby increasing marrow stiffness. Bone marrow stiffness has been recently recognized as an important regulator of blood cell development, notably by modifying the fate and differentiation process of hematopoietic or mesenchymal stem cells. This review surveys the different components of the ECM and their influence on stem cell development, with a focus on the impact of the ECM composition and stiffness on the megakaryocytic lineage in health and disease. Megakaryocyte maturation and the biogenesis of their progeny, the platelets, are thought to respond to environmental mechanical forces through a number of mechanosensors, including integrins and mechanosensitive ion channels, reviewed here.
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine; Whitaker Cardiovascular Institute, Boston University School of Medicine; Boston Massachusetts
| | - Catherine Leon
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S 949, FMTS; Strasbourg F-67000 France
| | - Seng Kah Ng
- Department of Medicine; Whitaker Cardiovascular Institute, Boston University School of Medicine; Boston Massachusetts
| | - Pierre Mangin
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S 949, FMTS; Strasbourg F-67000 France
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S 949, FMTS; Strasbourg F-67000 France
| | - Katya Ravid
- Department of Medicine; Whitaker Cardiovascular Institute, Boston University School of Medicine; Boston Massachusetts
| |
Collapse
|
15
|
Taylor KA, Wilson DGS, Harper MT, Pugh N. Extracellular chloride is required for efficient platelet aggregation. Platelets 2017; 29:79-83. [PMID: 28727479 PMCID: PMC5796487 DOI: 10.1080/09537104.2017.1332367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Anion channels perform a diverse range of functions and have been implicated in ATP release, volume regulation, and phosphatidylserine exposure. Platelets have been shown to express several anion channels but their function is incompletely understood. Due to a paucity of specific pharmacological blockers, we investigated the effect of extracellular chloride substitution on platelet activation using aggregometry and flow cytometry. In the absence of extracellular chloride, we observed a modest reduction of the maximum aggregation response to thrombin or collagen-related peptide. However, the rate of aggregation was substantially reduced in a manner that was dependent on the extracellular chloride concentration and aggregation in the absence of chloride was noticeably biphasic, indicative of impaired secondary signaling. This was further investigated by targeting secondary agonists with aspirin and apyrase or by blockade of the ADP receptor P2Y12. Under these conditions, the rates of aggregation were comparable to those recorded in the absence of extracellular chloride. Finally, we assessed platelet granule release by flow cytometry and report a chloride-dependent element of alpha, but not dense, granule secretion. Taken together these data support a role for anion channels in the efficient induction of platelet activation, likely via enhancement of secondary signaling pathways.
Collapse
Affiliation(s)
- Kirk A Taylor
- a Department of Biomedical and Forensic Sciences , Anglia Ruskin University , Cambridge , UK
| | - Darren G S Wilson
- a Department of Biomedical and Forensic Sciences , Anglia Ruskin University , Cambridge , UK
| | - Matthew T Harper
- b Department of Pharmacology , Cambridge University , Cambridge , UK
| | - Nicholas Pugh
- a Department of Biomedical and Forensic Sciences , Anglia Ruskin University , Cambridge , UK
| |
Collapse
|
16
|
Venom-derived peptide inhibitors of voltage-gated potassium channels. Neuropharmacology 2017; 127:124-138. [PMID: 28689025 DOI: 10.1016/j.neuropharm.2017.07.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/02/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022]
Abstract
Voltage-gated potassium channels play a key role in human physiology and pathology. Reflecting their importance, numerous channelopathies have been characterised that arise from mutations in these channels or from autoimmune attack on the channels. Voltage-gated potassium channels are also the target of a broad range of peptide toxins from venomous organisms, including sea anemones, scorpions, spiders, snakes and cone snails; many of these peptides bind to the channels with high potency and selectivity. In this review we describe the various classes of peptide toxins that block these channels and illustrate the broad range of three-dimensional structures that support channel blockade. The therapeutic opportunities afforded by these peptides are also highlighted. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
|
17
|
Tsai FC, Lin YC, Chang SH, Chang GJ, Hsu YJ, Lin YM, Lee YS, Wang CL, Yeh YH. Differential left-to-right atria gene expression ratio in human sinus rhythm and atrial fibrillation: Implications for arrhythmogenesis and thrombogenesis. Int J Cardiol 2016; 222:104-112. [PMID: 27494721 DOI: 10.1016/j.ijcard.2016.07.103] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) causes atrial remodeling, and the left atrium (LA) is the favored substrate for maintaining AF. It remains unclear if AF remodels both atria differently and contributes to LA arrhythmogenesis and thrombogenesis. Therefore, we wished to characterize the transcript profiles in the LA and right atrium (RA) in sinus rhythm (SR) and AF respectively. METHODS Paired LA and RA appendages acquired from patients receiving cardiac surgery were used for ion-channel- and whole-exome-based transcriptome analysis. The ultrastructure was evaluated by immunohistochemistry. RESULTS Twenty-two and twenty ion-channels and transporters were differentially expressed between the LA and RA in AF and SR, respectively. Among these, 15 genes were differentially expressed in parallel between AF and SR. AF was associated with increased LA/RA expression ratio in 9 ion channel-related genes, including genes related to calcium handling. In microarray, AF was associated with a differential LA/RA gene expression ratio in 309 genes, and was involved in atherosclerosis-related signaling. AF was associated with the upregulation of thrombogenesis-related genes in the LA appendage, including P2Y12, CD 36 and ApoE. Immunohistochemistry showed higher expressions of collagen-1, oxidative stress and TGF-β1 in the RA compared to the LA. CONCLUSIONS AF was associated with differential LA-to-RA gene expression related to specific ion channels and pathways as well as upregulation of thrombogenesis-related genes in the LA appendage. Targeting the molecular mechanisms underlying the LA-to-RA difference and AF-related remodeling in the LA appendage may help provide new therapeutic options in treating AF and preventing thromboembolism in AF.
Collapse
Affiliation(s)
- Feng-Chun Tsai
- Division of Cardiac Surgery, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yen-Chen Lin
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang-Gung University College of Medicine, Chang-Gung University, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Min Lin
- School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Yun-Shien Lee
- Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan
| | - Chun-Li Wang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
18
|
Wright JR, Amisten S, Goodall AH, Mahaut-Smith MP. Transcriptomic analysis of the ion channelome of human platelets and megakaryocytic cell lines. Thromb Haemost 2016; 116:272-84. [PMID: 27277069 PMCID: PMC5080539 DOI: 10.1160/th15-11-0891] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/30/2016] [Indexed: 11/05/2022]
Abstract
Ion channels have crucial roles in all cell types and represent important therapeutic targets. Approximately 20 ion channels have been reported in human platelets; however, no systematic study has been undertaken to define the platelet channelome. These membrane proteins need only be expressed at low copy number to influence function and may not be detected using proteomic or transcriptomic microarray approaches. In our recent work, quantitative real-time PCR (qPCR) provided key evidence that Kv1.3 is responsible for the voltage-dependent K+ conductance of platelets and megakaryocytes. The present study has expanded this approach to assess relative expression of 402 ion channels and channel regulatory genes in human platelets and three megakaryoblastic/erythroleukaemic cell lines. mRNA levels in platelets are low compared to other blood cells, therefore an improved method of isolating platelets was developed. This used a cocktail of inhibitors to prevent formation of leukocyte-platelet aggregates, and a combination of positive and negative immunomagnetic cell separation, followed by rapid extraction of mRNA. Expression of 34 channel-related transcripts was quantified in platelets, including 24 with unknown roles in platelet function, but that were detected at levels comparable to ion channels with established roles in haemostasis or thrombosis. Trace expression of a further 50 ion channel genes was also detected. More extensive channelomes were detected in MEG-01, CHRF-288-11 and HEL cells (195, 185 and 197 transcripts, respectively), but lacked several channels observed in the platelet. These "channelome" datasets provide an important resource for further studies of ion channel function in the platelet and megakaryocyte.
Collapse
Affiliation(s)
| | | | | | - Martyn P Mahaut-Smith
- Prof. Martyn Mahaut-Smith, PhD, Department of Molecular and Cell Biology, Henry Wellcome Building, University of Leicester, Leicester, LEI 7RH, UK, Tel.: +44 116 229 7135, E-mail:
| |
Collapse
|
19
|
Pérez-Verdaguer M, Capera J, Serrano-Novillo C, Estadella I, Sastre D, Felipe A. The voltage-gated potassium channel Kv1.3 is a promising multitherapeutic target against human pathologies. Expert Opin Ther Targets 2015; 20:577-91. [DOI: 10.1517/14728222.2016.1112792] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Burnstock G. Blood cells: an historical account of the roles of purinergic signalling. Purinergic Signal 2015; 11:411-34. [PMID: 26260710 PMCID: PMC4648797 DOI: 10.1007/s11302-015-9462-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 07/23/2015] [Indexed: 12/17/2022] Open
Abstract
The involvement of purinergic signalling in the physiology of erythrocytes, platelets and leukocytes was recognised early. The release of ATP and the expression of purinoceptors and ectonucleotidases on erythrocytes in health and disease are reviewed. The release of ATP and ADP from platelets and the expression and roles of P1, P2Y(1), P2Y(12) and P2X1 receptors on platelets are described. P2Y(1) and P2X(1) receptors mediate changes in platelet shape, while P2Y(12) receptors mediate platelet aggregation. The changes in the role of purinergic signalling in a variety of disease conditions are considered. The successful use of P2Y(12) receptor antagonists, such as clopidogrel and ticagrelor, for the treatment of thrombosis, myocardial infarction and stroke is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK.
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
21
|
Kazama I, Ejima Y, Endo Y, Toyama H, Matsubara M, Baba A, Tachi M. Chlorpromazine-induced changes in membrane micro-architecture inhibit thrombopoiesis in rat megakaryocytes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2805-12. [DOI: 10.1016/j.bbamem.2015.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/16/2015] [Accepted: 08/18/2015] [Indexed: 01/10/2023]
|
22
|
Osman A, Hitzler WE, Ameur A, Provost P. Differential Expression Analysis by RNA-Seq Reveals Perturbations in the Platelet mRNA Transcriptome Triggered by Pathogen Reduction Systems. PLoS One 2015; 10:e0133070. [PMID: 26172280 PMCID: PMC4501785 DOI: 10.1371/journal.pone.0133070] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/23/2015] [Indexed: 11/18/2022] Open
Abstract
Platelet concentrates (PCs) are prepared at blood banks for transfusion to patients in certain clinical conditions associated with a low platelet count. To prevent transfusion-transmitted infections via PCs, different pathogen reduction (PR) systems have been developed that inactivate the nucleic acids of contaminating pathogens by chemical cross-linking, a mechanism that may also affect platelets' nucleic acids. We previously reported that treatment of stored platelets with the PR system Intercept significantly reduced the level of half of the microRNAs that were monitored, induced platelet activation and compromised the platelet response to physiological agonists. Using genome-wide differential expression (DE) RNA sequencing (RNA-Seq), we now report that Intercept markedly perturbs the mRNA transcriptome of human platelets and alters the expression level of >800 mRNAs (P<0.05) compared to other PR systems and control platelets. Of these, 400 genes were deregulated with DE corresponding to fold changes (FC) ≥ 2. At the p-value < 0.001, as many as 147 genes were deregulated by ≥ 2-fold in Intercept-treated platelets, compared to none in the other groups. Finally, integrated analysis combining expression data for microRNA (miRNA) and mRNA, and involving prediction of miRNA-mRNA interactions, disclosed several positive and inverse correlations between miRNAs and mRNAs in stored platelets. In conclusion, this study demonstrates that Intercept markedly deregulates the platelet mRNA transcriptome, concomitant with reduced levels of mRNA-regulatory miRNAs. These findings should enlighten authorities worldwide when considering the implementation of PR systems, that target nucleic acids and are not specific to pathogens, for the management of blood products.
Collapse
Affiliation(s)
- Abdimajid Osman
- Department of Clinical Chemistry, Region Östergötland, Ingång 64, Linköping, Sweden
- Department of Clinical and Experimental Medicine, University of Linköping, Linköping, Sweden
| | - Walter E. Hitzler
- Transfusion Center, University Medical Center of the Johannes Gutenberg University Mainz, Hochhaus Augustusplatz, Mainz, Germany
| | - Adam Ameur
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala, Uppsala University, Uppsala, Sweden
| | - Patrick Provost
- Université Laval CHUQ Research Center / CHUL 2705 Blvd Laurier, Quebec, QC, Canada
| |
Collapse
|
23
|
Leanza L, Venturini E, Kadow S, Carpinteiro A, Gulbins E, Becker KA. Targeting a mitochondrial potassium channel to fight cancer. Cell Calcium 2015; 58:131-8. [DOI: 10.1016/j.ceca.2014.09.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/10/2014] [Accepted: 09/11/2014] [Indexed: 12/11/2022]
|
24
|
Wu KC, Kuo CS, Chao CC, Huang CC, Tu YK, Chan P, Leung YM. Role of voltage-gated K(+) channels in regulating Ca(2+) entry in rat cortical astrocytes. J Physiol Sci 2015; 65:171-7. [PMID: 25617267 PMCID: PMC10717881 DOI: 10.1007/s12576-015-0356-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/09/2015] [Indexed: 01/11/2023]
Abstract
Astrocytes have multiple functions such as provision of nourishment and mechanical support to the nervous system, helping to clear extracellular metabolites of neurons and modulating synaptic transmission by releasing gliotransmitters. In excitable cells, voltage-gated K(+) (Kv) channels serve to repolarize during action potentials. Astrocytes are considered non-excitable cells since they are not able to generate action potentials. There is an abundant expression of various Kv channels in astrocytes but the functions of these Kv channels remain unclear. We examined whether these astrocyte Kv channels regulate astrocyte "excitability" in the form of cytosolic Ca(2+) signaling. Electrophysiological examination revealed that neonatal rat cortical astrocytes possessed both delayed rectifier type and A-type Kv channels. Pharmacological blockade of both delayed rectifier Kv channels by TEA and A-type Kv channels by quinidine significantly suppressed store-operated Ca(2+) influx; however, TEA alone or quinidine alone did not suffice to cause such suppression. TEA and quinidine together dramatically enhanced current injection-triggered membrane potential overshoot (depolarization); either drug alone caused much smaller enhancements. Taken together, the results suggest both delayed rectifier and A-type Kv channels regulate astrocyte Ca(2+) signaling via controlling membrane potential.
Collapse
Affiliation(s)
- King-Chuen Wu
- Department of Anesthesiology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chang-Shin Kuo
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung, 40402 Taiwan
| | - Chia-Chia Chao
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chieh-Chen Huang
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yuan-Kun Tu
- Orthopedic Department, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Paul Chan
- Division of Cardiology, Department of Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yuk-Man Leung
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung, 40402 Taiwan
| |
Collapse
|
25
|
Yang W, Feng J, Xiang F, Xie Z, Zhang G, Sabatier JM, Cao Z, Li W, Chen Z, Wu Y. Endogenous animal toxin-like human β-defensin 2 inhibits own K(+) channels through interaction with channel extracellular pore region. Cell Mol Life Sci 2015; 72:845-53. [PMID: 25238780 PMCID: PMC11113244 DOI: 10.1007/s00018-014-1715-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 11/25/2022]
Abstract
Human potassium channels are widely inhibited by peptide toxins from venomous animals. However, no human endogenous peptide inhibitor has been discovered so far. In this study, we demonstrate for the first time using electrophysiological techniques, that endogenous human β-defensin 2 (hBD2) is able to selectively and dose-dependently inhibit the human voltage-gated Kv1.3 channel at picomolar peptide concentration. The co-immunoprecipitation assays further supported the selective binding of hBD2 to Kv1.3 channel. Using mutagenesis experiments, we found that the outer pore domain of Kv1.3 channel was the binding site of hBD2, which is similar to the interacting site of Kv1.3 channel recognized by animal toxin inhibitors. The hBD2 was able to suppress IL-2 production through inhibition of Kv1.3 channel currents in human Jurkat cells, which was further confirmed by the lack of hBD2 activity on IL-2 production after Kv1.3 knockdown in these cells. More interestingly, hBD2 was also found to efficiently inhibit Kv1.3 channel currents and suppress IL-2 production in both human primary CD3(+) T cells and peripheral mononuclear cells from either healthy donors or psoriasis patients. Our findings not only evidenced hBD2 as the first characterized endogenous peptide inhibitor of human potassium channels, but also paved a promising avenue to investigate newly discovered function of hBD2 as Kv1.3 channel inhibitor in the immune system and other fields.
Collapse
Affiliation(s)
- Weishan Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Jing Feng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Fang Xiang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Zili Xie
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Guoyi Zhang
- Institute of Dermatology, Chinese Academy of Medicine Sciences, Nanjing, 210042 China
| | - Jean-Marc Sabatier
- Laboratoire INSERM UMR 1097, Université d’Aix-Marseille, 163, Avenue de Luminy, Case 939, TPR2 INSERM, 13288 Marseille Cedex 09, France
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
- Center for BioDrug Research, Wuhan University, Wuhan, 430072 China
| | - Wenxin Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
- Center for BioDrug Research, Wuhan University, Wuhan, 430072 China
| | - Zongyun Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
- Center for BioDrug Research, Wuhan University, Wuhan, 430072 China
| |
Collapse
|
26
|
Lang F, Gawaz M, Borst O. The serum- & glucocorticoid-inducible kinase in the regulation of platelet function. Acta Physiol (Oxf) 2015; 213:181-90. [PMID: 24947805 DOI: 10.1111/apha.12334] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/02/2014] [Accepted: 06/15/2014] [Indexed: 12/23/2022]
Abstract
The serum- and glucocorticoid-inducible kinase 1 (SGK1) is expressed in megakaryocytes and circulating platelets. In megakaryocytes, SGK1 activates transcription factor nuclear factor kappa-B (NF-κB), which in turn stimulates expression of Orai1, a Ca(2+) channel protein accomplishing store-operated Ca(2+) enrty (SOCE). SGK1 enhances SOCE and several Ca(2+) -sensitive platelet functions, including degranulation, integrin αII b β3 activation, phosphatidylserine exposure, aggregation and thrombus formation. As shown in other cell types, stimulators of SGK1 expression include ischaemia, oxidative stress, hyperglycaemia, advanced glycation end products (AGEs) and a variety of hormones such as glucocorticoids, mineralocorticoids, transforming growth factor beta (TGFβ), interleukin 6 (IL-6), platelet-derived growth factor (PDGF), thrombin and endothelin. Thus, SGK1-sensitive Ca(2+) signalling may contribute to altered platelet function in several clinical conditions including inflammation, metabolic syndrome, diabetes mellitus and chronic renal failure. Nevertheless, further studies are needed defining the contribution of altered SGK1 expression and activity to physiology and pathophysiology of platelets.
Collapse
Affiliation(s)
- F. Lang
- Department of Physiology; University of Tübingen; Tübingen Germany
| | - M. Gawaz
- Department of Cardiology & Cardiovascular Medicine; University of Tübingen; Tübingen Germany
| | - O. Borst
- Department of Physiology; University of Tübingen; Tübingen Germany
- Department of Cardiology & Cardiovascular Medicine; University of Tübingen; Tübingen Germany
| |
Collapse
|
27
|
Kazama I. Physiological significance of delayed rectifier K(+) channels (Kv1.3) expressed in T lymphocytes and their pathological significance in chronic kidney disease. J Physiol Sci 2015; 65:25-35. [PMID: 25096892 PMCID: PMC10717717 DOI: 10.1007/s12576-014-0331-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/14/2014] [Indexed: 11/24/2022]
Abstract
T lymphocytes predominantly express delayed rectifier K(+) channels (Kv1.3) in their plasma membranes. More than 30 years ago, patch-clamp studies revealed that the channels play crucial roles in facilitating the calcium influx necessary to trigger lymphocyte activation and proliferation. In addition to selective channel inhibitors that have been developed, we recently showed physiological evidence that drugs such as nonsteroidal anti-inflammatory drugs, antibiotics, and anti-hypertensives effectively suppress the channel currents in lymphocytes, and thus exert immunosuppressive effects. Using experimental animal models, previous studies revealed the pathological relevance between the expression of ion channels and the progression of renal diseases. As an extension, we recently demonstrated that the overexpression of lymphocyte Kv1.3 channels contributed to the progression of chronic kidney disease (CKD) by promoting cellular proliferation and interstitial fibrosis. Together with our in-vitro results, the studies indicated the therapeutic potency of Kv1.3-channel inhibitors in the treatment or the prevention of CKD.
Collapse
Affiliation(s)
- Itsuro Kazama
- Department of Physiology I, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan,
| |
Collapse
|
28
|
Abstract
The field of mitochondrial ion channels has recently seen substantial progress, including the molecular identification of some of the channels. An integrative approach using genetics, electrophysiology, pharmacology, and cell biology to clarify the roles of these channels has thus become possible. It is by now clear that many of these channels are important for energy supply by the mitochondria and have a major impact on the fate of the entire cell as well. The purpose of this review is to provide an up-to-date overview of the electrophysiological properties, molecular identity, and pathophysiological functions of the mitochondrial ion channels studied so far and to highlight possible therapeutic perspectives based on current information.
Collapse
|
29
|
Cerecedo D, Martínez-Vieyra I, Alonso-Rangel L, Benítez-Cardoza C, Ortega A. Epithelial sodium channel modulates platelet collagen activation. Eur J Cell Biol 2014; 93:127-36. [PMID: 24679405 DOI: 10.1016/j.ejcb.2014.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 01/30/2014] [Accepted: 02/24/2014] [Indexed: 12/14/2022] Open
Abstract
Activated platelets adhere to the exposed subendothelial extracellular matrix and undergo a rapid cytoskeletal rearrangement resulting in shape change and release of their intracellular dense and alpha granule contents to avoid hemorrhage. A central step in this process is the elevation of the intracellular Ca(2+) concentration through its release from intracellular stores and on throughout its influx from the extracellular space. The Epithelial sodium channel (ENaC) is a highly selective Na(+) channel involved in mechanosensation, nociception, fluid volume homeostasis, and control of arterial blood pressure. The present study describes the expression, distribution, and participation of ENaC in platelet migration and granule secretion using pharmacological inhibition with amiloride. Our biochemical and confocal analysis in suspended and adhered platelets suggests that ENaC is associated with Intermediate filaments (IF) and with Dystrophin-associated proteins (DAP) via α-syntrophin and β-dystroglycan. Migration assays, quantification of soluble P-selectin, and serotonin release suggest that ENaC is dispensable for migration and alpha and dense granule secretion, whereas Na(+) influx through this channel is fundamental for platelet collagen activation.
Collapse
Affiliation(s)
- Doris Cerecedo
- Laboratorio de Hematobiología, Escuela Nacional de Medicina y Homeopatía (ENMH), Instituto Politécnico Nacional (IPN), Mexico City, Mexico.
| | - Ivette Martínez-Vieyra
- Laboratorio de Hematobiología, Escuela Nacional de Medicina y Homeopatía (ENMH), Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Lea Alonso-Rangel
- Laboratorio de Hematobiología, Escuela Nacional de Medicina y Homeopatía (ENMH), Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Claudia Benítez-Cardoza
- Laboratorio de Bioquímica, Escuela Nacional de Medicina y Homeopatía (ENMH), Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Arturo Ortega
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav-IPN), Mexico City, Mexico
| |
Collapse
|
30
|
Kazama I, Maruyama Y, Baba A. Amphipath-induced plasma membrane curvature controls microparticle formation from adipocytes: novel therapeutic implications for metabolic disorders. Med Hypotheses 2013; 82:196-8. [PMID: 24368036 DOI: 10.1016/j.mehy.2013.11.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 11/25/2013] [Accepted: 11/30/2013] [Indexed: 12/18/2022]
Abstract
Microparticles produced from the membrane surface of adipocytes promote lipid biosynthesis and angiogenesis in adipose tissues. Thus, they are deeply associated with the onset of metabolic disorders. Despite our understanding of their roles in physiological or pathological responses, we know little about the mechanism by which microparticles are produced from adipocytes. Based on our previous studies using rat megakaryocytes or mast cells during exocytosis, we proposed that membrane curvature induced by amphiphilic reagents, such as chlorpromazine or salicylate, facilitate or inhibit the formation of microparticles. Since the plasma membranes in adipocytes share many common biophysiological features with those in megakaryocytes or mast cells during exocytosis, the same stimulatory or inhibitory mechanism of microparticle formation would exist in adipocytes. Therefore, we hypothesize here that amphiphilic reagents would also change the membrane curvature in adipocytes, and that such changes would facilitate or inhibit the microparticle formation from adipocytes. Our hypothesis is unique because it sheds light for the first time on the physiological mechanism by which microparticles are produced in adipocytes. It is also important because the idea could have novel therapeutic implications for metabolic disorders that are triggered by increases in the microparticle formation.
Collapse
Affiliation(s)
- Itsuro Kazama
- Department of Physiology I, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan.
| | - Yoshio Maruyama
- Department of Physiology I, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Asuka Baba
- Department of Physiology I, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
31
|
Duque A, Gazula VR, Kaczmarek LK. Expression of Kv1.3 potassium channels regulates density of cortical interneurons. Dev Neurobiol 2013; 73:841-55. [PMID: 23821603 DOI: 10.1002/dneu.22105] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 06/19/2013] [Accepted: 06/21/2013] [Indexed: 01/27/2023]
Abstract
The Kv1.3 protein is a member of the large family of voltage-dependent K+ subunits (Kv channels), which assemble to form tetrameric membrane-spanning channels that provide a selective pore for the conductance of K+ across the cell membrane. Kv1.3 differs from most other Kv channels in that deletion of Kv1.3 gene produces very striking changes in development and structure of the olfactory bulb, where Kv1.3 is expressed at high levels, resulting in a lower threshold for detection of odors, an increased number of synaptic glomeruli and alterations in the levels of a variety of neuronal signaling molecules. Because Kv1.3 is also expressed in the cerebral cortex, we have now examined the effects of deletion of the Kv1.3 gene on the expression of interneuron populations of the cerebral cortex. Using unbiased stereology we found an increase in the number of parvalbumin (PV) cells in whole cerebral cortex of Kv1.3-/- mice relative to that in wild-type mice, and a decrease in the number of calbindin (CB), calretinin (CR), neuropeptide Y (NPY), vasoactive intestinal peptide (VIP), and somatostatin (SOM) interneurons. These changes are accompanied by a decrease in the cortical volume such that the cell density of PV interneurons is significantly increased and that of SOM neurons is decreased in Kv1.3-/- animals. Our studies suggest that, as in the olfactory bulb, Kv1.3 plays a unique role in neuronal differentiation and/or survival of interneuron populations and that expression of Kv1.3 is required for normal cortical function.
Collapse
Affiliation(s)
- Alvaro Duque
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, 06520
| | | | | |
Collapse
|
32
|
Olivera A, Kitamura Y, Wright LD, Allende ML, Chen W, Kaneko-Goto T, Yoshihara Y, Proia RL, Rivera J. Sphingosine-1-phosphate can promote mast cell hyper-reactivity through regulation of contactin-4 expression. J Leukoc Biol 2013; 94:1013-24. [PMID: 23904439 DOI: 10.1189/jlb.0313163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Both genes and the environment are determinants in the susceptibility to allergies and may alter the severity of the disease. We explored whether an increase in the levels of the lipid mediator S1P in vivo, a condition found during allergic asthma, could affect the sensitivity or the response of MCs to IgE/Ag and the onset of allergic disease. We found that increasing S1P levels by genetic deletion of S1P lyase, the enzyme catabolizing S1P, led to elevated activity of circulating tryptase. Accordingly, MCs of S1P lyase-deficient mice were mostly degranulated in the tissues and showed enhanced calcium levels, degranulation, and cytokine production in response to IgE/Ag in vitro. Th 1-skewed mice (C57BL/6) had lower levels of S1P in circulation and histamine responses than did Th 2-skewed (129/Sv) mice. However, when S1P levels were increased by pharmacologic inhibition of S1P lyase, the C57BL/6 mice showed increased histamine release into the circulation and anaphylactic responses similar to those in the 129/Sv mice. Culturing of MCs in the presence of S1P enhanced their degranulation responses, and when the S1P-treated MCs were used to reconstitute MC-deficient (Kit(W-sh)) mice, they caused enhanced anaphylaxis. Gene expression arrays in S1P lyase-deficient MCs and MCs treated with S1P continuously revealed increased expression of numerous genes, including the adhesion molecule CNTN4,which contributed to the enhanced responses. Our findings argue that dysregulation in the metabolism of S1P is a contributing factor in modulating MC responsiveness and the allergic response.
Collapse
Affiliation(s)
- Ana Olivera
- 1.Laboratory of Molecular Immunogenetics, NIAMS, NIH, Building 10, Room 13C103, Bethesda, MD 20892. ; J.R., E-mail:
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kazama I, Maruyama Y, Nakamichi S. Aspirin-induced microscopic surface changes stimulate thrombopoiesis in rat megakaryocytes. Clin Appl Thromb Hemost 2012; 20:318-25. [PMID: 23076773 DOI: 10.1177/1076029612461845] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
During the process of thrombopoiesis, invaginations of the plasma membrane occur in megakaryocytes. Since acetylsalicylic acid (aspirin), the most commonly used anti-inflammatory and antiplatelet drug, interacts with the lipid bilayers of the plasma membranes, this drug would affect the process of thrombopoiesis. In the present study, employing a standard patch-clamp whole-cell recording technique, we examined the effects of aspirin on delayed rectifier K(+)-channel (Kv1.3) currents and the membrane capacitance in megakaryocytes. Using confocal imaging of di-8-butyl-amino-naphthyl-ethylene-pyridinium-propyl-sulfonate (di-8-ANEPPS) staining, we also monitored the membrane invaginations in megakaryocytes. Aspirin suppressed both the peak and the pulse-end currents with a significant increase in the membrane capacitance. Massive di-8-ANEPPS staining after treatment with aspirin demonstrated the impaired membrane micro-architecture of megakaryocytes. This study demonstrated for the first time that aspirin induces microscopic surface changes in megakaryocytes. Such surface changes were thought to stimulate thrombopoiesis in megakaryocytes as detected by the increase in the membrane invaginations.
Collapse
Affiliation(s)
- Itsuro Kazama
- 1Department of Physiology I, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | | | | |
Collapse
|
34
|
Abstract
Ion channels are transmembrane proteins that play ubiquitous roles in cellular homeostasis and activation. In addition to their recognized role in the regulation of ionic permeability and thus membrane potential, some channel proteins possess intrinsic kinase activity, directly interact with integrins or are permeable to molecules up to ≈1000 Da. The small size and anuclear nature of the platelet has often hindered progress in understanding the role of specific ion channels in hemostasis, thrombosis and other platelet-dependent events. However, with the aid of transgenic mice and 'surrogate' patch clamp recordings from primary megakaryocytes, important unique contributions to platelet function have been identified for several classes of ion channel. Examples include ATP-gated P2X1 channels, Orai1 store-operated Ca2+ channels, voltage-gated Kv1.3 channels, AMPA and kainate glutamate receptors and connexin gap junction channels. Furthermore, evidence exists that some ion channels, such as NMDA glutamate receptors, contribute to megakaryocyte development. This review examines the evidence for expression of a range of ion channels in the platelet and its progenitor cell, and highlights the distinct roles that these proteins may play in health and disease.
Collapse
Affiliation(s)
- M P Mahaut-Smith
- Department of Cell Physiology & Pharmacology, University of Leicester, Leicester, UK.
| |
Collapse
|
35
|
Leanza L, Henry B, Sassi N, Zoratti M, Chandy KG, Gulbins E, Szabò I. Inhibitors of mitochondrial Kv1.3 channels induce Bax/Bak-independent death of cancer cells. EMBO Mol Med 2012; 4:577-93. [PMID: 22496117 PMCID: PMC3407946 DOI: 10.1002/emmm.201200235] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 02/17/2012] [Accepted: 02/21/2012] [Indexed: 02/01/2023] Open
Abstract
Overcoming the resistance of tumours to chemotherapy, often due to downregulation of Bax and Bak, represents a significant clinical challenge. It is therefore important to identify novel apoptosis inducers that bypass Bax and Bak. Potassium channels are emerging as oncological targets and a crucial role of mitochondrial Kv1.3 in apoptosis has been demonstrated. Here we report for the first time that Psora-4, PAP-1 and clofazimine, three distinct membrane-permeant inhibitors of Kv1.3, induce death by directly targeting the mitochondrial channel in multiple human and mouse cancer cell lines. Importantly, these drugs activated the intrinsic apoptotic pathway also in the absence of Bax and Bak, a result in agreement with the current mechanistic model for mitochondrial Kv1.3 action. Genetic deficiency or short interfering RNA (siRNA)-mediated downregulation of Kv1.3 abrogated the effects of the drugs. Intraperitoneal injection of clofazimine reduced tumour size by 90% in an orthotopic melanoma B16F10 mouse model in vivo, while no adverse effects were observed in several healthy tissues. The study indicates that inhibition of mitochondrial Kv1.3 might be a novel therapeutic option for the induction of cancer cell death independent of Bax and Bak.
Collapse
Affiliation(s)
- Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
36
|
Zeng XC, Zhang L, Nie Y, Luo X. Identification and molecular characterization of three new K+-channel specific toxins from the Chinese scorpion Mesobuthus martensii Karsch revealing intronic number polymorphism and alternative splicing in duplicated genes. Peptides 2012; 34:311-23. [PMID: 22230549 DOI: 10.1016/j.peptides.2011.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 12/21/2011] [Accepted: 12/21/2011] [Indexed: 11/23/2022]
Abstract
K(+)-channel specific toxins from scorpions are powerful probes used in the structural and functional characterization of different subfamilies of K(+)-channels which are thought to be the most diverse ion channels. However, only a limited number of K(+)-channel toxins have been identified from scorpions so far; moreover, little is known about the mechanisms for the generation of a combinatorial peptide library in a venom gland of a scorpion. Here, we identified and characterized three new K(+)-channel toxin-like peptides from the scorpion Mesobuthus martensii Karsch, which were referred to as BmKcug1, BmKcug2 and BmKcugx, respectively. BmKcug1 and BmKcug2 are two new members of α-KTx1 subfamily, and have been classified as α-KTx1.14 and α-KTx1.15, respectively. BmKcugx represents a new subfamily of K(+)-channel specific toxins which was classified into α-KTx22. BmKcugx was thus classified as α-KTx22.1. Genomic analysis demonstrated that BmKcugx gene has two exons interrupted by an intron inserted in the signal peptide encoding region, whereas BmKcug1a (a close homologue of BmKcug1)/BmKcug2 gene was interrupted by two introns, located within the 5'UTR of the gene and in the signal peptide encoding region, respectively. Transcriptomic analysis for the venom glands of M. martensii Karsch indicated that the abundances of the transcripts of BmKcug1a and BmKcug2 are much higher than that of BmKcugx; it suggests that the intron in 5'UTR could markedly increase the expression level of the K(+)-channel toxins. Alignment of the genomic sequences of BmKcug1a and BmKcug2 revealed that an alternative splicing event occurred at the intron 1-exon 2 junction in the 5'UTR of BmKcug2 transcript.
Collapse
Affiliation(s)
- Xian-Chun Zeng
- Department of Biological Science and Technology, School of Environmental Studies & State Key Laboratory of Biogeology and Environmental Geology, China University of Geosciences (Wuhan), Wuhan 430074, China.
| | | | | | | |
Collapse
|
37
|
Kazama I, Maruyama Y, Murata Y. Suppressive effects of nonsteroidal anti-inflammatory drugs diclofenac sodium, salicylate and indomethacin on delayed rectifier K+-channel currents in murine thymocytes. Immunopharmacol Immunotoxicol 2012; 34:874-8. [PMID: 22409730 DOI: 10.3109/08923973.2012.666249] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lymphocytes predominantly express delayed rectifier K(+)-channels (Kv1.3) in their plasma membranes, and the channels play crucial roles in the lymphocyte activation and proliferation. Since nonsteroidal anti-inflammatory drugs (NSAIDs), the most commonly used analgesic and antipyretic drugs, exert immunomodulatory effects, they would affect the channel currents in lymphocytes. In the present study, employing the standard patch-clamp whole-cell recording technique, we examined the effects of diclofenac sodium, salicylate and indomethacin on the channel currents in murine thymocytes and the membrane capacitance. Diclofenac sodium and salicylate significantly suppressed the pulse-end currents of the channel. However, indomethacin suppressed both the peak and the pulse-end currents with a significant increase in the membrane capacitance. This study demonstrated for the first time that NSAIDs, such as diclofenac sodium, salicylate and indomethacin, exert inhibitory effects on thymocyte Kv1.3-channel currents. The slow inactivation pattern induced by indomethacin was thought to be associated with microscopic changes in the plasma membrane surface detected by the increase in the membrane capacitance.
Collapse
Affiliation(s)
- Itsuro Kazama
- Department of Physiology I, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan.
| | | | | |
Collapse
|
38
|
Ramanathan G, Gupta S, Thielmann I, Pleines I, Varga-Szabo D, May F, Mannhalter C, Dietrich A, Nieswandt B, Braun A. Defective diacylglycerol-induced Ca2+ entry but normal agonist-induced activation responses in TRPC6-deficient mouse platelets. J Thromb Haemost 2012; 10:419-29. [PMID: 22176814 DOI: 10.1111/j.1538-7836.2011.04596.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Platelet adhesion, activation and aggregation at sites of vascular injury are essential processes for primary hemostasis. Elevation of the intracellular Ca(2+) concentration is a central event in platelet activation but the underlying mechanisms are not fully understood. Store-operated calcium entry (SOCE) through Orai1 was shown to be the main Ca(2+) influx pathway in murine platelets, but there are additional non-store-operated Ca(2+) (non-SOC) and receptor operated Ca(2+) (ROC) channels expressed in the platelet plasma membrane. OBJECTIVE Canonical transient receptor potential (TRPC) channel 6 is found both in human and murine platelets and has been proposed to mediate diacylglycerol (DAG) activated ROCE but also a role in the regulation of SOCE has been suggested. METHODS To investigate the function of TRPC6 in platelet Ca(2+) signaling and activation, we analyzed platelets from mice deficient in TRPC6 using a wide range of in vitro and in vivo assays. RESULTS In the mutant platelets, DAG activated Ca(2+) influx was found to be abolished. However, this did not significantly affect SOCE or agonist induced Ca(2+) responses. Platelet function in vitro and in vivo was also unaltered in the absence of TRPC6. CONCLUSION Our results indicate that DAG activated ROCE is mediated exclusively by TRPC6 in murine platelets, but this Ca(2+) influx has no major functional relevance for hemostasis and thrombosis. Further, in contrast to previous suggestions, based on studies with human platelets, TRPC6 appears to play an insignificant role in the regulation of SOCE in murine platelets.
Collapse
Affiliation(s)
- G Ramanathan
- Chair of Vascular Medicine, DFG Research Center for Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Amisten S. A rapid and efficient platelet purification protocol for platelet gene expression studies. Methods Mol Biol 2012; 788:155-172. [PMID: 22130707 DOI: 10.1007/978-1-61779-307-3_12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Isolation of pure platelet samples from whole blood is crucial for the study of platelet gene expression. The main obstacles to overcome in order to successfully isolate platelets from whole blood include (1) platelet activation; (2) leukocyte and red blood cell contamination, and (3) time-dependent platelet mRNA degradation. This chapter describes a rapid and highly efficient method for isolating human circulating platelets from small volumes of whole blood based on efficient inhibition of platelet activation and leukocyte removal by filtration followed by magnetic bead-depletion of residual contaminating leukocytes and red blood cells. Also described are methods for RNA extraction, cDNA synthesis, and platelet gene expression studies using both quantitative real-time PCR and microarray.
Collapse
Affiliation(s)
- Stefan Amisten
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Oxford University, The Churchill Hospital, Oxford, UK.
| |
Collapse
|
40
|
Zhu Z, Zhou H, Yu X, Chen L, Zhang H, Ren S, Wu Y, Luo D. Potential regulatory role of calsequestrin in platelet Ca(2+) homeostasis and its association with platelet hyperactivity in diabetes mellitus. J Thromb Haemost 2012; 10:116-24. [PMID: 22060633 DOI: 10.1111/j.1538-7836.2011.04550.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Altered Ca(2+) homeostasis contributes significantly to platelet hyperactivity in diabetes mellitus. Calsequestrin (CSQ), as a Ca(2+) buffer protein in the sarcoplasmic reticulum, also regulates the Ca(2+) release process in muscles. We hypothesized that CSQ may be expressed in platelets, but is altered and involved in diabetic platelet Ca(2+) abnormalities and hyperaggregability. METHODS CSQ expression in platelets from streptozotocin-induced type 1 diabetes rats, type 2 diabetes volunteers and Goto-Kakizaki rats were analyzed by western blotting and RT-qPCR. Platelet Ca(2+) and aggregation were evaluated with Fura2 and an aggregometer, respectively. RESULTS Platelets from diabetic patients and rats exhibited increased resting Ca(2+) levels, and hyperactive Ca(2+) and aggregation responses to agonists. This enhanced basal Ca(2+) was largely dependent on intracellular Ca(2+) and insensitive to inositol 1,4,5-trisphosphate receptor (IP(3)R) antagonism. Additionally, the expression of the skeletal CSQ isotype (CSQ-1) was detected in both rat and human platelets, but its levels were significantly lowered in diabetic platelets as compared with normal platelets. Impairment of CSQ by trifluoperazine caused concentration-dependent Ca(2+) release in normal platelets and HEK293 cells. Knocking down CSQ-1 in HEK293 cells resulted in increased leakage of Ca(2+), which was also insensitive to IP(3)R inhibition, and exaggerated Ca(2+) release following carbachol treatment. CONCLUSIONS Downregulation of CSQ-1 in diabetic platelets and impairment of CSQ-1 in normal cells leads to disturbed Ca(2+) release, demonstrating a potential role for CSQ-1 in the regulation of the platelet Ca(2+) release process and a possible causal contribution to diabetic platelet hyperactivity.
Collapse
Affiliation(s)
- Z Zhu
- Department of Pharmacology, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Fortin DL, Dunn TW, Fedorchak A, Allen D, Montpetit R, Banghart MR, Trauner D, Adelman JP, Kramer RH. Optogenetic photochemical control of designer K+ channels in mammalian neurons. J Neurophysiol 2011; 106:488-96. [PMID: 21525363 PMCID: PMC3129715 DOI: 10.1152/jn.00251.2011] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 04/25/2011] [Indexed: 11/22/2022] Open
Abstract
Currently available optogenetic tools, including microbial light-activated ion channels and transporters, are transforming systems neuroscience by enabling precise remote control of neuronal firing, but they tell us little about the role of indigenous ion channels in controlling neuronal function. Here, we employ a chemical-genetic strategy to engineer light sensitivity into several mammalian K(+) channels that have different gating and modulation properties. These channels provide the means for photoregulating diverse electrophysiological functions. Photosensitivity is conferred on a channel by a tethered ligand photoswitch that contains a cysteine-reactive maleimide (M), a photoisomerizable azobenzene (A), and a quaternary ammonium (Q), a K(+) channel pore blocker. Using mutagenesis, we identify the optimal extracellular cysteine attachment site where MAQ conjugation results in pore blockade when the azobenzene moiety is in the trans but not cis configuration. With this strategy, we have conferred photosensitivity on channels containing Kv1.3 subunits (which control axonal action potential repolarization), Kv3.1 subunits (which contribute to rapid-firing properties of brain neurons), Kv7.2 subunits (which underlie "M-current"), and SK2 subunits (which are Ca(2+)-activated K(+) channels that contribute to synaptic responses). These light-regulated channels may be overexpressed in genetically targeted neurons or substituted for native channels with gene knockin technology to enable precise optopharmacological manipulation of channel function.
Collapse
Affiliation(s)
- Doris L Fortin
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kazama I, Endo Y, Toyama H, Ejima Y, Kurosawa S, Murata Y, Matsubara M, Maruyama Y. Compensatory thrombopoietin production from the liver and bone marrow stimulates thrombopoiesis of living rat megakaryocytes in chronic renal failure. NEPHRON EXTRA 2011; 1:147-56. [PMID: 22470388 PMCID: PMC3290854 DOI: 10.1159/000333018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND/AIMS Decreased thrombopoiesis has been ascribed a role in the pathogenesis of uremic bleeding in chronic renal failure (CRF). However, serum thrombopoietin (TPO) levels are usually elevated in CRF patients, suggesting increased thrombopoiesis. The aim of this study was to determine the thrombopoietic activity in CRF. METHODS Male Sprague-Dawley rats that underwent 5/6 nephrectomy were used as the model of CRF. Age-matched sham-operated rats were used as controls. Single megakaryocytes were isolated from the rat bone marrow, and their size distribution was examined. Megakaryocyte membrane invaginations were monitored by confocal imaging of di-8-ANEPPS staining, and patch clamp whole-cell recordings of membrane capacitance. TPO gene expression was assessed in various tissues. RESULTS Circulating platelet counts and the number of large megakaryocytes were increased in the bone marrow of CRF rats. Massive di-8-ANEPPS staining and increased membrane capacitance in large megakaryocytes demonstrated increased membrane invaginations. Unaffected Kv1.3-channel currents per cell surface area demonstrated unaltered channel densities. TPO transcription was decreased in the renal cortex but increased in the liver and bone marrow of CRF rats. CONCLUSION Increased thrombopoiesis in CRF was thought to be a reactive mechanism to platelet dysfunction. Increased TPO production from the liver and bone marrow compensated for decreased production from damaged kidneys.
Collapse
Affiliation(s)
| | - Yasuhiro Endo
- Department of Anesthesiology, Tohoku University Hospital, Sendai, Japan
| | - Hiroaki Toyama
- Department of Anesthesiology, Tohoku University Hospital, Sendai, Japan
| | - Yutaka Ejima
- Department of Anesthesiology, Tohoku University Hospital, Sendai, Japan
| | - Shin Kurosawa
- Department of Anesthesiology, Tohoku University Hospital, Sendai, Japan
| | | | - Mitsunobu Matsubara
- Division of Molecular Medicine, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | |
Collapse
|
43
|
Choi BH, Hahn SJ. Kv1.3: a potential pharmacological target for diabetes. Acta Pharmacol Sin 2010; 31:1031-5. [PMID: 20711225 DOI: 10.1038/aps.2010.133] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
K(+) channels, which are ubiquitous membrane proteins, play a central role in regulating the resting membrane potential and the shape and duration of the action potential in pancreatic beta-cells. There are at least three types of K(+) channels (K(ATP), K(Ca), and Kv2.1 channels) that are involved in glucose-stimulated insulin secretion in pancreatic beta-cells, and one type (Kv1.3) that is associated with the regulation of insulin sensitivity in peripheral target tissues. This article reviews the function of Kv1.3 channels that contribute to mediating insulin action in insulin-sensitive tissues. Pharmacological strategies for targeting Kv1.3 are then discussed with a focus on a rationale for the potential therapeutic use of Kv1.3 blocker in diabetic treatment.
Collapse
|
44
|
Affiliation(s)
- Michael Emerson
- Platelet Biology Group, National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|