1
|
Iglesias-Martínez-Almeida M, Campos-Ríos A, Freiría-Martínez L, Rivera-Baltanás T, Rodrígues-Amorím D, Diz-Chaves Y, Comis-Tuche M, Fernández-Palleiro P, Rodríguez-Jamardo C, Ramos-García S, Rodríguez-Tébar A, Del Carmen Vallejo-Curto M, Campos-Pérez JA, López-García M, de Las Heras E, García-Caballero A, Olivares JM, Lamas JA, Spuch C. Characterization and modulation of voltage-gated potassium channels in human lymphocytes in schizophrenia. Schizophr Res 2024; 270:260-272. [PMID: 38944972 DOI: 10.1016/j.schres.2024.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND It is known that the immune system is dysregulated in schizophrenia, having a state similar to chronic neuroinflammation. The origin of this process is unknown, but it is known that T and B lymphocytes, which are components of the adaptive immune system, play an important role in the pathogenic mechanisms of schizophrenia. METHODS We analysed the membrane of PBMCs from patients diagnosed with schizophrenia through proteomic analysis (n = 5 schizophrenia and n = 5 control). We found the presence of the Kv1.3 voltage-gated potassium channel and its auxiliary subunit β1 (KCNAB1) and β2 (KCNAB2). From a sample of 90 participants, we carried out a study on lymphocytes with whole-cell patch-clamp experiments (n = 7 schizophrenia and n = 5 control), western blot (n = 40 schizophrenia and n = 40 control) and confocal microscopy to evaluate the presence and function of different channels. Kv in both cells. RESULTS We demonstrated the overexpression of Kv1.1, Kv1.2, Kv1.3, Kv1.6, Kv4.2, Kv4.3 and Kv7.2 channels in PBMCs from patients with schizophrenia. This study represents a groundbreaking exploration, as it involves an electrophysiological analysis performed on T and B lymphocytes from patients diagnosed of schizophrenia compared to healthy participants. We observed that B lymphocytes exhibited an increase in output current along with greater peak current amplitude and voltage conductance curves among patients with schizophrenia compared with healthy controls. CONCLUSIONS This study showed the importance of the B lymphocyte in schizophrenia. We know that the immune system is altered in schizophrenia, but the physiological mechanisms of this system are not very well known. We suggest that the B lymphocyte may be relevant in the pathophysiology of schizophrenia and that it should be investigated in more depth, opening a new field of knowledge and possibilities for new treatments combining antipsychotics and immunomodulators. The limitation is that all participants received antipsychotic medication, which may have influenced the differences observed between patients and controls. This implies that more studies need to be done where the groups can be separated according to the antipsychotic drug.
Collapse
Affiliation(s)
- Marta Iglesias-Martínez-Almeida
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; University of Vigo, Vigo, Spain
| | - Ana Campos-Ríos
- Laboratory of neuroscience, University of Vigo, Department of Functional Biology and Health Sciences, Vigo, Spain; Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), SERGAS-UVIGO, Vigo, Spain
| | - Luis Freiría-Martínez
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; University of Vigo, Vigo, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; University of Vigo, Vigo, Spain; CIBERSAM, Madrid, Spain
| | - Daniela Rodrígues-Amorím
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States
| | | | - María Comis-Tuche
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Cynthia Rodríguez-Jamardo
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; University of Vigo, Vigo, Spain
| | - Silvia Ramos-García
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | | | | | - Jose Antonio Campos-Pérez
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Marta López-García
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Elena de Las Heras
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Alejandro García-Caballero
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Jose M Olivares
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; CIBERSAM, Madrid, Spain
| | - Jose A Lamas
- Laboratory of neuroscience, University of Vigo, Department of Functional Biology and Health Sciences, Vigo, Spain; Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), SERGAS-UVIGO, Vigo, Spain
| | - Carlos Spuch
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; CIBERSAM, Madrid, Spain.
| |
Collapse
|
2
|
Navarro-Pérez M, Capera J, Benavente-Garcia A, Cassinelli S, Colomer-Molera M, Felipe A. Kv1.3 in the spotlight for treating immune diseases. Expert Opin Ther Targets 2024; 28:67-82. [PMID: 38316438 DOI: 10.1080/14728222.2024.2315021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Kv1.3 is the main voltage-gated potassium channel of leukocytes from both the innate and adaptive immune systems. Channel function is required for common processes such as Ca2+ signaling but also for cell-specific events. In this context, alterations in Kv1.3 are associated with multiple immune disorders. Excessive channel activity correlates with numerous autoimmune diseases, while reduced currents result in increased cancer prevalence and immunodeficiencies. AREAS COVERED This review offers a general view of the role of Kv1.3 in every type of leukocyte. Moreover, diseases stemming from dysregulations of the channel are detailed, as well as current advances in their therapeutic research. EXPERT OPINION Kv1.3 arises as a potential immune target in a variety of diseases. Several lines of research focused on channel modulation have yielded positive results. However, among the great variety of specific channel blockers, only one has reached clinical trials. Future investigations should focus on developing simpler administration routes for channel inhibitors to facilitate their entrance into clinical trials. Prospective Kv1.3-based treatments will ensure powerful therapies while minimizing undesired side effects.
Collapse
Affiliation(s)
- María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Anna Benavente-Garcia
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Silvia Cassinelli
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Magalí Colomer-Molera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
3
|
Abstract
Rapid fluctuations in the plasma membrane potential (Vm) provide the basis underlying the action potential waveform in electrically excitable cells; however, a growing body of literature shows that the Vm is also functionally instructive in nonexcitable cells, including cancer cells. Various ion channels play a key role in setting and fine tuning the Vm in cancer and stromal cells within the tumor microenvironment (TME), raising the possibility that the Vm could be targeted therapeutically using ion channel-modulating compounds. Emerging evidence points to the Vm as a viable therapeutic target, given its functional significance in regulating cell cycle progression, migration, invasion, immune infiltration, and pH regulation. Several compounds are now undergoing clinical trials and there is increasing interest in therapeutic manipulation of the Vm via application of pulsed electric fields. The purpose of this article is to update the reader on the significant recent and ongoing progress to elucidate the functional significance of Vm regulation in tumors, to highlight key remaining questions and the prospect of future therapeutic targeting. In particular, we focus on key developments in understanding the functional consequences of Vm alteration on tumor development via the activation of small GTPase (K-Ras and Rac1) signaling, as well as the impact of Vm changes within the heterogeneous TME on immune cell function and cancer progression.
Collapse
Affiliation(s)
- Ming Yang
- York Biomedical Research Institute, Department of Biology, University of York, Heslington, United Kingdom
| | - William J Brackenbury
- York Biomedical Research Institute, Department of Biology, University of York, Heslington, United Kingdom
| |
Collapse
|
4
|
Fan L, Huang X, Xue W, Xie W, Tong R, Chen J, Wu T. Effects of Ruanmailing in Blocking Early Stages of Atherosclerosis by TNF- α Regulation via Kir2.1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2836880. [PMID: 35432558 PMCID: PMC9010176 DOI: 10.1155/2022/2836880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022]
Abstract
Objective Ruanmailing oral solution consists of 16 herbs, has anti-lipid peroxidation activity, protects vascular endothelial cells, and improves vascular elasticity. It is an effective drug for the treatment of atherosclerosis (AS). The objective of this study was to investigate the mechanism underlying the antiatherosclerotic effects of Ruanmailing oral solution. Methods Macrophages were isolated, cultured, and divided into the macrophage control; macrophage foam cell; and low-, medium-, and high-concentration Ruanmailing groups. Cell proliferation was analyzed by cell counting kit-8 (CCK-8) assay, and the expression levels of inward-rectifier potassium ion channel 2.1 (Kir2.1) and tumor necrosis factor (TNF)-α were detected by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot analyses. Results CCK-8 assay results showed that the tested concentrations of Ruanmailing solution did not affect macrophage proliferation. RT-PCR and Western blot assays indicated that TNF-α expression increased significantly with the formation of macrophage foam cells (P < 0.05). In addition, significant decreases in Kir2.1 and TNF-α expression were observed following treatment with various concentrations of Ruanmailing (P < 0.05). Conclusion Based on the results, Ruanmailing affects macrophage foam cell formation by regulating Kir2.1 expression, which in turn reduces TNF-α expression and exerts antiatherosclerotic effects. These findings provide a scientific basis for the use of traditional Chinese medicine for AS treatment.
Collapse
Affiliation(s)
- Liufang Fan
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, Fujian, China
| | - Xuanbin Huang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, Fujian, China
| | - Wenjuan Xue
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, Fujian, China
| | - Weyan Xie
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, Fujian, China
| | - Ruxi Tong
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, Fujian, China
| | - Jinshui Chen
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, Fujian, China
| | - Tianmin Wu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, Fujian, China
| |
Collapse
|
5
|
Vallejo-Gracia A, Sastre D, Colomer-Molera M, Solé L, Navarro-Pérez M, Capera J, Roig SR, Pedrós-Gámez O, Estadella I, Szilágyi O, Panyi G, Hajdú P, Felipe A. KCNE4-dependent functional consequences of Kv1.3-related leukocyte physiology. Sci Rep 2021; 11:14632. [PMID: 34272451 PMCID: PMC8285421 DOI: 10.1038/s41598-021-94015-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
The voltage-dependent potassium channel Kv1.3 plays essential roles in the immune system, participating in leukocyte activation, proliferation and apoptosis. The regulatory subunit KCNE4 acts as an ancillary peptide of Kv1.3, modulates K+ currents and controls channel abundance at the cell surface. KCNE4-dependent regulation of the oligomeric complex fine-tunes the physiological role of Kv1.3. Thus, KCNE4 is crucial for Ca2+-dependent Kv1.3-related leukocyte functions. To better understand the role of KCNE4 in the regulation of the immune system, we manipulated its expression in various leukocyte cell lines. Jurkat T lymphocytes exhibit low KCNE4 levels, whereas CY15 dendritic cells, a model of professional antigen-presenting cells, robustly express KCNE4. When the cellular KCNE4 abundance was increased in T cells, the interaction between KCNE4 and Kv1.3 affected important T cell physiological features, such as channel rearrangement in the immunological synapse, cell growth, apoptosis and activation, as indicated by decreased IL-2 production. Conversely, ablation of KCNE4 in dendritic cells augmented proliferation. Furthermore, the LPS-dependent activation of CY15 cells, which induced Kv1.3 but not KCNE4, increased the Kv1.3-KCNE4 ratio and increased the expression of free Kv1.3 without KCNE4 interaction. Our results demonstrate that KCNE4 is a pivotal regulator of the Kv1.3 channelosome, which fine-tunes immune system physiology by modulating Kv1.3-associated leukocyte functions.
Collapse
Affiliation(s)
- Albert Vallejo-Gracia
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain.,Virology and Immunology, Gladstone Institutes, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Daniel Sastre
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain
| | - Magalí Colomer-Molera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain
| | - Laura Solé
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain.,Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain
| | - Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain
| | - Sara R Roig
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain
| | - Oriol Pedrós-Gámez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain
| | - Irene Estadella
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain
| | - Orsolya Szilágyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 400, 1 Egyetem Sq., Debrecen, 4032, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 400, 1 Egyetem Sq., Debrecen, 4032, Hungary
| | - Péter Hajdú
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 400, 1 Egyetem Sq., Debrecen, 4032, Hungary
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028, Barcelona, Spain.
| |
Collapse
|
6
|
Minutti-Zanella C, Gil-Leyva EJ, Vergara I. Immunomodulatory properties of molecules from animal venoms. Toxicon 2021; 191:54-68. [PMID: 33417946 DOI: 10.1016/j.toxicon.2020.12.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/02/2020] [Accepted: 12/30/2020] [Indexed: 10/22/2022]
Abstract
The immune system can amplify or decrease the strength of its response when it is stimulated by chemical or biological substances that act as immunostimulators, immunosuppressants, or immunoadjuvants. Immunomodulation is a progressive approach to treat a diversity of pathologies with promising results, including autoimmune disorders and cancer. Animal venoms are a mixture of chemical compounds that include proteins, peptides, amines, salts, polypeptides, enzymes, among others, which produce the toxic effect. Since the discovery of captopril in the early 1980s, other components from snakes, spiders, scorpions, and marine animal venoms have been demonstrated to be useful for treating several human diseases. The valuable progress in fields such as venomics, molecular biology, biotechnology, immunology, and others has been crucial to understanding the interaction of toxins with the immune system and its application on immune pathologies. More in-depth knowledge of venoms' components and multi-disciplinary studies could facilitate their transformation into effective novel immunotherapies. This review addresses advances and research of molecules from venoms that have immunomodulatory properties.
Collapse
Affiliation(s)
- C Minutti-Zanella
- Departamento de Ciencias Químico-Biológicas, Universidad de Las Américas Puebla, ExHda. Sta. Catarina Mártir s/n, San Andrés Cholula, 72820, Puebla, Mexico
| | - E J Gil-Leyva
- Departamento de Ciencias Químico-Biológicas, Universidad de Las Américas Puebla, ExHda. Sta. Catarina Mártir s/n, San Andrés Cholula, 72820, Puebla, Mexico
| | - I Vergara
- Departamento de Ciencias Químico-Biológicas, Universidad de Las Américas Puebla, ExHda. Sta. Catarina Mártir s/n, San Andrés Cholula, 72820, Puebla, Mexico.
| |
Collapse
|
7
|
Zöphel D, Hof C, Lis A. Altered Ca 2+ Homeostasis in Immune Cells during Aging: Role of Ion Channels. Int J Mol Sci 2020; 22:ijms22010110. [PMID: 33374304 PMCID: PMC7794837 DOI: 10.3390/ijms22010110] [Citation(s) in RCA: 3] [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: 12/11/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
Aging is an unstoppable process and begins shortly after birth. Each cell of the organism is affected by the irreversible process, not only with equal density but also at varying ages and with different speed. Therefore, aging can also be understood as an adaptation to a continually changing cellular environment. One of these very prominent changes in age affects Ca2+ signaling. Especially immune cells highly rely on Ca2+-dependent processes and a strictly regulated Ca2+ homeostasis. The intricate patterns of impaired immune cell function may represent a deficit or compensatory mechanisms. Besides, altered immune function through Ca2+ signaling can profoundly affect the development of age-related disease. This review attempts to summarize changes in Ca2+ signaling due to channels and receptors in T cells and beyond in the context of aging.
Collapse
Affiliation(s)
| | | | - Annette Lis
- Correspondence: ; Tel.: +49-(0)-06841-1616318; Fax: +49-(0)-6841-1616302
| |
Collapse
|
8
|
Serrano-Albarrás A, Cirera-Rocosa S, Sastre D, Estadella I, Felipe A. Fighting rheumatoid arthritis: Kv1.3 as a therapeutic target. Biochem Pharmacol 2019; 165:214-220. [DOI: 10.1016/j.bcp.2019.03.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/12/2019] [Indexed: 01/18/2023]
|
9
|
Dong X, Wei L, Guo X, Yang Z, Wu C, Li P, Lu L, Qi H, Shi Y, Hu X, Wu L, Chen L, Liu W. Dlg1 Maintains Dendritic Cell Function by Securing Voltage-Gated K + Channel Integrity. THE JOURNAL OF IMMUNOLOGY 2019; 202:3187-3197. [PMID: 31028120 DOI: 10.4049/jimmunol.1900089] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) play key roles in Ab responses by presenting Ags to lymphocytes and by producing proinflammatory cytokines. In this study, we reported that DC-specific knockout of discs large homologue 1 (Dlg1) resulted in a significantly reduced capacity to mediate Ab responses to both thymus-independent and thymus-dependent Ags in Dlg1 fl/flCd11c-Cre-GFP mice. Mechanistically, Dlg1-deficient DCs showed severely impaired endocytosis and phagocytosis capacities upon Ag exposure. In parallel, loss of Dlg1 significantly jeopardized the proinflammatory cytokine production by DCs upon TLR stimulation. Thus, Dlg1-deficient DCs lost their functions to support innate and adaptive immunities. At a cellular level, Dlg1 exhibited an indispensable function to maintain membrane potential changes by securing potassium ion (K+) efflux and subsequent calcium ion (Ca2+) influx events in DCs upon stimulation, both of which are known to be required for proper function of DCs. At a molecular level, Dlg1 did so by retaining the integrity of voltage-gated K+ channels (including Kv1.3) in DCs. The loss of Dlg1 led to a decreased expression of K+ channels, resulting in impaired membrane potential changes and, as a consequence, reduced proinflammatory cytokine production, compromised Ag endocytosis, and phagocytosis. In conclusion, this study provided, to our knowledge, a novel insight into Dlg1 and the voltage-gated K+ channels axis in DC functions.
Collapse
Affiliation(s)
- Xuejiao Dong
- Ministry of Education Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Laboratory for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Lisi Wei
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Xueheng Guo
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, China.,National Education Examinations Authority, Beijing 100084, China
| | - Zhiyong Yang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20851
| | - Peiyu Li
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China.,Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People's Hospital, Guangdong Medical University, Shenzhen 518052, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China
| | - Hai Qi
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yan Shi
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaoyu Hu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Li Wu
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, China;
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China;
| | - Wanli Liu
- Ministry of Education Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Laboratory for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing 100084, China;
| |
Collapse
|
10
|
Yang JF, Cheng N, Ren S, Liu XM, Li XT. Characterization and molecular basis for the block of Kv1.3 channels induced by carvedilol in HEK293 cells. Eur J Pharmacol 2018; 834:206-212. [PMID: 30016664 DOI: 10.1016/j.ejphar.2018.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/04/2018] [Accepted: 07/13/2018] [Indexed: 10/28/2022]
Abstract
Carvedilol is a non-selective β-adrenoreceptor antagonist and exhibits a wide range of biological activities. The voltage-gated K+ (Kv) channel is one of the target ion channels of this compound. The rapidly activating Kv1.3 channel is expressed in several different tissues and plays an important role in the regulation of physiological functions, including cell proliferation and apoptosis. However, little is known about the possible action of carvedilol on Kv1.3 currents. Using the whole-cell configuration of the patch-clamp technique, we have revealed that exposure to carvedilol produced a concentration-dependent blocking of Kv1.3 channels heterologously expressed in HEK293 cells, with an IC50 value of 9.7 μM. This chemical decelerated the deactivation tail current of Kv1.3 currents, resulting in a tail crossover phenomenon. In addition, carvedilol generated a markedly hyperpolarizing shift (20 mV) of the inactivation curve, but failed to affect the activation curve. Mutagenesis experiments of Kv1.3 channels identified G427 and H451, two related sites of TEA block, as important residues for carvedilol-mediated blocking. The present results suggest that carvedilol acts directly on Kv1.3 currents by inducing closed- and open-channel block and helps to elucidate the mechanisms of action of this compound on Kv channels.
Collapse
Affiliation(s)
- Jin-Feng Yang
- College of Life Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Neng Cheng
- College of Life Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Sheng Ren
- College of Life Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Xiang-Ming Liu
- GongQing Institute of Science and Technology, Gongqing City 332020, China
| | - Xian-Tao Li
- College of Life Science, South-Central University for Nationalities, Wuhan 430074, China.
| |
Collapse
|
11
|
Lowinus T, Bose T, Busse S, Busse M, Reinhold D, Schraven B, Bommhardt UHH. Immunomodulation by memantine in therapy of Alzheimer's disease is mediated through inhibition of Kv1.3 channels and T cell responsiveness. Oncotarget 2018; 7:53797-53807. [PMID: 27462773 PMCID: PMC5288222 DOI: 10.18632/oncotarget.10777] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/09/2016] [Indexed: 12/20/2022] Open
Abstract
Memantine is approved for the treatment of advanced Alzheimer's disease (AD) and reduces glutamate-mediated neuronal excitotoxicity by antagonism of N-methyl-D-aspartate receptors. In the pathophysiology of AD immune responses deviate and infectious side effects are observed during memantine therapy. However, the particular effects of memantine on human T lymphocytes are unresolved. Here, we provide evidence that memantine blocks Kv1.3 potassium channels, inhibits CD3-antibody- and alloantigen-induced proliferation and suppresses chemokine-induced migration of peripheral blood T cells of healthy donors. Concurrent with the in vitro data, CD4+ T cells from AD patients receiving therapeutic doses of memantine show a transient decline of Kv1.3 channel activity and a long-lasting reduced proliferative response to alloantigens in mixed lymphocyte reactions. Furthermore, memantine treatment provokes a profound depletion of peripheral blood memory CD45RO+ CD4+ T cells. Thus, standard doses of memantine profoundly reduce T cell responses in treated patients through blockade of Kv1.3 channels. This may normalize deviant immunopathology in AD and contribute to the beneficial effects of memantine, but may also account for the enhanced infection rate.
Collapse
Affiliation(s)
- Theresa Lowinus
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Tanima Bose
- Molecular Physiology, Leibniz Institute for Neurobiology, Magdeburg, Germany.,Current address: Lee Kong Chian School of Medicine, Singapore
| | - Stefan Busse
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Mandy Busse
- Department of Pediatric Pulmonology & Allergology, Medical University of Hannover, Hannover, Germany
| | - Dirk Reinhold
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Department of Immune Control, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ursula H H Bommhardt
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
12
|
Tanner MR, Beeton C. Differences in ion channel phenotype and function between humans and animal models. FRONT BIOSCI-LANDMRK 2018; 23:43-64. [PMID: 28930537 PMCID: PMC5626566 DOI: 10.2741/4581] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ion channels play crucial roles in regulating a broad range of physiological processes. They form a very large family of transmembrane proteins. Their diversity results from not only a large number of different genes encoding for ion channel subunits but also the ability of subunits to assemble into homo- or heteromultimers, the existence of splice variants, and the expression of different regulatory subunits. These characteristics and the existence of very selective modulators make ion channels very attractive targets for therapy in a wide variety of pathologies. Some ion channels are already being targeted in the clinic while many more are being evaluated as novel drug targets in both clinical and preclinical studies. Advancing ion channel modulators from the bench to the clinic requires their assessment for safety and efficacy in animal models. While extrapolating results from one species to another is tempting, doing such without careful evaluation of the ion channels in different species presents a risk as the translation is not always straightforward. Here, we discuss differences between species in terms of ion channels expressed in selected tissues, differing roles of ion channels in some cell types, variable response to pharmacological agents, and human channelopathies that cannot fully be replicated in animal models.
Collapse
Affiliation(s)
- Mark R Tanner
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston TX 77030, and Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston TX 77030
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston TX 77030, and Center for Drug Discovery and Biology of Inflammation Center, Baylor College of Medicine, Houston TX 77030,
| |
Collapse
|
13
|
Kubota T, Correa AM, Bezanilla F. Mechanism of functional interaction between potassium channel Kv1.3 and sodium channel NavBeta1 subunit. Sci Rep 2017; 7:45310. [PMID: 28349975 PMCID: PMC5368567 DOI: 10.1038/srep45310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/22/2017] [Indexed: 11/29/2022] Open
Abstract
The voltage-gated potassium channel subfamily A member 3 (Kv1.3) dominantly expresses on T cells and neurons. Recently, the interaction between Kv1.3 and NavBeta1 subunits has been explored through ionic current measurements, but the molecular mechanism has not been elucidated yet. We explored the functional interaction between Kv1.3 and NavBeta1 through gating current measurements using the Cut-open Oocyte Voltage Clamp (COVC) technique. We showed that the N-terminal 1–52 sequence of hKv1.3 disrupts the channel expression on the Xenopus oocyte membrane, suggesting a potential role as regulator of hKv1.3 expression in neurons and lymphocytes. Our gating currents measurements showed that NavBeta1 interacts with the voltage sensing domain (VSD) of Kv1.3 through W172 in the transmembrane segment and modifies the gating operation. The comparison between G-V and Q-V with/without NavBeta1 indicates that NavBeta1 may strengthen the coupling between hKv1.3-VSD movement and pore opening, inducing the modification of kinetics in ionic activation and deactivation.
Collapse
Affiliation(s)
- Tomoya Kubota
- Department of Biochemistry and Molecular Biology, The University of Chicago, 929 E. 57th street, Chicago, IL 60637, USA
| | - Ana M Correa
- Department of Biochemistry and Molecular Biology, The University of Chicago, 929 E. 57th street, Chicago, IL 60637, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, The University of Chicago, 929 E. 57th street, Chicago, IL 60637, USA
| |
Collapse
|
14
|
Eil R, Vodnala SK, Clever D, Klebanoff CA, Sukumar M, Pan JH, Palmer DC, Gros A, Yamamoto TN, Patel SJ, Guittard GC, Yu Z, Carbonaro V, Okkenhaug K, Schrump DS, Linehan WM, Roychoudhuri R, Restifo NP. Ionic immune suppression within the tumour microenvironment limits T cell effector function. Nature 2016; 537:539-543. [PMID: 27626381 PMCID: PMC5204372 DOI: 10.1038/nature19364] [Citation(s) in RCA: 447] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 08/15/2016] [Indexed: 12/15/2022]
Abstract
Tumours progress despite being infiltrated by tumour-specific effector T cells. Tumours contain areas of cellular necrosis, which are associated with poor survival in a variety of cancers. Here, we show that necrosis releases intracellular potassium ions into the extracellular fluid of mouse and human tumours, causing profound suppression of T cell effector function. Elevation of the extracellular potassium concentration ([K+]e) impairs T cell receptor (TCR)-driven Akt-mTOR phosphorylation and effector programmes. Potassium-mediated suppression of Akt-mTOR signalling and T cell function is dependent upon the activity of the serine/threonine phosphatase PP2A. Although the suppressive effect mediated by elevated [K+]e is independent of changes in plasma membrane potential (Vm), it requires an increase in intracellular potassium ([K+]i). Accordingly, augmenting potassium efflux in tumour-specific T cells by overexpressing the potassium channel Kv1.3 lowers [K+]i and improves effector functions in vitro and in vivo and enhances tumour clearance and survival in melanoma-bearing mice. These results uncover an ionic checkpoint that blocks T cell function in tumours and identify potential new strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Robert Eil
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Suman K Vodnala
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - David Clever
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Christopher A Klebanoff
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Madhusudhanan Sukumar
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jenny H Pan
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Douglas C Palmer
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Alena Gros
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Tori N Yamamoto
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Shashank J Patel
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Geoffrey C Guittard
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Zhiya Yu
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Valentina Carbonaro
- Laboratory of Lymphocyte Signalling and Development, The Babraham, Institute, Cambridge, UK
| | - Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, The Babraham, Institute, Cambridge, UK
| | - David S Schrump
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - W Marston Linehan
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Rahul Roychoudhuri
- Laboratory of Lymphocyte Signalling and Development, The Babraham, Institute, Cambridge, UK
| | - Nicholas P Restifo
- National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
15
|
Ehling P, Meuth P, Eichinger P, Herrmann AM, Bittner S, Pawlowski M, Pankratz S, Herty M, Budde T, Meuth SG. Human T cells in silico: Modelling their electrophysiological behaviour in health and disease. J Theor Biol 2016; 404:236-250. [PMID: 27288542 DOI: 10.1016/j.jtbi.2016.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/27/2016] [Accepted: 06/01/2016] [Indexed: 01/27/2023]
Abstract
Although various types of ion channels are known to have an impact on human T cell effector functions, their exact mechanisms of influence are still poorly understood. The patch clamp technique is a well-established method for the investigation of ion channels in neurons and T cells. However, small cell sizes and limited selectivity of pharmacological blockers restrict the value of this experimental approach. Building a realistic T cell computer model therefore can help to overcome these kinds of limitations as well as reduce the overall experimental effort. The computer model introduced here was fed off ion channel parameters from literature and new experimental data. It is capable of simulating the electrophysiological behaviour of resting and activated human CD4(+) T cells under basal conditions and during extracellular acidification. The latter allows for the very first time to assess the electrophysiological consequences of tissue acidosis accompanying most forms of inflammation.
Collapse
Affiliation(s)
- Petra Ehling
- Department of Neurology, and Institute of Translational Neurology, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany.
| | - Patrick Meuth
- Department of Neurology, and Institute of Translational Neurology, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany
| | - Paul Eichinger
- Department of Neurology, and Institute of Translational Neurology, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany; Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München TUM, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Alexander M Herrmann
- Department of Neurology, and Institute of Translational Neurology, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany
| | - Stefan Bittner
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Matthias Pawlowski
- Department of Neurology, and Institute of Translational Neurology, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany; Wellcome Trust and MRC Cambridge Stem Cell Institute, and Anne McLaren Laboratory for Regenerative Medicine, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge, UK
| | - Susann Pankratz
- Department of Neurology, and Institute of Translational Neurology, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany
| | - Michael Herty
- RWTH Aachen University, Mathematics (Continuous optimization), Templergraben 55, 52056 Aachen, Germany
| | - Thomas Budde
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Robert-Koch-Str. 27a, 48149 Münster, Germany
| | - Sven G Meuth
- Department of Neurology, and Institute of Translational Neurology, Westfälische Wilhelms-Universität Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany
| |
Collapse
|
16
|
Pucca MB, Bertolini TB, Cerni FA, Bordon KCF, Peigneur S, Tytgat J, Bonato VL, Arantes EC. Immunosuppressive evidence of Tityus serrulatus toxins Ts6 and Ts15: insights of a novel K(+) channel pattern in T cells. Immunology 2016; 147:240-50. [PMID: 26595158 DOI: 10.1111/imm.12559] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/05/2015] [Accepted: 11/10/2015] [Indexed: 12/25/2022] Open
Abstract
The voltage-gated potassium channel Kv1.3 is a novel target for immunomodulation of autoreactive effector memory T cells, which play a major role in the pathogenesis of autoimmune diseases. In this study, the Ts6 and Ts15 toxins isolated from Tityus serrulatus (Ts) were investigated for their immunosuppressant roles on CD4(+) cell subsets: naive, effector (TEF ), central memory (TCM) and effector memory (TEM). The electrophysiological assays confirmed that both toxins were able to block Kv1.3 channels. Interestingly, an extended Kv channel screening shows that Ts15 blocks Kv2.1 channels. Ts6 and Ts15 significantly inhibit the proliferation of TEM cells and interferon-γ production; however, Ts15 also inhibits other CD4(+) cell subsets (naive, TEF and TCM). Based on the Ts15 inhibitory effect of proliferation of all CD4(+) cell subsets, and based on its blocking effect on Kv2.1, we investigated the Kv2.1 expression in T cells. The assays showed that CD4(+) and CD8(+) cells express the Kv2.1 channels mainly extracellularly with TCM cells expressing the highest number of Kv2.1 channels. We also provide in vivo experimental evidence to the protective effect of Ts6 and Ts15 on delayed-type hypersensitivity reaction. Altogether, this study presents the immunosuppressive behaviour of Ts6 and Ts15 toxins, indicating that these toxins could be promising candidates for autoimmune disease therapy. Moreover, this is the first report illustrating the involvement of a novel K(+) channel subtype, Kv2.1, and its distribution in T-cell subsets.
Collapse
Affiliation(s)
- Manuela B Pucca
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Thaís B Bertolini
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Felipe A Cerni
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Karla C F Bordon
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Steve Peigneur
- Toxicology and Pharmacology, University of Leuven, Leuven, Belgium
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven, Leuven, Belgium
| | - Vânia L Bonato
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Eliane C Arantes
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
17
|
Voltage-Gated K+ Channel, Kv3.3 Is Involved in Hemin-Induced K562 Differentiation. PLoS One 2016; 11:e0148633. [PMID: 26849432 PMCID: PMC4743930 DOI: 10.1371/journal.pone.0148633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/20/2016] [Indexed: 12/31/2022] Open
Abstract
Voltage-gated K+ (Kv) channels are well known to be involved in cell proliferation. However, even though cell proliferation is closely related to cell differentiation, the relationship between Kv channels and cell differentiation remains poorly investigated. This study demonstrates that Kv3.3 is involved in K562 cell erythroid differentiation. Down-regulation of Kv3.3 using siRNA-Kv3.3 increased hemin-induced K562 erythroid differentiation through decreased activation of signal molecules such as p38, cAMP response element-binding protein, and c-fos. Down-regulation of Kv3.3 also enhanced cell adhesion by increasing integrin β3 and this effect was amplified when the cells were cultured with fibronectin. The Kv channels, or at least Kv3.3, appear to be associated with cell differentiation; therefore, understanding the mechanisms of Kv channel regulation of cell differentiation would provide important information regarding vital cellular processes.
Collapse
|
18
|
Normal human CD4(+) helper T cells express Kv1.1 voltage-gated K(+) channels, and selective Kv1.1 block in T cells induces by itself robust TNFα production and secretion and activation of the NFκB non-canonical pathway. J Neural Transm (Vienna) 2015; 123:137-57. [PMID: 26611796 DOI: 10.1007/s00702-015-1446-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 08/16/2015] [Indexed: 10/22/2022]
Abstract
TNFα is a very potent and pleiotropic pro-inflammatory cytokine, essential to the immune system for eradicating cancer and microorganisms, and to the nervous system, for brain development and ongoing function. Yet, excess and/or chronic TNFα secretion causes massive tissue damage in autoimmune, inflammatory and neurological diseases and injuries. Therefore, many patients with autoimmune/inflammatory diseases receive anti-TNFα medications. TNFα is secreted primarily by CD4(+) T cells, macrophages, monocytes, neutrophils and NK cells, mainly after immune stimulation. Yet, the cause for the pathologically high and chronic TNFα secretion is unknown. Can blocking of a particular ion channel in T cells induce by itself TNFα secretion? Such phenomenon was never revealed or even hypothesized. In this interdisciplinary study we discovered that: (1) normal human T cells express Kv1.1 voltage-gated potassium channel mRNA, and the Kv1.1 membrane-anchored protein channel; (2) Kv1.1 is expressed in most CD4(+)CD3(+) helper T cells (mean CD4(+)CD3(+)Kv1.1(+) T cells of 7 healthy subjects: 53.09 ± 22.17 %), but not in CD8(+)CD3(+) cytotoxic T cells (mean CD8(+)CD3(+)Kv1.1(+) T cells: 4.12 ± 3.04 %); (3) electrophysiological whole-cell recordings in normal human T cells revealed Kv currents; (4) Dendrotoxin-K (DTX-K), a highly selective Kv1.1 blocker derived from snake toxin, increases the rate of rise and decay of Kv currents in both resting and activated T cells, without affecting the peak current; (5) DTX-K by itself induces robust TNFα production and secretion by normal human T cells, without elevating IFNγ, IL-4 and IL-10; (6) intact Ca(2+) channels are required for DTX-induced TNFα secretion; (7) selective anti-Kv1.1 antibodies also induce by themselves TNFα secretion; (8) DTX-K activates NFκB in normal human T cells via the unique non-canonical-pathway; (9) injection of Kv1.1-blocked human T cells to SCID mice, causes recruitment of resident mouse cells into the liver, alike reported after TNFα injection into the brain. Based on our discoveries we speculate that abnormally blocked Kv1.1 in T cells (and other immune cells?), due to either anti-Kv1.1 autoimmune antibodies, or Kv1.1-blocking toxins alike DTX-K, or Kv1.1-blocking genetic mutations, may be responsible for the chronic/excessive TNFα in autoimmune/inflammatory diseases. Independently, we also hypothesize that selective block of Kv1.1 in CD4(+) T cells of patients with cancer or chronic infectious diseases could be therapeutic, since it may: a. augment beneficial secretion and delivery of TNFα to the disease-affected sites; b. induce recruitment and extravasation of curative immune cells and factors; c. improve accessibility of drugs to the brain and few peripheral organs thanks to TNFα-induced increased permeability of organ's barriers.
Collapse
|
19
|
Toxins Targeting the Kv1.3 Channel: Potential Immunomodulators for Autoimmune Diseases. Toxins (Basel) 2015; 7:1749-64. [PMID: 25996605 PMCID: PMC4448172 DOI: 10.3390/toxins7051749] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/05/2015] [Accepted: 05/05/2015] [Indexed: 12/23/2022] Open
Abstract
Autoimmune diseases are usually accompanied by tissue injury caused by autoantigen-specific T-cells. KV1.3 channels participate in modulating calcium signaling to induce T-cell proliferation, immune activation and cytokine production. Effector memory T (TEM)-cells, which play major roles in many autoimmune diseases, are controlled by blocking KV1.3 channels on the membrane. Toxins derived from animal venoms have been found to selectively target a variety of ion channels, including KV1.3. By blocking the KV1.3 channel, these toxins are able to suppress the activation and proliferation of TEM cells and may improve TEM cell-mediated autoimmune diseases, such as multiple sclerosis and type I diabetes mellitus.
Collapse
|
20
|
Abstract
Ion channels and transporters mediate the transport of charged ions across hydrophobic lipid membranes. In immune cells, divalent cations such as calcium, magnesium, and zinc have important roles as second messengers to regulate intracellular signaling pathways. By contrast, monovalent cations such as sodium and potassium mainly regulate the membrane potential, which indirectly controls the influx of calcium and immune cell signaling. Studies investigating human patients with mutations in ion channels and transporters, analysis of gene-targeted mice, or pharmacological experiments with ion channel inhibitors have revealed important roles of ionic signals in lymphocyte development and in innate and adaptive immune responses. We here review the mechanisms underlying the function of ion channels and transporters in lymphocytes and innate immune cells and discuss their roles in lymphocyte development, adaptive and innate immune responses, and autoimmunity, as well as recent efforts to develop pharmacological inhibitors of ion channels for immunomodulatory therapy.
Collapse
Affiliation(s)
- Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, California 95616
| | - Edward Y. Skolnik
- Division of Nephrology, New York University School of Medicine, New York, NY 10016
- Department of Molecular Pathogenesis, New York University School of Medicine, New York, NY 10016
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016
- The Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
21
|
Zhang T, Luo XJ, Sai WB, Yu MF, Li WE, Ma YF, Chen W, Zhai K, Qin G, Guo D, Zheng YM, Wang YX, Shen JH, Ji G, Liu QH. Non-selective cation channels mediate chloroquine-induced relaxation in precontracted mouse airway smooth muscle. PLoS One 2014; 9:e101578. [PMID: 24992312 PMCID: PMC4081631 DOI: 10.1371/journal.pone.0101578] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/06/2014] [Indexed: 02/01/2023] Open
Abstract
Bitter tastants can induce relaxation in precontracted airway smooth muscle by activating big-conductance potassium channels (BKs) or by inactivating voltage-dependent L-type Ca2+ channels (VDLCCs). In this study, a new pathway for bitter tastant-induced relaxation was defined and investigated. We found nifedipine-insensitive and bitter tastant chloroquine-sensitive relaxation in epithelium-denuded mouse tracheal rings (TRs) precontracted with acetylcholine (ACH). In the presence of nifedipine (10 µM), ACH induced cytosolic Ca2+ elevation and cell shortening in single airway smooth muscle cells (ASMCs), and these changes were inhibited by chloroquine. In TRs, ACH triggered a transient contraction under Ca2+-free conditions, and, following a restoration of Ca2+, a strong contraction occurred, which was inhibited by chloroquine. Moreover, the ACH-activated whole-cell and single channel currents of non-selective cation channels (NSCCs) were blocked by chloroquine. Pyrazole 3 (Pyr3), an inhibitor of transient receptor potential C3 (TRPC3) channels, partially inhibited ACH-induced contraction, intracellular Ca2+ elevation, and NSCC currents. These results demonstrate that NSCCs play a role in bitter tastant-induced relaxation in precontracted airway smooth muscle.
Collapse
Affiliation(s)
- Ting Zhang
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Xiao-Jing Luo
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Wen-Bo Sai
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Meng-Fei Yu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Wen-Er Li
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yun-Fei Ma
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Weiwei Chen
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Kui Zhai
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Gangjian Qin
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Donglin Guo
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
- Lankenau Institute for Medical Research & Main Line Health Heart Center, Wynnewood, Pennsylvania, United States of America
| | - Yun-Min Zheng
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, United States of America
| | - Yong-Xiao Wang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, United States of America
| | - Jin-Hua Shen
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- * E-mail: (QHL); (GJ)
| | - Qing-Hua Liu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
- * E-mail: (QHL); (GJ)
| |
Collapse
|
22
|
Ishii T, Warabi E, Siow RCM, Mann GE. Sequestosome1/p62: a regulator of redox-sensitive voltage-activated potassium channels, arterial remodeling, inflammation, and neurite outgrowth. Free Radic Biol Med 2013; 65:102-116. [PMID: 23792273 DOI: 10.1016/j.freeradbiomed.2013.06.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 12/14/2022]
Abstract
Sequestosome1/p62 (SQSTM1) is an oxidative stress-inducible protein regulated by the redox-sensitive transcription factor Nrf2. It is not an antioxidant but known as a multifunctional regulator of cell signaling with an ability to modulate targeted or selective degradation of proteins through autophagy. SQSTM1 implements these functions through physical interactions with different types of proteins including atypical PKCs, nonreceptor-type tyrosine kinase p56(Lck) (Lck), polyubiquitin, and autophagosomal factor LC3. One of the notable physiological functions of SQSTM1 is the regulation of redox-sensitive voltage-gated potassium (Kv) channels which are composed of α and β subunits: (Kvα)4 (Kvβ)4. Previous studies have established that SQSTM1 scaffolds PKCζ, enhancing phosphorylation of Kvβ which induces inhibition of pulmonary arterial Kv1.5 channels under acute hypoxia. Recent studies reveal that Lck indirectly interacts with Kv1.3 α subunits and plays a key role in acute hypoxia-induced Kv1.3 channel inhibition in T lymphocytes. Kv1.3 channels provide a signaling platform to modulate the migration and proliferation of arterial smooth muscle cells and activation of T lymphocytes, and hence have been recognized as a therapeutic target for treatment of restenosis and autoimmune diseases. In this review, we focus on the functional interactions of SQSTM1 with Kv channels through two key partners aPKCs and Lck. Furthermore, we provide molecular insights into the functions of SQSTM1 in suppression of proliferation of arterial smooth muscle cells and neointimal hyperplasia following carotid artery ligation, in T lymphocyte differentiation and activation, and in NGF-induced neurite outgrowth in PC12 cells.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8575, Japan
| | - Richard C M Siow
- Cardiovascular Division, British Heart Foundation Centre of Research Excellence, School of Medicine, King's College London, London SE1 9NH, UK
| | - Giovanni E Mann
- Cardiovascular Division, British Heart Foundation Centre of Research Excellence, School of Medicine, King's College London, London SE1 9NH, UK
| |
Collapse
|
23
|
Vallejo-Gracia A, Bielanska J, Hernández-Losa J, Castellví J, Ruiz-Marcellan MC, Ramón y Cajal S, Condom E, Manils J, Soler C, Comes N, Ferreres JC, Felipe A. Emerging role for the voltage-dependent K+channel Kv1.5 in B-lymphocyte physiology: expression associated with human lymphoma malignancy. J Leukoc Biol 2013; 94:779-89. [DOI: 10.1189/jlb.0213094] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
24
|
Rao S, Liu X, Freedman BD, Behrens EM. Spleen tyrosine kinase (Syk)-dependent calcium signals mediate efficient CpG-induced exocytosis of tumor necrosis factor α (TNFα) in innate immune cells. J Biol Chem 2013; 288:12448-58. [PMID: 23515313 DOI: 10.1074/jbc.m113.454405] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Pattern recognition receptors expressed by cells of the innate immune system initiate the immune response upon recognition of microbial products. Activation of pattern recognition receptors result in the production and release of proinflammatory cytokines, including TNFα and IL-6. Because these cytokines promote disparate effector cell responses, understanding the signaling pathways involved in their regulation is critical for directing the immune response. Using macrophages and dendritic cells deficient in spleen tyrosine kinase (Syk), we identified a novel pathway by which TNFα trafficking and secretion are regulated by Syk following stimulation with CpG DNA. In the absence of PLCγ2, a Syk substrate, or the calcium-responsive kinase calcium calmodulin kinase II, CpG-induced TNFα secretion was impaired. Forced calcium mobilization rescued the TNFα secretion defect in Syk-deficient cells. In contrast to its effect on TNFα, Syk deficiency did not affect IL-6 secretion, suggesting that Syk-dependent signals participate in differential sorting of cytokines, thus tailoring the cytokine response. Our data report a novel pathway for TNFα regulation and provide insight into non-transcriptional mechanisms for shaping cytokine responses.
Collapse
Affiliation(s)
- Sheila Rao
- Division of Pediatric Rheumatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
25
|
Solé L, Vallejo-Gracia A, Roig SR, Serrano-Albarrás A, Marruecos L, Manils J, Gómez D, Soler C, Felipe A. KCNE gene expression is dependent on the proliferation and mode of activation of leukocytes. Channels (Austin) 2013; 7:85-96. [PMID: 23327879 DOI: 10.4161/chan.23258] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Voltage-dependent K (+) (Kv) channels are tightly regulated during the immune system response. Leukocytes have a limited repertoire of Kv channels, whose physiological role is under intense investigation. A functional Kv channel is an oligomeric complex composed of pore-forming and ancillary subunits. The KCNE gene family is a novel group of modulatory Kv channel elements in leukocytes. Here, we characterized the gene expression of KCNEs (1-5) in leukocytes and investigated their regulation during leukocyte proliferation and mode of activation. Murine bone-marrow-derived macrophages, human Jurkat T-lymphocytes and human Raji B-cells were analyzed. KCNEs (1-5) are expressed in all leukocytes lineages. Most KCNE mRNAs show cell cycle-dependent regulation and are differentially regulated under specific insults. Our results further suggest a new and yet undefined physiological role for KCNE subunits in the immune system. Putative associations of these ancillary proteins with Kv channels would yield a wide variety of biophysically and pharmacologically distinct channels that fine-tune the immunological response.
Collapse
Affiliation(s)
- Laura Solé
- Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular; Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gocke AR, Lebson LA, Grishkan IV, Hu L, Nguyen HM, Whartenby KA, Chandy KG, Calabresi PA. Kv1.3 deletion biases T cells toward an immunoregulatory phenotype and renders mice resistant to autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2012; 188:5877-86. [PMID: 22581856 DOI: 10.4049/jimmunol.1103095] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increasing evidence suggests ion channels have critical functions in the differentiation and plasticity of T cells. Kv1.3, a voltage-gated K(+) channel, is a functional marker and a pharmacological target for activated effector memory T cells. Selective Kv1.3 blockers have been shown to inhibit proliferation and cytokine production by human and rat effector memory T cells. We used Kv1.3 knockout (KO) mice to investigate the mechanism by which Kv1.3 blockade affects CD4(+) T cell differentiation during an inflammatory immune-mediated disease. Kv1.3 KO animals displayed significantly lower incidence and severity of myelin oligodendrocyte glycoprotein (MOG) peptide-induced experimental autoimmune encephalomyelitis. Kv1.3 was the only K(V) channel expressed in MOG 35-55-specific CD4(+) T cell blasts, and no K(V) current was present in MOG-specific CD4(+) T cell-blasts from Kv1.3 KO mice. Fewer CD4(+) T cells migrated to the CNS in Kv1.3 KO mice following disease induction, and Ag-specific proliferation of CD4(+) T cells from these mice was impaired with a corresponding cell-cycle delay. Kv1.3 was required for optimal expression of IFN-γ and IL-17, whereas its absence led to increased IL-10 production. Dendritic cells from Kv1.3 KO mice fully activated wild-type CD4(+) T cells, indicating a T cell-intrinsic defect in Kv1.3 KO mice. The loss of Kv1.3 led to a suppressive phenotype, which may contribute to the mechanism by which deletion of Kv1.3 produces an immunotherapeutic effect. Skewing of CD4(+) T cell differentiation toward Ag-specific regulatory T cells by pharmacological blockade or genetic suppression of Kv1.3 might be beneficial for therapy of immune-mediated diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Anne R Gocke
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Ion channels in autoimmune neurodegeneration. FEBS Lett 2011; 585:3836-42. [DOI: 10.1016/j.febslet.2011.03.065] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 03/26/2011] [Accepted: 03/28/2011] [Indexed: 11/23/2022]
|
28
|
Felipe A, Soler C, Comes N. Kv1.5 in the immune system: the good, the bad, or the ugly? Front Physiol 2010; 1:152. [PMID: 21423392 PMCID: PMC3059964 DOI: 10.3389/fphys.2010.00152] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 10/28/2010] [Indexed: 11/13/2022] Open
Abstract
For the last 20 years, knowledge of the physiological role of voltage-dependent potassium channels (Kv) in the immune system has grown exponentially. Leukocytes express a limited repertoire of Kv channels, which contribute to the membrane potential. These proteins are involved in the immune response and are therefore considered good pharmacological targets. Although there is a clear consensus about the physiological relevance of Kv1.3, the expression and the role of Kv1.5 are controversial. However, recent reports indicate that certain heteromeric Kv1.3/Kv1.5 associations may provide insight on Kv1.5. Here, we summarize what is known about this issue and highlight the role of Kv1.5 partnership interactions that could be responsible for this debate. The Kv1.3/Kv1.5 heterotetrameric composition of the channel and their possible differential associations with accessory regulatory proteins warrant further investigation.
Collapse
Affiliation(s)
- Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona Barcelona, Spain.
| | | | | |
Collapse
|
29
|
Haffner CD, Thomson SA, Guo Y, Petrov K, Larkin A, Banker P, Schaaf G, Dickerson S, Gobel J, Gillie D, Condreay JP, Poole C, Carpenter T, Ulrich J. Substituted N-{3-[(1,1-dioxido-1,2-benzothiazol-3-yl)(phenyl)amino]propyl}benzamide analogs as potent Kv1.3 ion channel blockers. Part 2. Bioorg Med Chem Lett 2010; 20:6989-92. [PMID: 20974533 DOI: 10.1016/j.bmcl.2010.09.131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 09/23/2010] [Accepted: 09/24/2010] [Indexed: 10/19/2022]
Abstract
We report the synthesis and in vitro activity of a series of novel substituted N-{3-[(1,1-dioxido-1,2-benzothiazol-3-yl)(phenyl)amino]propyl}benzamide analogs. These analogs showed potent inhibitory activity against Kv1.3. Several demonstrated similar potency to the known Kv1.3 inhibitor PAP-1 when tested under the IonWorks patch clamp assay conditions. Two compounds 13i and 13rr were advanced further as potential tool compounds for in vivo validation studies.
Collapse
Affiliation(s)
- Curt D Haffner
- Department of Medicinal Chemistry, GlaxoSmithKline Research and Development, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Rangaraju S, Chi V, Pennington MW, Chandy KG. Kv1.3 potassium channels as a therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2010; 13:909-24. [PMID: 19538097 DOI: 10.1517/14728220903018957] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We discuss the potential use of inhibitors of Kv1.3 potassium channels in T lymphocytes as therapeutics for multiple sclerosis. Current treatment strategies target the immune system in a non-selective manner. The resulting general immunosuppression, toxic side-effects and increased risk of opportunistic infections create the need for more selective therapeutics. Autoreactive effector-memory T (T(EM)) cells, considered to be major mediators of autoimmunity, express large numbers of Kv1.3 channels. Selective blockers of Kv1.3 inhibit calcium signaling, cytokine production and proliferation of T(EM) cells in vitro, and T(EM) cell-motility in vivo. Kv1.3 blockers ameliorate disease in animal models of multiple sclerosis, rheumatoid arthritis, type 1 diabetes mellitus and contact dermatitis without compromising the protective immune response to acute infections. Kv1.3 blockers have a good safety profile in rodents and primates.
Collapse
Affiliation(s)
- Srikant Rangaraju
- University of California, Department of Physiology and Biophysics, Irvine, California 92697, USA
| | | | | | | |
Collapse
|
31
|
Inhibition of the K+ channel KCa3.1 ameliorates T cell-mediated colitis. Proc Natl Acad Sci U S A 2010; 107:1541-6. [PMID: 20080610 DOI: 10.1073/pnas.0910133107] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The calcium-activated K(+) channel KCa3.1 plays an important role in T lymphocyte Ca(2+) signaling by helping to maintain a negative membrane potential, which provides an electrochemical gradient to drive Ca(2+) influx. To assess the role of KCa3.1 channels in lymphocyte activation in vivo, we studied T cell function in KCa3.1(-/-) mice. CD4 T helper (i.e., Th0) cells isolated from KCa3.1(-/-) mice lacked KCa3.1 channel activity, which resulted in decreased T cell receptor-stimulated Ca(2+) influx and IL-2 production. Although loss of KCa3.1 did not interfere with CD4 T cell differentiation, both Ca(2+) influx and cytokine production were impaired in KCa3.1(-/-) Th1 and Th2 CD4 T cells, whereas T-regulatory and Th17 function were normal. We found that inhibition of KCa3.1(-/-) protected mice from developing severe colitis in two mouse models of inflammatory bowel disease, which were induced by (i) the adoptive transfer of mouse naïve CD4 T cells into rag2(-/-) recipients and (ii) trinitrobenzene sulfonic acid. Pharmacologic inhibitors of KCa3.1 have already been shown to be safe in humans. Thus, if these preclinical studies continue to show efficacy, it may be possible to rapidly test whether KCa3.1 inhibitors are efficacious in patients with inflammatory bowel diseases such as Crohn's disease and ulcerative colitis.
Collapse
|
32
|
Abstract
The human genome encodes 40 voltage-gated K(+) channels (K(V)), which are involved in diverse physiological processes ranging from repolarization of neuronal and cardiac action potentials, to regulating Ca(2+) signalling and cell volume, to driving cellular proliferation and migration. K(V) channels offer tremendous opportunities for the development of new drugs to treat cancer, autoimmune diseases and metabolic, neurological and cardiovascular disorders. This Review discusses pharmacological strategies for targeting K(V) channels with venom peptides, antibodies and small molecules, and highlights recent progress in the preclinical and clinical development of drugs targeting the K(V)1 subfamily, the K(V)7 subfamily (also known as KCNQ), K(V)10.1 (also known as EAG1 and KCNH1) and K(V)11.1 (also known as HERG and KCNH2) channels.
Collapse
|
33
|
Abstract
Dynamic changes in cytoplasmic calcium concentration dictate the immunological fate and functions of lymphocytes. During the past few years, important details have been revealed about the mechanism of store-operated calcium entry in lymphocytes, including the molecular identity of calcium release-activated calcium (CRAC) channels and the endoplasmic reticulum (ER) calcium sensor (STIM1) responsible for CRAC channel activation following calcium depletion of stores. However, details of the potential fine regulation of CRAC channel activation that may be imposed on lymphocytes following physiologic stimulation within an inflammatory environment have not been fully addressed. In this review, we discuss several underexplored aspects of store-operated (CRAC-mediated) and store-independent calcium signaling in B lymphocytes. First, we discuss results suggesting that coupling between stores and CRAC channels may be regulated, allowing for fine tuning of CRAC channel activation following depletion of ER stores. Second, we discuss mechanisms that sustain the duration of calcium entry via CRAC channels. Finally, we discuss distinct calcium permeant non-selective cation channels (NSCCs) that are activated by innate stimuli in B cells, the potential means by which these innate calcium signaling pathways and CRAC channels crossregulate one another, and the mechanistic basis and physiologic consequences of innate calcium signaling.
Collapse
Affiliation(s)
- Leslie B King
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
34
|
Solé L, Roura-Ferrer M, Pérez-Verdaguer M, Oliveras A, Calvo M, Fernández-Fernández JM, Felipe A. KCNE4 suppresses Kv1.3 currents by modulating trafficking, surface expression and channel gating. J Cell Sci 2009; 122:3738-48. [PMID: 19773357 DOI: 10.1242/jcs.056689] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Voltage-dependent potassium channels (Kv) play a crucial role in the activation and proliferation of leukocytes. Kv channels are either homo- or hetero-oligomers. This composition modulates their surface expression and serves as a mechanism for regulating channel activity. Kv channel interaction with accessory subunits provides mechanisms for channels to respond to stimuli beyond changes in membrane potential. Here, we demonstrate that KCNE4 (potassium voltage-gated channel subfamily E member 4), but not KCNE2, functions as an inhibitory Kv1.3 partner in leukocytes. Kv1.3 trafficking, targeting and activity are altered by the presence of KCNE4. KCNE4 decreases current density, slows activation, accelerates inactivation, increases cumulative inactivation, retains Kv1.3 in the ER and impairs channel targeting to lipid raft microdomains. KCNE4 associates with Kv1.3 in the ER and decreases the number of Kv1.3 channels at the cell surface, which diminishes cell excitability. Kv1.3 and KCNE4 are differentially regulated upon activation or immunosuppression in macrophages. Thus, lipopolysaccharide-induced activation increases Kv1.3 and KCNE4 mRNA, whereas dexamethasone triggers a decrease in Kv1.3 with no changes in KCNE4. The channelosome composition determines the activity and affects surface expression and membrane localization. Therefore, KCNE4 association might play a crucial role in controlling immunological responses. Our results indicate that KCNE ancillary subunits could be new targets for immunomodulation.
Collapse
Affiliation(s)
- Laura Solé
- Departament de Bioquímica i Biologia Molecular, Molecular Physiology Laboratory, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 645, 08028 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
For more than 25 years, it has been widely appreciated that Ca2+ influx is essential to trigger T-lymphocyte activation. Patch clamp analysis, molecular identification, and functional studies using blockers and genetic manipulation have shown that a unique contingent of ion channels orchestrates the initiation, intensity, and duration of the Ca2+ signal. Five distinct types of ion channels--Kv1.3, KCa3.1, Orai1+ stromal interacting molecule 1 (STIM1) [Ca2+-release activating Ca2+ (CRAC) channel], TRPM7, and Cl(swell)--comprise a network that performs functions vital for ongoing cellular homeostasis and for T-cell activation, offering potential targets for immunomodulation. Most recently, the roles of STIM1 and Orai1 have been revealed in triggering and forming the CRAC channel following T-cell receptor engagement. Kv1.3, KCa3.1, STIM1, and Orai1 have been found to cluster at the immunological synapse following contact with an antigen-presenting cell; we discuss how channels at the synapse might function to modulate local signaling. Immuno-imaging approaches are beginning to shed light on ion channel function in vivo. Importantly, the expression pattern of Ca2+ and K+ channels and hence the functional network can adapt depending upon the state of differentiation and activation, and this allows for different stages of an immune response to be targeted specifically.
Collapse
Affiliation(s)
- Michael D Cahalan
- Department of Physiology and Biophysics, and the Institute for Immunology, University of California, Irvine, Irvine, CA 92697-4561, USA.
| | | |
Collapse
|
36
|
Nolz JC, Medeiros RB, Mitchell JS, Zhu P, Freedman BD, Shimizu Y, Billadeau DD. WAVE2 regulates high-affinity integrin binding by recruiting vinculin and talin to the immunological synapse. Mol Cell Biol 2007; 27:5986-6000. [PMID: 17591693 PMCID: PMC1952166 DOI: 10.1128/mcb.00136-07] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
T-cell-receptor (TCR)-mediated integrin activation is required for T-cell-antigen-presenting cell conjugation and adhesion to extracellular matrix components. While it has been demonstrated that the actin cytoskeleton and its regulators play an essential role in this process, no mechanism has been established which directly links TCR-induced actin polymerization to the activation of integrins. Here, we demonstrate that TCR stimulation results in WAVE2-ARP2/3-dependent F-actin nucleation and the formation of a complex containing WAVE2, ARP2/3, vinculin, and talin. The verprolin-connecting-acidic (VCA) domain of WAVE2 mediates the formation of the ARP2/3-vinculin-talin signaling complex and talin recruitment to the immunological synapse (IS). Interestingly, although vinculin is not required for F-actin or integrin accumulation at the IS, it is required for the recruitment of talin. In addition, RNA interference of either WAVE2 or vinculin inhibits activation-dependent induction of high-affinity integrin binding to VCAM-1. Overall, these findings demonstrate a mechanism in which signals from the TCR produce WAVE2-ARP2/3-mediated de novo actin polymerization, leading to integrin clustering and high-affinity binding through the recruitment of vinculin and talin.
Collapse
Affiliation(s)
- Jeffrey C Nolz
- Department of Immunology and Division of Oncology Research, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Rebeaud F, Hailfinger S, Thome M. Dlgh1 and Carma1 MAGUK proteins contribute to signal specificity downstream of TCR activation. Trends Immunol 2007; 28:196-200. [PMID: 17395537 DOI: 10.1016/j.it.2007.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 02/26/2007] [Accepted: 03/16/2007] [Indexed: 10/23/2022]
Abstract
Mitogen-activated protein kinases (MAPKs), including p38 and c-Jun N-terminal kinase (JNK), have a key role in T cell receptor (TCR)-induced gene transcription but their precise mechanism of activation is not well understood. The findings of two recent papers provide new insight into the activation of p38 and JNK by the membrane-associated guanylate kinase (MAGUK) family members Dlgh1 and Carma1, respectively, and show how distinct MAGUK proteins control specific aspects of TCR-mediated MAPK activation.
Collapse
Affiliation(s)
- Fabien Rebeaud
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | | | | |
Collapse
|
38
|
Azam P, Sankaranarayanan A, Homerick D, Griffey S, Wulff H. Targeting effector memory T cells with the small molecule Kv1.3 blocker PAP-1 suppresses allergic contact dermatitis. J Invest Dermatol 2007; 127:1419-29. [PMID: 17273162 PMCID: PMC1929164 DOI: 10.1038/sj.jid.5700717] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The voltage-gated potassium channel Kv1.3 has been recently identified as a molecular target that allows for selective pharmacological suppression of effector memory T (T(EM)) cells without affecting the function of naïve and central memory T cells. We here investigated whether PAP-1, a small molecule Kv1.3 blocker (EC50=2 nM), could suppress allergic contact dermatitis (ACD). In a rat model of ACD, we first confirmed that the infiltrating cells in the elicitation phase are indeed CD8+ CD45RC- memory T cells with high Kv1.3 expression. In accordance with its selective effect on T(EM) cells, PAP-1 did not impair sensitization, but potently suppressed oxazolone-induced inflammation by inhibiting the infiltration of CD8+ T cells and reducing the production of the inflammatory cytokines IFN-gamma, IL-2, and IL-17 when administered intraperitoneally or orally during the elicitation phase. PAP-1 was equally effective when applied topically, demonstrating that it effectively penetrates skin. We further show that PAP-1 is not a sensitizer or an irritant and exhibits no toxicity in a 28-day toxicity study. Based on these results we propose that PAP-1 could potentially be developed into a drug for the topical treatment of inflammatory skin diseases such as psoriasis.
Collapse
Affiliation(s)
- Philippe Azam
- Department of Medical Pharmacology and Toxicology, University of California, Davis, California, USA
| | | | - Daniel Homerick
- Department of Medical Pharmacology and Toxicology, University of California, Davis, California, USA
| | - Stephen Griffey
- Comparative Pathology Laboratory University of California, Davis, California, USA
| | - Heike Wulff
- Department of Medical Pharmacology and Toxicology, University of California, Davis, California, USA
| |
Collapse
|
39
|
Villalonga N, Escalada A, Vicente R, Sánchez-Tilló E, Celada A, Solsona C, Felipe A. Kv1.3/Kv1.5 heteromeric channels compromise pharmacological responses in macrophages. Biochem Biophys Res Commun 2007; 352:913-8. [PMID: 17157812 DOI: 10.1016/j.bbrc.2006.11.120] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Accepted: 11/21/2006] [Indexed: 12/31/2022]
Abstract
Voltage-dependent K(+) (Kv) channels are involved in the immune response. Kv1.3 is highly expressed in activated macrophages and T-effector memory cells of autoimmune disease patients. Macrophages are actively involved in T-cell activation by cytokine production and antigen presentation. However, unlike T-cells, macrophages express Kv1.5, which is resistant to Kv1.3-drugs. We demonstrate that mononuclear phagocytes express different Kv1.3/Kv1.5 ratios, leading to biophysically and pharmacologically distinct channels. Therefore, Kv1.3-based treatments to alter physiological responses, such as proliferation and activation, are impaired by Kv1.5 expression. The presence of Kv1.5 in the macrophagic lineage should be taken into account when designing Kv1.3-based therapies.
Collapse
Affiliation(s)
- Núria Villalonga
- Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Avda. Diagonal 645, E-08028 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Eck SC, Zhu P, Pepper M, Bensinger SJ, Freedman BD, Laufer TM. Developmental alterations in thymocyte sensitivity are actively regulated by MHC class II expression in the thymic medulla. THE JOURNAL OF IMMUNOLOGY 2006; 176:2229-37. [PMID: 16455979 DOI: 10.4049/jimmunol.176.4.2229] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Developing thymocytes are positively selected if they respond to self-MHC-peptide complexes, yet mature T cells are not activated by those same self-complexes. To avoid autoimmunity, positive selection must be followed by a period of maturation when the cellular response to TCR signals is altered. The mechanisms that mediate this postselection developmental tuning remain largely unknown. Specifically, it is unknown whether developmental tuning is a preprogrammed outcome of positive selection or if it is sensitive to ongoing interactions between the thymocyte and the thymic stroma. We probed the requirement for MHC class II-TCR interactions in postselection maturation by studying single positive (SP) CD4 thymocytes from K14/A(beta)(b) mice, in which CD4 T cells cannot interact with MHC class II in the thymic medulla. We report here that SP CD4 thymocytes must receive MHC class II signals to avoid hyperactive responses to TCR signals. This hyperactivity correlates with decreased expression of CD5; however, developmental tuning can occur independently of CD5, correlating instead with differences in the distribution of Lck. Thus, the maturation of postselection SP CD4 thymocytes is an active process mediated by ongoing interactions between the T cell and MHC class II molecules. This represents a novel mechanism by which the thymic medulla prevents autoreactivity.
Collapse
Affiliation(s)
- Steven C Eck
- Department of Medicine, University of Pennsylvania, Philadephia, 19104, USA
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Multiple sclerosis is a chronic inflammatory autoimmune disease of the central nervous system characterized by demyelination and axonal damage that result in disabling neurological deficits. Here the authors explain the rationale for the use of inhibitors of the Kv1.3 K+ channel in immune cells as a therapy for multiple sclerosis and other autoimmune disorders.
Collapse
Affiliation(s)
- Christine Beeton
- Department of Physiology and Biophysics, Medical School, University of California, Irvine, 92697, USA
| | | |
Collapse
|
42
|
Liu X, Zhu P, Freedman BD. Multiple eicosanoid-activated nonselective cation channels regulate B-lymphocyte adhesion to integrin ligands. Am J Physiol Cell Physiol 2005; 290:C873-82. [PMID: 16251477 DOI: 10.1152/ajpcell.00229.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Arachidonic acid (AA) is a substrate for a variety of proinflammatory mediators, which are generated by cyclooxygenases (COXs), lipoxygenases (LOXs), and cytochrome P-450 (CYP450) enzymes. COX (e.g., PGs and prostacyclins) and LOX (e.g., leukotrienes) products have well-established proinflammatory roles; however, little is known about the functions of CYP450 products in leukocytes. We previously found that mechanical strain generated by subjecting lymphocytes to hypotonic challenge triggered AA production and that two CYP450 products of AA, 5,6-epoxyeicosatrienoic acid (5,6-EET) and 20-hydroxyeicosatetraenoic acid (20-HETE), as well as a product of LOX, 5-(S)-hydroperoxyeicosatetrenoic acid (5-HPETE), induced Ca(2+) entry into primary B cells. The main goal of the present studies, therefore, was to define the biophysically properties of eicosanoid-activated channels responsible for Ca(2+) entry and the physiological consequences of activating these channels, including their role in mechanical signaling. We found that 5,6-EET, 20-HETE, and 5-HPETE each activated distinct Ca(2+)-permeant nonselective cation channels (NSCCs) in primary B cells. These NSCCs each regulate plasma membrane potential and B-cell adhesion to integrin ligands ICAM-1 and VCAM-1. Thus our data demonstrate that proinflammatory mediators produced in response to osmotic and/or physical stress play a direct role in regulating the B-cell membrane potential and their adhesion to specific ECM proteins. These results not only have important implications for understanding normal mechanisms of B-cell activation, differentiation, and trafficking but also point to novel targets for modulating the pathogenesis of B-cell-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 368E Old Vet Bldg., 3800 Spruce St., Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
43
|
Liu QH, Liu X, Wen Z, Hondowicz B, King L, Monroe J, Freedman BD. Distinct Calcium Channels Regulate Responses of Primary B Lymphocytes to B Cell Receptor Engagement and Mechanical Stimuli. THE JOURNAL OF IMMUNOLOGY 2004; 174:68-79. [PMID: 15611229 DOI: 10.4049/jimmunol.174.1.68] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Intracellular Ca(2+) plays a central role in controlling lymphocyte function. Nonetheless, critical gaps remain in our understanding of the mechanisms that regulate its concentration. Although Ca(2+)-release-activated calcium (CRAC) channels are the primary Ca(2+) entry pathways in T cells, additional pathways appear to be operative in B cells. Our efforts to delineate these pathways in primary murine B cells reveal that Ca(2+)-permeant nonselective cation channels (NSCCs) operate in a cooperative fashion with CRAC. Interestingly, these non-CRAC channels are selectively activated by mechanical stress, although the mechanism overlaps with BCR-activated pathways, suggesting that they may operate in concert to produce functionally diverse Ca(2+) signals. NSCCs also regulate the membrane potential, which activates integrin-dependent binding of B cells to extracellular matrix elements involved in their trafficking and localization within secondary lymphoid organs. Thus, CRAC and distinct Ca(2+) permeant NSCCs are differentially activated by the BCR and mechanical stimuli and regulate distinct aspects of B cell physiology.
Collapse
Affiliation(s)
- Qing-Hua Liu
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Wulff H, Knaus HG, Pennington M, Chandy KG. K+ channel expression during B cell differentiation: implications for immunomodulation and autoimmunity. THE JOURNAL OF IMMUNOLOGY 2004; 173:776-86. [PMID: 15240664 DOI: 10.4049/jimmunol.173.2.776] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Using whole-cell patch-clamp, fluorescence microscopy and flow cytometry, we demonstrate a switch in potassium channel expression during differentiation of human B cells from naive to memory cells. Naive and IgD(+)CD27(+) memory B cells express small numbers of the voltage-gated Kv1.3 and the Ca(2+)-activated intermediate-conductance IKCa1 channel when quiescent, and increase IKCa1 expression 45-fold upon activation with no change in Kv1.3 levels. In contrast, quiescent class-switched memory B cells express high levels of Kv1.3 ( approximately 2000 channels/cell) and maintain their Kv1.3(high) expression after activation. Consistent with their channel phenotypes, proliferation of naive and IgD(+)CD27(+) memory B cells is suppressed by the specific IKCa1 inhibitor TRAM-34 but not by the potent Kv1.3 blocker Stichodactyla helianthus toxin, whereas the proliferation of class-switched memory B cells is suppressed by Stichodactyla helianthus toxin but not TRAM-34. These changes parallel those reported for T cells. Therefore, specific Kv1.3 and IKCa1 inhibitors may have use in therapeutic manipulation of selective lymphocyte subsets in immunological disorders.
Collapse
Affiliation(s)
- Heike Wulff
- Department of Medical Pharmacology and Toxicology, University of California, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
45
|
Nam JH, Woo JE, Uhm DY, Kim SJ. Membrane-delimited regulation of novel background K+ channels by MgATP in murine immature B cells. J Biol Chem 2004; 279:20643-54. [PMID: 15014072 DOI: 10.1074/jbc.m312547200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In WEHI-231, a representative immature B cell line, Ca(2+) entry is paradoxically augmented by treatment with 2-aminoethoxydiphenyl borate (2-APB), a blocker of inositol 1,4,5-trisphosphate receptor and of nonselective cation channels (Nam, J. H., Yun, S. S., Kim, T. J., Uhm, D.-Y., and Kim, S. J. (2003) FEBS Lett. 535, 113-118). The initial goal of the present study was to elucidate the effects of 2-APB on membrane currents, which revealed the presence of novel K(+) channels in WEHI-231 cells. Under whole-cell patch clamp conditions, 2-APB induced background K(+) current (I(K,bg)) and hyperpolarization in WEHI-231 cells. Lowering of intracellular MgATP also induced the I(K,bg). The I(K,bg) was blocked by micromolar concentrations of quinidine but not by tetraethylammonium. In a single channel study, two types of voltage-independent K(+) channels were found with large (346 picosiemens) and medium conductance (112 picosiemens), named BK(bg) and MK(bg), respectively. The excision of membrane patches (inside-out (i-o) patches) greatly increased the P(o) of BK(bg). In i-o patches, cytoplasmic MgATP (IC(50) = 0.18 mm) decreased the BK(bg) activity, although non-hydrolyzable adenosine 5'-(beta,gamma-imino)triphosphate had no effect. A pretreatment with Al(3+) or wortmannin (50 microm) blocked the inhibitory effects of MgATP. A direct application of phosphoinositide 4,5-bisphosphate (10 microm) inhibited the BK(bg) activity. Meanwhile, the activity of MK(bg) was unaffected by MgATP. In cell-attached conditions, the BK(bg) activity was largely increased by 2-APB. In i-o patches, however, the MgATP-induced inhibition of BK(bg) was weakly reversed by the addition of 2-APB. In summary, WEHI-231 cells express the unique background K(+) channels. The BK(bg)s are inhibited by membrane-delimited elevation of phosphoinositide 4,5-bisphosphate. The activation of BK(bg) would hyperpolarize the membrane, which augments the calcium influx in WEHI-231 cells.
Collapse
Affiliation(s)
- Joo Hyun Nam
- Department of Physiology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Changan-Gu, Cheoncheon-Dong 300, Suwon 440-746, Korea
| | | | | | | |
Collapse
|
46
|
Xu J, Wang P, Li Y, Li G, Kaczmarek LK, Wu Y, Koni PA, Flavell RA, Desir GV. The voltage-gated potassium channel Kv1.3 regulates peripheral insulin sensitivity. Proc Natl Acad Sci U S A 2004; 101:3112-7. [PMID: 14981264 PMCID: PMC365752 DOI: 10.1073/pnas.0308450100] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Kv1.3 is a voltage-gated potassium (K) channel expressed in a number of tissues, including fat and skeletal muscle. Channel inhibition improves experimental autoimmune encephalitis, in part by reducing IL-2 and tumor necrosis factor production by peripheral T lymphocytes. Gene inactivation causes mice (Kv1.3-/-) exposed to a high-fat diet to gain less weight and be less obese than littermate control. Interestingly, although Kv1.3-/- mice on the high-calorie diet gain weight, they remain euglycemic, with low blood insulin levels. This observation prompted us to examine the effect of Kv1.3 gene inactivation and inhibition on peripheral glucose homeostasis and insulin sensitivity. Here we show that Kv1.3 gene deletion and channel inhibition increase peripheral insulin sensitivity in vivo. Baseline and insulin-stimulated glucose uptake are increased in adipose tissue and skeletal muscle of Kv1.3-/- mice. Inhibition of Kv1.3 activity facilitates the translocation of the glucose transporter, GLUT4, to the plasma membrane. It also suppresses c-JUN terminal kinase activity in fat and skeletal muscle and decreases IL-6 and tumor necrosis factor secretion by adipose tissue. We conclude that Kv1.3 inhibition improves insulin sensitivity by increasing the amount of GLUT4 at the plasma membrane. These results pinpoint a pathway through which K channels regulate peripheral glucose homeostasis, and identify Kv1.3 as a pharmacologic target for the treatment of diabetes.
Collapse
Affiliation(s)
- Jianchao Xu
- Department of Medicine,Yale University School of Medicine, and Veterans Affairs Medical Center, New Haven, CT, USA
| | | | | | | | | | | | | | | | | |
Collapse
|