1
|
Boda VK, Yasmen N, Jiang J, Li W. Pathophysiological significance and modulation of the transient receptor potential canonical 3 ion channel. Med Res Rev 2024; 44:2510-2544. [PMID: 38715347 PMCID: PMC11452291 DOI: 10.1002/med.22048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024]
Abstract
Transient receptor potential canonical 3 (TRPC3) protein belongs to the TRP family of nonselective cation channels. Its activation occurs by signaling through a G protein-coupled receptor (GPCR) and a phospholipase C-dependent (PLC) pathway. Perturbations in the expression of TRPC3 are associated with a plethora of pathophysiological conditions responsible for disorders of the cardiovascular, immune, and central nervous systems. The recently solved cryo-EM structure of TRPC3 provides detailed inputs about the underlying mechanistic aspects of the channel, which in turn enables more efficient ways of designing small-molecule modulators. Pharmacologically targeting TRPC3 in animal models has demonstrated great efficacy in treating diseases including cancers, neurological disorders, and cardiovascular diseases. Despite extensive scientific evidence supporting some strong correlations between the expression and activity of TRPC3 and various pathophysiological conditions, therapeutic strategies based on its pharmacological modulations have not led to clinical trials. The development of small-molecule TRPC3 modulators with high safety, sufficient brain penetration, and acceptable drug-like profiles remains in progress. Determining the pathological mechanisms for TRPC3 involvement in human diseases and understanding the requirements for a drug-like TRPC3 modulator will be valuable in advancing small-molecule therapeutics to future clinical trials. In this review, we provide an overview of the origin and activation mechanism of TRPC3 channels, diseases associated with irregularities in their expression, and new development in small-molecule modulators as potential therapeutic interventions for treating TRPC3 channelopathies.
Collapse
Affiliation(s)
- Vijay K. Boda
- Department of Pharmaceutical Sciences, and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Nelufar Yasmen
- Department of Pharmaceutical Sciences, and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| |
Collapse
|
2
|
Xue C, Li X, Ba L, Shen Y, Sun Z, Gu J, Yang Y, Han Q, Zhao RC. Irisin mediates beiging of adipose-derived mesenchymal stem cells through binding to TRPC3. BMC Biol 2022; 20:95. [PMID: 35501783 PMCID: PMC9063202 DOI: 10.1186/s12915-022-01287-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Beiging of white fat plays an important role in energy metabolism. Beige adipocytes contribute to the regulation of body weight and body temperature through expenditure of chemical energy to produce heat, and they have therefore recently attracted considerable attention as potential targets for therapeutic approaches in metabolic disorders, including obesity. All adipocytes, including beige adipocytes, differentiate from mesenchymal stem cells (MSCs), which may provide an important path for clinical intervention; however, the mechanism of beiging of human adipose cell-derived MSCs is not fully understood. Here, we provide insights on the role of IRISIN, which is known to be secreted by skeletal muscle and promote beiging of white fat. RESULTS We established an IRISIN-induced mesenchymal stem cell beiging model and found that IRISIN protein interacts with the MSC membrane protein TRPC3. This interaction results in calcium influx and consequential activation of Erk and Akt signaling pathways, which causes phosphorylation of PPARγ. The phosphorylated PPARγ enters the nucleus and binds the UCP1 promoter region. Furthermore, the role of TRPC3 in the beiging of MSCs was largely abolished in Trpc3-/- mice. We additionally demonstrate that the calcium concentration in the brain of mice increases upon IRISIN stimulation, followed by an increase in the content of excitatory amino acids and norepinephrine, while Trpc3-/- mice exhibit the reverse effect. CONCLUSIONS We found that TRPC3 is a key factor in irisin-induced beiging of MSCs, which may provide a new target pathway in addressing metabolic disorders. Our results additionally suggest that the interaction of irisin with TRPC3 may affect multiple tissues, including the brain.
Collapse
Affiliation(s)
- Chunling Xue
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China.
| | - Xuechun Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China
| | - Li Ba
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China
| | - Yamei Shen
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China
| | - Zhao Sun
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Junjie Gu
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Ying Yang
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China.
| |
Collapse
|
3
|
Distribution and Assembly of TRP Ion Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1349:111-138. [PMID: 35138613 DOI: 10.1007/978-981-16-4254-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the last several decades, a large family of ion channels have been identified and studied intensively as cellular sensors for diverse physical and/or chemical stimuli. Named transient receptor potential (TRP) channels, they play critical roles in various aspects of cellular physiology. A large number of human hereditary diseases are found to be linked to TRP channel mutations, and their dysregulations lead to acute or chronical health problems. As TRP channels are named and categorized mostly based on sequence homology rather than functional similarities, they exhibit substantial functional diversity. Rapid advances in TRP channel study have been made in recent years and reported in a vast body of literature; a summary of the latest advancements becomes necessary. This chapter offers an overview of current understandings of TRP channel distribution and subunit assembly.
Collapse
|
4
|
Zhang S, Romero LO, Deng S, Wang J, Li Y, Yang L, Hamilton DJ, Miller DD, Liao FF, Cordero-Morales JF, Wu Z, Li W. Discovery of a Highly Selective and Potent TRPC3 Inhibitor with High Metabolic Stability and Low Toxicity. ACS Med Chem Lett 2021; 12:572-578. [PMID: 33859797 PMCID: PMC8040052 DOI: 10.1021/acsmedchemlett.0c00571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
The overactivation of transient receptor potential canonical 3 (TRPC3) is associated with neurodegenerative diseases and hypertension. Pyrazole 3 (Pyr3) is reported as the most selective TRPC3 inhibitor, but it has two inherent structural limitations: (1) the labile ester moiety leads to its rapid hydrolysis to the inactive Pyr8 in vivo, and (2) the alkylating trichloroacrylic amide moiety is known to be toxic. To circumvent these limitations, we designed a series of conformationally restricted Pyr3 analogues and reported that compound 20 maintains high potency and selectivity for human TRPC3 over its closely related TRP channels. It has significantly improved metabolic stability compared with Pyr3 and has a good safety profile. Preliminary evaluation of 20 demonstrated its ability to rescue Aβ-induced neuron damage with similar potency to that of Pyr3 in vitro. Collectively, these results suggest that 20 represents a promising scaffold to potentially ameliorate the symptoms associated with TRPC3-mediated neurological and cardiovascular disorders.
Collapse
Affiliation(s)
- Sicheng Zhang
- Department
of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Luis O. Romero
- Department
of Physiology, the University of Tennessee
Health Science Center, Memphis, Tennessee 38163, United States
- Integrated
Biomedical Sciences Graduate Program, College
of Graduate Health Sciences, Memphis, Tennessee 38163, United States
| | - Shanshan Deng
- Department
of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jiaxing Wang
- Department
of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Yong Li
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Lei Yang
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - David J. Hamilton
- Department
of Comparative Medicine, College of Graduate Health Sciences, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D. Miller
- Department
of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Francesca-Fang Liao
- Department
of Pharmacology, Addiction Science, and Toxicology, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Julio F. Cordero-Morales
- Department
of Physiology, the University of Tennessee
Health Science Center, Memphis, Tennessee 38163, United States
| | - Zhongzhi Wu
- Department
of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Wei Li
- Department
of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
5
|
Froghi S, Grant CR, Tandon R, Quaglia A, Davidson B, Fuller B. New Insights on the Role of TRP Channels in Calcium Signalling and Immunomodulation: Review of Pathways and Implications for Clinical Practice. Clin Rev Allergy Immunol 2021; 60:271-292. [PMID: 33405100 PMCID: PMC7985118 DOI: 10.1007/s12016-020-08824-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 12/14/2022]
Abstract
Calcium is the most abundant mineral in the human body and is central to many physiological processes, including immune system activation and maintenance. Studies continue to reveal the intricacies of calcium signalling within the immune system. Perhaps the most well-understood mechanism of calcium influx into cells is store-operated calcium entry (SOCE), which occurs via calcium release-activated channels (CRACs). SOCE is central to the activation of immune system cells; however, more recent studies have demonstrated the crucial role of other calcium channels, including transient receptor potential (TRP) channels. In this review, we describe the expression and function of TRP channels within the immune system and outline associations with murine models of disease and human conditions. Therefore, highlighting the importance of TRP channels in disease and reviewing potential. The TRP channel family is significant, and its members have a continually growing number of cellular processes. Within the immune system, TRP channels are involved in a diverse range of functions including T and B cell receptor signalling and activation, antigen presentation by dendritic cells, neutrophil and macrophage bactericidal activity, and mast cell degranulation. Not surprisingly, these channels have been linked to many pathological conditions such as inflammatory bowel disease, chronic fatigue syndrome and myalgic encephalomyelitis, atherosclerosis, hypertension and atopy.
Collapse
Affiliation(s)
- Saied Froghi
- Department of HPB & Liver Transplantation, Royal Free Hospital, Pond St, Hampstead, London, NW3 2QG, UK. .,Division of Surgery & Interventional Sciences/University College London (UCL), Royal Free Hospital, Pond Street, Hampstead, London, NW3 2QG, UK. .,HCA Senior Clinical Fellow (HPB & Liver Transplant), Wellington Hospital, St Johns Wood, London, UK.
| | - Charlotte R Grant
- Department of HPB & Liver Transplantation, Royal Free Hospital, Pond St, Hampstead, London, NW3 2QG, UK
| | - Radhika Tandon
- Sheffield Medical School, Beech Hill Road, Sheffield, UK, S10 2RX
| | - Alberto Quaglia
- Department of Pathology, Royal Free Hospital, Pond Street, Hampstead, London, NW3 2QG, UK
| | - Brian Davidson
- Department of HPB & Liver Transplantation, Royal Free Hospital, Pond St, Hampstead, London, NW3 2QG, UK.,Division of Surgery & Interventional Sciences/University College London (UCL), Royal Free Hospital, Pond Street, Hampstead, London, NW3 2QG, UK
| | - Barry Fuller
- Division of Surgery & Interventional Sciences/University College London (UCL), Royal Free Hospital, Pond Street, Hampstead, London, NW3 2QG, UK
| |
Collapse
|
6
|
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
|
7
|
DHA induces Jurkat T-cell arrest in G2/M phase of cell cycle and modulates the plasma membrane expression of TRPC3/6 channels. Biochimie 2020; 181:169-175. [PMID: 33333171 DOI: 10.1016/j.biochi.2020.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/28/2020] [Accepted: 12/12/2020] [Indexed: 12/23/2022]
Abstract
We investigated whether docosahexaenoic acid (DHA), a dietary n-3 fatty acid, modulates calcium (Ca2+) signaling and cell cycle progression in human Jurkat T-cells. Our study demonstrates that DHA inhibited Jurkat T-cell cycle progression by blocking their passage from S phase to G2/M phase. In addition, DHA decreased the plasma membrane expression of TRPC3 and TRPC6 calcium channels during T-cell proliferation. Interestingly, this fatty acid increased plasma membrane expression of TRPC6 after 24 h of mitogenic stimulation by phorbol-13-myristate-12-acetate (PMA) and ionomycin. These variations in the membrane expression of TRPC3 and TRPC6 channels were not directly correlated with the mRNA expression, indicating that it was a post-translational phenomenon. DHA increased free intracellular calcium concentrations, [Ca2+]i, via opening TRPC3 and TRPC6 channels. We conclude that the anti-proliferative effect of DHA might involve the modulation of TRPC3 and TRPC6 channels in human T-cells.
Collapse
|
8
|
Acharya TK, Tiwari A, Majhi RK, Goswami C. TRPM8 channel augments T-cell activation and proliferation. Cell Biol Int 2020; 45:198-210. [PMID: 33090595 DOI: 10.1002/cbin.11483] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 09/21/2020] [Accepted: 10/10/2020] [Indexed: 12/25/2022]
Abstract
The transient receptor potential melastatin 8 (TRPM8) is an ion channel that has been widely studied as a cold-sensitive nociceptor. However, its importance in nonneuronal cells is mostly unexplored. Here, we describe the presence and functional significance of endogenous TRPM8, a nonselective Ca2+ -channel in T cell functions. The major pool of TRPM8 resides at the T cell surface and its surface accumulation significantly increases in activated T cells. TRPM8 activation synergizes with T-cell receptor (TCR) stimulation to increase CD25, CD69 levels and enhances secretion of proinflammatory cytokine tumor necrosis factor. However, TRPM8 inhibition does not restrict TCR stimulation mediated activation of T cells, indicating that unlike the heat-sensitive TRPV1 and TRPV4 channels, the cold-sensitive TRPM8 channel may be dispensable during T-cell activation, at least in mice. In this study, we demonstrate that TRPM8 promotes TCR-induced intracellular calcium increase. TRPM8 activation is beneficial for T-cell activation and differentiation into effector cells. TRPM8 inhibition during the T-cell activation process may lead to altered phenotype and reduced proliferation, without affecting cell viability. These results collectively establish TRPM8 as a functional calcium channel whose activation may be utilized for mounting an effective immune response. The findings of this study will be relevant to the regulation and response of T cells during cell-mediated immunity. These results will likely further our understanding on the role of ion channels in T-cell activation.
Collapse
Affiliation(s)
- Tusar K Acharya
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Odisha, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Ankit Tiwari
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Odisha, India
| | - Rakesh K Majhi
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Odisha, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Chandan Goswami
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Odisha, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
9
|
Pethő Z, Najder K, Carvalho T, McMorrow R, Todesca LM, Rugi M, Bulk E, Chan A, Löwik CWGM, Reshkin SJ, Schwab A. pH-Channeling in Cancer: How pH-Dependence of Cation Channels Shapes Cancer Pathophysiology. Cancers (Basel) 2020; 12:E2484. [PMID: 32887220 PMCID: PMC7565548 DOI: 10.3390/cancers12092484] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/20/2022] Open
Abstract
Tissue acidosis plays a pivotal role in tumor progression: in particular, interstitial acidosis promotes tumor cell invasion, and is a major contributor to the dysregulation of tumor immunity and tumor stromal cells. The cell membrane and integral membrane proteins commonly act as important sensors and transducers of altered pH. Cell adhesion molecules and cation channels are prominent membrane proteins, the majority of which is regulated by protons. The pathophysiological consequences of proton-sensitive ion channel function in cancer, however, are scarcely considered in the literature. Thus, the main focus of this review is to highlight possible events in tumor progression and tumor immunity where the pH sensitivity of cation channels could be of great importance.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Karolina Najder
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Tiago Carvalho
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
| | - Luca Matteo Todesca
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Micol Rugi
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Etmar Bulk
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Alan Chan
- Percuros B.V., 2333 CL Leiden, The Netherlands;
| | - Clemens W. G. M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
- Department of Oncology CHUV, UNIL and Ludwig Cancer Center, 1011 Lausanne, Switzerland
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| |
Collapse
|
10
|
Nam JH, Kim WK. The Role of TRP Channels in Allergic Inflammation and its Clinical Relevance. Curr Med Chem 2020; 27:1446-1468. [PMID: 30474526 DOI: 10.2174/0929867326666181126113015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 09/03/2018] [Accepted: 11/07/2018] [Indexed: 12/24/2022]
Abstract
Allergy refers to an abnormal adaptive immune response to non-infectious environmental substances (allergen) that can induce various diseases such as asthma, atopic dermatitis, and allergic rhinitis. In this allergic inflammation, various immune cells, such as B cells, T cells, and mast cells, are involved and undergo complex interactions that cause a variety of pathophysiological conditions. In immune cells, calcium ions play a crucial role in controlling intracellular Ca2+ signaling pathways. Cations, such as Na+, indirectly modulate the calcium signal generation by regulating cell membrane potential. This intracellular Ca2+ signaling is mediated by various cation channels; among them, the Transient Receptor Potential (TRP) family is present in almost all immune cell types, and each channel has a unique function in regulating Ca2+ signals. In this review, we focus on the role of TRP ion channels in allergic inflammatory responses in T cells and mast cells. In addition, the TRP ion channels, which are attracting attention in clinical practice in relation to allergic diseases, and the current status of the development of therapeutic agents that target TRP channels are discussed.
Collapse
Affiliation(s)
- Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea.,Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do 10326, Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do 10326, Korea.,Department of Internal Medicine Graduate School of Medicine, Dongguk University, 27 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do 10326, Korea
| |
Collapse
|
11
|
Yang PC, Jafri MS. Ca 2+ signaling in T lymphocytes: the interplay of the endoplasmic reticulum, mitochondria, membrane potential, and CRAC channels on transcription factor activation. Heliyon 2020; 6:e03526. [PMID: 32181396 PMCID: PMC7063158 DOI: 10.1016/j.heliyon.2020.e03526] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/12/2018] [Accepted: 02/28/2020] [Indexed: 11/16/2022] Open
Abstract
T cell receptor stimulation initiates a cascade of reactions that cause an increase in intracellular calcium (Ca2+) concentration mediated through inositol 1,4,5-trisphosphate (IP3). To understand the basic mechanisms by which the immune response in T cells is activated, it is useful to understand the signaling pathways that contain important targets for drugs in a quantitative fashion. A computational model helps us to understand how the selected elements in the pathways interact with each other, and which component plays the crucial role in systems. We have developed a mathematical model to explore the mechanism for controlling transcription factor activity, which regulates gene expression, by the modulation of calcium signaling triggered during T cell activation. The model simulates the activation and modulation of Ca2+ release-activated Ca2+ (CRAC) channels by mitochondrial dynamics and depletion of endoplasmic reticulum (ER) store, and also includes membrane potential in T-cells. The model simulates the experimental finding that increases in Ca2+ current enhances the activation of transcription factors and the Ca2+ influx through CRAC is also essential for the NFAT and NFκB activation. The model also suggests that plasma membrane Ca2+-ATPase (PMCA) controls a majority of the extrusion of Ca2+ and modulates the activation of CRAC channels. Furthermore, the model simulations explain how the complex interaction of the endoplasmic reticulum, membrane potential, mitochondria, and ion channels such as CRAC channels control T cell activation.
Collapse
Affiliation(s)
- Pei-Chi Yang
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, 95616, USA
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA
| | - M. Saleet Jafri
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 20201, USA
| |
Collapse
|
12
|
Arimilli S, Makena P, Prasad GL. Combustible Cigarette and Smokeless Tobacco Product Preparations Differentially Regulate Intracellular Calcium Mobilization in HL60 Cells. Inflammation 2020; 42:1641-1651. [PMID: 31190105 PMCID: PMC6719334 DOI: 10.1007/s10753-019-01025-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Changes in the level of intracellular calcium ([Ca2+]i) are central to leukocyte signaling and immune response. Although evidence suggests that cigarette smoking affects inflammatory response via an increase in intracellular calcium, it remains unclear if the use of smokeless tobacco (e.g., moist snuff) elicits a similar response. In this study, we evaluated the effects of tobacco product preparations (TPPs), including total particulate matter (TPM) from 3R4F reference cigarettes, smokeless tobacco extract (STE) from 2S3 reference moist snuff, and nicotine alone on Ca2+ mobilization in HL60 cells. Treatment with TPM, but not STE or nicotine alone, significantly increased [Ca2+]i in a concentration-dependent manner in HL60 cells. Moreover, TPM-induced [Ca2+]i increase was not related to extracellular Ca2+ and did not require the activation of the IP3 pathway nor involved the transient receptor potential (TRP) channels. Our findings indicate that, in cells having either intact or depleted endoplasmic reticulum (ER) Ca2+ stores, TPM-mediated [Ca2+]i increase involves cytosolic Ca2+ pools other than thapsigargin-sensitive ER Ca2+ stores. These results, for the first time, demonstrate that TPM triggers [Ca2+]i increases, while significantly higher nicotine equivalent doses of STE or nicotine alone, did not affect [Ca2+]i under the experimental conditions. In summary, our study suggests that in contrast with STE or nicotine preparations, TPM activates Ca2+ signaling pathways in HL60 cells. The differential effect of combustible and non-combustible TPPs on Ca2+ mobilization could be a useful in vitro endpoint for tobacco product evaluation.
Collapse
Affiliation(s)
- S Arimilli
- Eurofins Lancaster Laboratories PSS, Winston-Salem, NC, 27105, USA
| | - P Makena
- RAI Services Company, Winston-Salem, NC, 27105, USA.
| | - G L Prasad
- RAI Services Company, Winston-Salem, NC, 27105, USA
| |
Collapse
|
13
|
Abstract
Calcium (Ca2+) signalling is of paramount importance to immunity. Regulated increases in cytosolic and organellar Ca2+ concentrations in lymphocytes control complex and crucial effector functions such as metabolism, proliferation, differentiation, antibody and cytokine secretion and cytotoxicity. Altered Ca2+ regulation in lymphocytes leads to various autoimmune, inflammatory and immunodeficiency syndromes. Several types of plasma membrane and organellar Ca2+-permeable channels are functional in T cells. They contribute highly localized spatial and temporal Ca2+ microdomains that are required for achieving functional specificity. While the mechanistic details of these Ca2+ microdomains are only beginning to emerge, it is evident that through crosstalk, synergy and feedback mechanisms, they fine-tune T cell signalling to match complex immune responses. In this article, we review the expression and function of various Ca2+-permeable channels in the plasma membrane, endoplasmic reticulum, mitochondria and endolysosomes of T cells and their role in shaping immunity and the pathogenesis of immune-mediated diseases.
Collapse
Affiliation(s)
- Mohamed Trebak
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Jean-Pierre Kinet
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Fenninger F, Jefferies WA. What's Bred in the Bone: Calcium Channels in Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2019; 202:1021-1030. [PMID: 30718290 DOI: 10.4049/jimmunol.1800837] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/22/2018] [Indexed: 12/30/2022]
Abstract
Calcium (Ca2+) is an important second messenger in lymphocytes and is essential in regulating various intracellular pathways that control critical cell functions. Ca2+ channels are located in the plasma membrane and intracellular membranes, facilitating Ca2+ entry into the cytoplasm. Upon Ag receptor stimulation, Ca2+ can enter the lymphocyte via the Ca2+ release-activated Ca2+ channel found in the plasma membrane. The increase of cytosolic Ca2+ modulates signaling pathways, resulting in the transcription of target genes implicated in differentiation, activation, proliferation, survival, and apoptosis of lymphocytes. Along with Ca2+ release-activated Ca2+ channels, several other channels have been found in the membranes of T and B lymphocytes contributing to key cellular events. Among them are the transient receptor potential channels, the P2X receptors, voltage-dependent Ca2+ channels, and the inositol 1,4,5-trisphosphate receptor as well as the N-methyl-d-aspartate receptors. In this article, we review the contributions of these channels to mediating Ca2+ currents that drive specific lymphocyte functions.
Collapse
Affiliation(s)
- Franz Fenninger
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Wilfred A Jefferies
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada; .,Department of Microbiology and Immunology, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada.,Vancouver Prostate Centre, University of British Columbia, Vancouver V6H 3Z6, British Columbia, Canada.,Centre for Blood Research, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada.,The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada; and.,Department of Zoology, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| |
Collapse
|
15
|
TRPC-mediated Ca 2+ signaling and control of cellular functions. Semin Cell Dev Biol 2019; 94:28-39. [PMID: 30738858 DOI: 10.1016/j.semcdb.2019.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/30/2019] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
Abstract
Canonical members of the TRP superfamily of ion channels have long been recognized as key elements of Ca2+ handling in a plethora of cell types. The emerging role of TRPC channels in human physiopathology has generated considerable interest in their pharmacological targeting, which requires detailed understanding of their molecular function. Although consent has been reached that receptor-phospholipase C (PLC) pathways and generation of lipid mediators constitute the prominent upstream signaling process that governs channel activity, multimodal sensing features of TRPC complexes have been demonstrated repeatedly. Downstream signaling by TRPC channels is similarly complex and involves the generation of local and global cellular Ca2+ rises, which are well-defined in space and time to govern specific cellular functions. These TRPC-mediated Ca2+ signals rely in part on Ca2+ permeation through the channels, but are essentially complemented by secondary mechanisms such as Ca2+ mobilization from storage sites and Na+/Ca2+ exchange, which involve coordinated interaction with signaling partners. Consequently, the control of cell functions by TRPC molecules is critically determined by dynamic assembly and subcellular targeting of the TRPC complexes. The very recent availability of high-resolution structure information on TRPC channel complexes has paved the way towards a comprehensive understanding of signal transduction by TRPC channels. Here, we summarize current concepts of cation permeation in TRPC complexes, TRPC-mediated shaping of cellular Ca2+ signals and the associated control of specific cell functions.
Collapse
|
16
|
Tiapko O, Groschner K. TRPC3 as a Target of Novel Therapeutic Interventions. Cells 2018; 7:cells7070083. [PMID: 30037143 PMCID: PMC6071100 DOI: 10.3390/cells7070083] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/18/2018] [Accepted: 07/20/2018] [Indexed: 01/25/2023] Open
Abstract
TRPC3 is one of the classical members of the mammalian transient receptor potential (TRP) superfamily of ion channels. TRPC3 is a molecule with intriguing sensory features including the direct recognition of and activation by diacylglycerols (DAG). Although TRPC3 channels are ubiquitously expressed, they appear to control functions of the cardiovascular system and the brain in a highly specific manner. Moreover, a role of TRPC3 in immunity, cancer, and tissue remodeling has been proposed, generating much interest in TRPC3 as a target for pharmacological intervention. Advances in the understanding of molecular architecture and structure-function relations of TRPC3 have been the foundations for novel therapeutic approaches, such as photopharmacology and optochemical genetics of TRPC3. This review provides an account of advances in therapeutic targeting of TRPC3 channels.
Collapse
Affiliation(s)
- Oleksandra Tiapko
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/D04, 8010 Graz, Austria.
| | - Klaus Groschner
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/D04, 8010 Graz, Austria.
| |
Collapse
|
17
|
Ramirez GA, Coletto LA, Sciorati C, Bozzolo EP, Manunta P, Rovere-Querini P, Manfredi AA. Ion Channels and Transporters in Inflammation: Special Focus on TRP Channels and TRPC6. Cells 2018; 7:E70. [PMID: 29973568 PMCID: PMC6070975 DOI: 10.3390/cells7070070] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/27/2018] [Accepted: 06/29/2018] [Indexed: 12/14/2022] Open
Abstract
Allergy and autoimmune diseases are characterised by a multifactorial pathogenic background. Several genes involved in the control of innate and adaptive immunity have been associated with diseases and variably combine with each other as well as with environmental factors and epigenetic processes to shape the characteristics of individual manifestations. Systemic or local perturbations in salt/water balance and in ion exchanges between the intra- and extracellular spaces or among tissues play a role. In this field, usually referred to as elementary immunology, novel evidence has been recently acquired on the role of members of the transient potential receptor (TRP) channel family in several cellular mechanisms of potential significance for the pathophysiology of the immune response. TRP canonical channel 6 (TRPC6) is emerging as a functional element for the control of calcium currents in immune-committed cells and target tissues. In fact, TRPC6 influences leukocytes’ tasks such as transendothelial migration, chemotaxis, phagocytosis and cytokine release. TRPC6 also modulates the sensitivity of immune cells to apoptosis and influences tissue susceptibility to ischemia-reperfusion injury and excitotoxicity. Here, we provide a view of the interactions between ion exchanges and inflammation with a focus on the pathogenesis of immune-mediated diseases and potential future therapeutic implications.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Lavinia A Coletto
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Clara Sciorati
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Enrica P Bozzolo
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Paolo Manunta
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Nephrology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Patrizia Rovere-Querini
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Angelo A Manfredi
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
18
|
Profiling calcium signals of in vitro polarized human effector CD4 + T cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:932-943. [PMID: 29626493 DOI: 10.1016/j.bbamcr.2018.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/29/2018] [Accepted: 04/03/2018] [Indexed: 12/21/2022]
Abstract
Differentiation of naïve CD4+ T cells into effector subtypes with distinct cytokine profiles and physiological roles is a tightly regulated process, the imbalance of which can lead to an inadequate immune response or autoimmune disease. The crucial role of Ca2+ signals, mainly mediated by the store operated Ca2+ entry (SOCE) in shaping the immune response is well described. However, it is unclear if human effector CD4+ T cell subsets show differential Ca2+ signatures in response to different stimulation methods. Herein, we provide optimized in vitro culture conditions for polarization of human CD4+ effector T cells and characterize their SOCE following both pharmacological store depletion and direct T-cell receptor (TCR) activation. Moreover, we measured whole cell Ca2+ release activated Ca2+ currents (ICRAC) and investigated whether the observed differences correlate to the expression of CRAC genes. Our results show that Ca2+ profiles of helper CD4+ Th1, Th2 and Th17 are distinct and in part shaped by the intensity of stimulation. Regulatory T cells (Treg) are unique being the subtype with the most prominent SOCE response. Analysis of in vivo differentiated Treg unraveled the role of differential expression of ORAI2 in fine-tuning signals in Treg vs. conventional CD4+ T cells.
Collapse
|
19
|
Actions and Regulation of Ionotropic Cannabinoid Receptors. ADVANCES IN PHARMACOLOGY 2017; 80:249-289. [PMID: 28826537 DOI: 10.1016/bs.apha.2017.04.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Almost three decades have passed since the identification of the two specific metabotropic receptors mediating cannabinoid pharmacology. Thereafter, many cannabinoid effects, both at central and peripheral levels, have been well documented and characterized. However, numerous evidences demonstrated that these pharmacological actions could not be attributable solely to the activation of CB1 and CB2 receptors since several important cannabimimetic actions have been found in biological systems lacking CB1 or CB2 gene such as in specific cell lines or transgenic mice. It is now well accepted that, beyond their receptor-mediated effects, these molecules can act also via CB1/CB2-receptor-independent mechanism. Cannabinoids have been demonstrated to modulate several voltage-gated channels (including Ca2+, Na+, and various type of K+ channels), ligand-gated ion channels (i.e., GABA, glycine), and ion-transporting membranes proteins such as transient potential receptor class (TRP) channels. The first direct, cannabinoid receptor-independent interaction was reported on the function of serotonin 5-HT3 receptor-ion channel complex. Similar effects were reported also on the other above mentioned ion channels. In the early ninety, studies searching for endogenous modulators of L-type Ca2+ channels identified anandamide as ligand for L-type Ca2+ channel. Later investigations indicated that other types of Ca2+ currents are also affected by endocannabinoids, and, in the late ninety, it was discovered that endocannabinoids activate the vanilloid receptor subtype 1 (TRPV1), and nowadays, it is known that (endo)cannabinoids gate at least five distinct TRP channels. This chapter focuses on cannabinoid regulation of ion channels and lays special emphasis on their action at transient receptor channels.
Collapse
|
20
|
Ma S, Jiang Y, Huang W, Li X, Li S. Role of Transient Receptor Potential Channels in Heart Transplantation: A Potential Novel Therapeutic Target for Cardiac Allograft Vasculopathy. Med Sci Monit 2017; 23:2340-2347. [PMID: 28516902 PMCID: PMC5444344 DOI: 10.12659/msm.901920] [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] [Indexed: 12/19/2022] Open
Abstract
Heart transplantation has evolved as the criterion standard therapy for end-stage heart failure, but its efficacy is limited by the development of cardiac allograft vasculopathy (CAV), a unique and rapidly progressive form of atherosclerosis in heart transplant recipients. Here, we briefly review the key processes in the development of CAV during heart transplantation and highlight the roles of transient receptor potential (TRP) channels in these processes during heart transplantation. Understanding the roles of TRP channels in contributing to the key procedures for the development of CAV during heart transplantation could provide basic scientific knowledge for the development of new preventive and therapeutic approaches to manage patients with CAV after heart transplantation.
Collapse
Affiliation(s)
- Shuo Ma
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, China (mainland).,The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Yue Jiang
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, China (mainland).,The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Weiting Huang
- The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Xintao Li
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, China (mainland).,The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Shuzhuang Li
- Department of Physiology, Dalian Medical University, Dalian, China (mainland)
| |
Collapse
|
21
|
Mizoguchi Y, Monji A. TRPC Channels and Brain Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 976:111-121. [DOI: 10.1007/978-94-024-1088-4_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
22
|
Lin VHC, Chen JJ, Liao CC, Lee SS, Chien EJ. The rapid immunosuppression in phytohemagglutinin-activated human T cells is inhibited by the proliferative Ca(2+) influx induced by progesterone and analogs. Steroids 2016; 111:71-78. [PMID: 26808612 DOI: 10.1016/j.steroids.2016.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 01/11/2016] [Accepted: 01/20/2016] [Indexed: 10/22/2022]
Abstract
Progesterone, an endogenous immunomodulator, suppresses human T-cell activation during pregnancy. A sustained Ca(2 +) influx is an important signal for T-cell proliferation after crosslinking of T-cell receptor/CD3 complexes by anti-CD3 antibodies or phytohemagglutinin (PHA). Progesterone targets cell membrane sites inducing rapid responses including elevated intracellular free calcium concentration ([Ca(2+)]i) and suppressed T-cell PHA-activated proliferation. Interestingly, both PHA and progesterone induce [Ca(2+)]i elevation, but it remains unclear whether the PHA-induced Ca(2+) influx is affected by progesterone leading to T-cell immunosuppression. Primary T-cells were isolated from human peripheral blood and the quench effect on intracellular fura-2 fluorescence of Mn(2+) was used to explore the responses to Ca(2+) influx with cell proliferation being determined by MTT assay. PHA-stimulated Ca(2+) influx was dose-dependently suppressed by progesterone and its agonist R5020, which correlated with PHA-activated T-cell proliferation inhibition. A similar dose-dependent suppression effect on cellular Ca(2+) influx and proliferation occurred with the TRPC channel inhibitor BTP2 and selective TRPC3 channel inhibitor Pyr3. In addition, two progesterone analogs, Org OD 02-0 and 20α-hydroxyprogesterone (20α-OHP), also produced dose-dependent suppression of Ca(2+) influx, but had no effect on proliferation. Finally, inhibition of PHA-activated T-cell proliferation by progesterone is further suppressed by 20α-OHP, but not by Org OD 02-0. Overall, progesterone and R5020 are able to rapidly decrease PHA-stimulated sustained Ca(2+) influx, probably via blockade of TRPC3 channels, which suppresses T-cell proliferation. Taken together, the roles of progesterone and its analogs regarding the rapid response Ca(2+) influx need to be further explored in relation to cytokine secretion and proliferation in activated T-cells.
Collapse
Affiliation(s)
- Veronica Hui-Chen Lin
- Institute and Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan, ROC
| | - Jiann-Jong Chen
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, New Taipei City 23143, Taiwan, ROC
| | - Chen-Chung Liao
- Proteomics Research Center, National Yang-Ming University, Taipei 11221, Taiwan, ROC
| | - Shinn-Shing Lee
- Department of Medicine, Cheng Hsin General Hospital, Taipei 11220, Taiwan, ROC.
| | - Eileen Jea Chien
- Institute and Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan, ROC; Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan, ROC; Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan, ROC.
| |
Collapse
|
23
|
Grandclément C, Pick H, Vogel H, Held W. NK Cells Respond to Haptens by the Activation of Calcium Permeable Plasma Membrane Channels. PLoS One 2016; 11:e0151031. [PMID: 26963818 PMCID: PMC4786276 DOI: 10.1371/journal.pone.0151031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 02/23/2016] [Indexed: 01/07/2023] Open
Abstract
Natural Killer (NK) cells mediate innate immunity to infected and transformed cells. Yet, NK cells can also mount hapten-specific recall responses thereby contributing to contact hypersensitivity (CHS). However, since NK cells lack antigen receptors that are used by the adaptive immune system to recognize haptens, it is not clear if NK cells respond directly to haptens and, if so, what mediates these responses. Here we show that among four haptens the two that are known to induce NK cell-dependent CHS trigger the rapid influx of extracellular Ca2+ into NK cells and lymphocyte cell lines. Thus lymphocytes can respond to haptens independent of antigen presentation and antigen receptors. We identify the Ca2+-permeable cation channel TRPC3 as a component of the lymphocyte response to one of these haptens. These data suggest that the response to the second hapten is based on a distinct mechanism, consistent with the capacity of NK cells to discriminate haptens. These findings raise the possibility that antigen-receptor independent activation of immune cells contributes to CHS.
Collapse
Affiliation(s)
- Camille Grandclément
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Epalinges, Switzerland
| | - Horst Pick
- Laboratory of Physical Chemistry of Polymers and Membranes, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Horst Vogel
- Laboratory of Physical Chemistry of Polymers and Membranes, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Werner Held
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Epalinges, Switzerland
- * E-mail:
| |
Collapse
|
24
|
Parenti A, De Logu F, Geppetti P, Benemei S. What is the evidence for the role of TRP channels in inflammatory and immune cells? Br J Pharmacol 2016; 173:953-69. [PMID: 26603538 DOI: 10.1111/bph.13392] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/25/2015] [Accepted: 11/10/2015] [Indexed: 12/11/2022] Open
Abstract
A complex network of many interacting mechanisms orchestrates immune and inflammatory responses. Among these, the cation channels of the transient receptor potential (TRP) family expressed by resident tissue cells, inflammatory and immune cells and distinct subsets of primary sensory neurons, have emerged as a novel and interrelated system to detect and respond to harmful agents. TRP channels, by means of their direct effect on the intracellular levels of cations and/or through the indirect modulation of a large series of intracellular pathways, orchestrate a range of cellular processes, such as cytokine production, cell differentiation and cytotoxicity. The contribution of TRP channels to the transition of inflammation and immune responses from a defensive early response to a chronic and pathological condition is also emerging as a possible underlying mechanism in various diseases. This review discusses the roles of TRP channels in inflammatory and immune cell function and provides an overview of the effects of inflammatory and immune TRP channels on the pathogenesis of human diseases.
Collapse
Affiliation(s)
- A Parenti
- Clinical Pharmacology and Oncology Unit, Department of Health Sciences, University of Florence, Florence, Italy
| | - F De Logu
- Clinical Pharmacology and Oncology Unit, Department of Health Sciences, University of Florence, Florence, Italy
| | - P Geppetti
- Clinical Pharmacology and Oncology Unit, Department of Health Sciences, University of Florence, Florence, Italy
| | - S Benemei
- Clinical Pharmacology and Oncology Unit, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
25
|
Bertin S, Raz E. Transient Receptor Potential (TRP) channels in T cells. Semin Immunopathol 2015; 38:309-19. [PMID: 26468011 DOI: 10.1007/s00281-015-0535-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/01/2015] [Indexed: 12/16/2022]
Abstract
The transient receptor potential (TRP) family of ion channels is widely expressed in many cell types and plays various physiological roles. Growing evidence suggests that certain TRP channels are functionally expressed in the immune system. Indeed, an increasing number of reports have demonstrated the functional expression of several TRP channels in innate and adaptive immune cells and have highlighted their critical role in the activation and function of these cells. However, very few reviews have been entirely dedicated to this subject. Here, we will summarize the recent findings with regards to TRP channel expression in T cells and discuss their emerging role as regulators of T cell activation and functions. Moreover, these studies suggest that beyond their pharmaceutical interest in pain management, certain TRP channels may represent potential novel therapeutic targets for various immune-related diseases.
Collapse
Affiliation(s)
- Samuel Bertin
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA.
| | - Eyal Raz
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| |
Collapse
|
26
|
Abstract
Store-operated calcium channels (SOCs) are a major pathway for calcium signaling in virtually all metozoan cells and serve a wide variety of functions ranging from gene expression, motility, and secretion to tissue and organ development and the immune response. SOCs are activated by the depletion of Ca(2+) from the endoplasmic reticulum (ER), triggered physiologically through stimulation of a diverse set of surface receptors. Over 15 years after the first characterization of SOCs through electrophysiology, the identification of the STIM proteins as ER Ca(2+) sensors and the Orai proteins as store-operated channels has enabled rapid progress in understanding the unique mechanism of store-operate calcium entry (SOCE). Depletion of Ca(2+) from the ER causes STIM to accumulate at ER-plasma membrane (PM) junctions where it traps and activates Orai channels diffusing in the closely apposed PM. Mutagenesis studies combined with recent structural insights about STIM and Orai proteins are now beginning to reveal the molecular underpinnings of these choreographic events. This review describes the major experimental advances underlying our current understanding of how ER Ca(2+) depletion is coupled to the activation of SOCs. Particular emphasis is placed on the molecular mechanisms of STIM and Orai activation, Orai channel properties, modulation of STIM and Orai function, pharmacological inhibitors of SOCE, and the functions of STIM and Orai in physiology and disease.
Collapse
Affiliation(s)
- Murali Prakriya
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| | - Richard S Lewis
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
27
|
Kumar A, Kumari S, Majhi RK, Swain N, Yadav M, Goswami C. Regulation of TRP channels by steroids: Implications in physiology and diseases. Gen Comp Endocrinol 2015; 220:23-32. [PMID: 25449179 DOI: 10.1016/j.ygcen.2014.10.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 10/09/2014] [Accepted: 10/10/2014] [Indexed: 01/26/2023]
Abstract
While effects of different steroids on the gene expression and regulation are well established, it is proven that steroids can also exert rapid non-genomic actions in several tissues and cells. In most cases, these non-genomic rapid effects of steroids are actually due to intracellular mobilization of Ca(2+)- and other ions suggesting that Ca(2+) channels are involved in such effects. Transient Receptor Potential (TRP) ion channels or TRPs are the largest group of non-selective and polymodal ion channels which cause Ca(2+)-influx in response to different physical and chemical stimuli. While non-genomic actions of different steroids on different ion channels have been established to some extent, involvement of TRPs in such functions is largely unexplored. In this review, we critically analyze the literature and summarize how different steroids as well as their metabolic precursors and derivatives can exert non-genomic effects by acting on different TRPs qualitatively and/or quantitatively. Such effects have physiological repercussion on systems such as in sperm cells, immune cells, bone cells, neuronal cells and many others. Different TRPs are also endogenously expressed in diverse steroid-producing tissues and thus may have importance in steroid synthesis as well, a process which is tightly controlled by the intracellular Ca(2+) concentrations. Tissue and cell-specific expression of TRP channels are also regulated by different steroids. Understanding of the crosstalk between TRP channels and different steroids may have strong significance in physiological, endocrinological and pharmacological context and in future these compounds can also be used as potential biomedicine.
Collapse
Affiliation(s)
- Ashutosh Kumar
- School of Biology, National Institute of Science Education and Research, Sachivalaya Marg, Bhubaneswar, Orissa 751005, India
| | - Shikha Kumari
- School of Biology, National Institute of Science Education and Research, Sachivalaya Marg, Bhubaneswar, Orissa 751005, India
| | - Rakesh Kumar Majhi
- School of Biology, National Institute of Science Education and Research, Sachivalaya Marg, Bhubaneswar, Orissa 751005, India
| | - Nirlipta Swain
- School of Biology, National Institute of Science Education and Research, Sachivalaya Marg, Bhubaneswar, Orissa 751005, India
| | - Manoj Yadav
- School of Biology, National Institute of Science Education and Research, Sachivalaya Marg, Bhubaneswar, Orissa 751005, India
| | - Chandan Goswami
- School of Biology, National Institute of Science Education and Research, Sachivalaya Marg, Bhubaneswar, Orissa 751005, India.
| |
Collapse
|
28
|
Majhi RK, Sahoo SS, Yadav M, Pratheek BM, Chattopadhyay S, Goswami C. Functional expression of TRPV channels in T cells and their implications in immune regulation. FEBS J 2015; 282:2661-81. [PMID: 25903376 DOI: 10.1111/febs.13306] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/02/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022]
Abstract
The importance of Ca(2+) signalling and temperature in the context of T cell activation is well known. However, the molecular identities of key players involved in such critical regulations are still unknown. In this work we explored the endogenous expression of transient receptor potential vanilloid (TRPV) channels, a group of thermosensitive and non-selective cation channels, in T cells. Using flow cytometry and confocal microscopy, we demonstrate that members belonging to the TRPV subfamily are expressed endogenously in the human T cell line Jurkat, in primary human T cells and in primary murine splenic T cells. We also demonstrate that TRPV1- and TRPV4-specific agonists, namely resiniferatoxin and 4α-phorbol-12,13-didecanoate, can cause Ca(2+) influx in T cells. Moreover, our results show that expression of these channels can be upregulated in T cells during concanavalin A-driven mitogenic and anti-CD3/CD28 stimulated TCR activation of T cells. By specific blocking of TRPV1 and TRPV4 channels, we found that these TRPV inhibitors may regulate mitogenic and T cell receptor mediated T cell activation and effector cytokine(s) production by suppressing tumour necrosis factor, interleukin-2 and interferon-γ release. These results may have broad implications in the context of cell-mediated immunity, especially T cell responses and their regulations, neuro-immune interactions and molecular understanding of channelopathies.
Collapse
Affiliation(s)
- Rakesh K Majhi
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Subhransu S Sahoo
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Manoj Yadav
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Belluru M Pratheek
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Subhasis Chattopadhyay
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| | - Chandan Goswami
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, Orissa, India
| |
Collapse
|
29
|
Nohara LL, Stanwood SR, Omilusik KD, Jefferies WA. Tweeters, Woofers and Horns: The Complex Orchestration of Calcium Currents in T Lymphocytes. Front Immunol 2015; 6:234. [PMID: 26052328 PMCID: PMC4440397 DOI: 10.3389/fimmu.2015.00234] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/30/2015] [Indexed: 11/28/2022] Open
Abstract
Elevation of intracellular calcium ion (Ca2+) levels is a vital event that regulates T lymphocyte homeostasis, activation, proliferation, differentiation, and apoptosis. The mechanisms that regulate intracellular Ca2+ signaling in lymphocytes involve tightly controlled concinnity of multiple ion channels, membrane receptors, and signaling molecules. T cell receptor (TCR) engagement results in depletion of endoplasmic reticulum (ER) Ca2+ stores and subsequent sustained influx of extracellular Ca2+ through Ca2+ release-activated Ca2+ (CRAC) channels in the plasma membrane. This process termed store-operated Ca2+ entry (SOCE) involves the ER Ca2+ sensing molecule, STIM1, and a pore-forming plasma membrane protein, ORAI1. However, several other important Ca2+ channels that are instrumental in T cell function also exist. In this review, we discuss the role of additional Ca2+ channel families expressed on the plasma membrane of T cells that likely contribute to Ca2+ influx following TCR engagement, which include the TRP channels, the NMDA receptors, the P2X receptors, and the IP3 receptors, with a focus on the voltage-dependent Ca2+ (CaV) channels.
Collapse
Affiliation(s)
- Lilian L Nohara
- Michael Smith Laboratories, University of British Columbia , Vancouver, BC , Canada ; Department of Microbiology and Immunology, University of British Columbia , Vancouver, BC , Canada
| | - Shawna R Stanwood
- Michael Smith Laboratories, University of British Columbia , Vancouver, BC , Canada ; Department of Microbiology and Immunology, University of British Columbia , Vancouver, BC , Canada
| | - Kyla D Omilusik
- Michael Smith Laboratories, University of British Columbia , Vancouver, BC , Canada ; Department of Microbiology and Immunology, University of British Columbia , Vancouver, BC , Canada
| | - Wilfred A Jefferies
- Michael Smith Laboratories, University of British Columbia , Vancouver, BC , Canada ; Department of Microbiology and Immunology, University of British Columbia , Vancouver, BC , Canada ; Centre for Blood Research, University of British Columbia , Vancouver, BC , Canada ; The Djavad Mowafaghian Centre for Brain Health, University of British Columbia , Vancouver, BC , Canada ; Department of Medical Genetics, University of British Columbia , Vancouver, BC , Canada ; Department of Zoology, University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
30
|
Christo SN, Diener KR, Hayball JD. The functional contribution of calcium ion flux heterogeneity in T cells. Immunol Cell Biol 2015; 93:694-704. [PMID: 25823995 DOI: 10.1038/icb.2015.34] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/15/2015] [Accepted: 02/16/2015] [Indexed: 12/30/2022]
Abstract
The role of intracellular calcium ion oscillations in T-cell physiology is being increasingly appreciated by studies that describe how unique temporal and spatial calcium ion signatures can control different signalling pathways. Within this review, we provide detailed mechanisms of calcium ion oscillations, and emphasise the pivotal role that calcium signalling plays in directing crucial events pertaining to T-cell functionality. We also describe methods of calcium ion quantification, and take the opportunity to discuss how a deeper understanding of calcium signalling combined with new detection and quantification methodologies can be used to better design immunotherapies targeting T-cell responses.
Collapse
Affiliation(s)
- Susan N Christo
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Kerrilyn R Diener
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia, Adelaide, South Australia, Australia.,Robinson Research Institute, School of Paediatrics and Reproductive Health, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - John D Hayball
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia, Adelaide, South Australia, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
31
|
Abstract
The calcium ion (Ca(2+)) is the main common second messenger involved in signaling transduction subsequent to immunoreceptor activation. Its rapid intracellular elevation induces multiple cellular responses, such as secretion, proliferation, mobility, and gene transcription. Intracellular levels of Ca(2+) need to reach a specific threshold to efficiently transduce the signal to activate transcription factors through the recruitment of Ca(2+)-binding molecules. However, since Ca(2+) cannot be metabolized, its intracellular concentration is tightly regulated to avoid the induction of programmed cell death. This highly controlled regulation of Ca(2+) homeostasis has recently been clarified by the uncovering of new ion channels. The regulation of these channels allows the role of Ca(2+) in Fc receptor transduction pathways to be more precisely defined.
Collapse
Affiliation(s)
- Tarik Attout
- Inserm U1149, Bichat Medical School, Paris, France,
| | | | | |
Collapse
|
32
|
Wu QY, Sun MR, Wu CL, Li Y, Du JJ, Zeng JY, Bi HL, Sun YH. Activation of calcium-sensing receptor increases TRPC3/6 expression in T lymphocyte in sepsis. Mol Immunol 2014; 64:18-25. [PMID: 25467798 DOI: 10.1016/j.molimm.2014.10.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 10/23/2014] [Indexed: 01/17/2023]
Abstract
Sepsis is a systemic inflammatory response syndrome induced by infection. T Lymphocytes play an important role in this disease. Transient receptor potential (TRP) channels and calcium-sensing receptors (CaSR) are expressed in lymphocytes to promote intracellular Ca(2+) release. However, data about the link between CaSR and TRP channels in septic T lymphocytes are few. In this study, by Ca(2+) imaging and Western blotting, we found that in septic rat peripheral blood T lymphocytes expressions of TRPC3 and TRPC6 proteins are higher. The SR/ER Ca(2+) ATPase inhibitor thapsigargin (TG) and CaSR agonist NPS R-568 also increased expressions of TRPC3 and TRPC6 proteins, which were reversed by PLC-IP3 channel blocker U73122 and TRPC channels inhibitor SKF96365. By Ca(2+) imaging, we found that the depletion of ER Ca(2+) stores by TG elicited a transient rise in cytoplasmic Ca(2+), followed by sustained increase depending on extracellular Ca(2+). But, SKF96365, not Verapamil (L-type channels inhibitor) and NiCl2 (Na(+)/Ca(2+) exchanger inhibitor), inhibited the relatively high [Ca(2+)]i. NPS R-568 also resulted in the same effect, and the duration of [Ca(2+)]i increase was eliminated completely by U73122 and was reduced in the absence of [Ca(2+)]o. NPS R-568 and TG increased the apoptotic ratio of septic T lymphocytes, which can be suppressed by SKF96365 and U73122. These results suggested that CaSR activation promoted the expression of TRPC3 and TRPC6 and enhanced T lymphocytes apoptosis through PLC-IP3 signaling pathway in sepsis.
Collapse
Affiliation(s)
- Qiu-yue Wu
- Department of Clinical Laboratory, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ming-rui Sun
- Department of Pharmacology, Qiqihaer Medical College, Qiqihaer 160001, China
| | - Chun-li Wu
- Department of Clinical Laboratory, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yang Li
- Department of Rheumatology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jing-jing Du
- Department of Clinical Laboratory, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jing-ya Zeng
- Department of Clinical Laboratory, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Hai-liang Bi
- Department of Clinical Laboratory, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yi-hua Sun
- Department of Clinical Laboratory, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
33
|
Bertin S, Aoki-Nonaka Y, de Jong PR, Nohara LL, Xu H, Stanwood SR, Srikanth S, Lee J, To K, Abramson L, Yu T, Han T, Touma R, Li X, González-Navajas JM, Herdman S, Corr M, Fu G, Dong H, Gwack Y, Franco A, Jefferies WA, Raz E. The ion channel TRPV1 regulates the activation and proinflammatory properties of CD4⁺ T cells. Nat Immunol 2014; 15:1055-1063. [PMID: 25282159 PMCID: PMC4843825 DOI: 10.1038/ni.3009] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/11/2014] [Indexed: 12/11/2022]
Abstract
TRPV1 is a Ca(2+)-permeable channel studied mostly as a pain receptor in sensory neurons. However, its role in other cell types is poorly understood. Here we found that TRPV1 was functionally expressed in CD4(+) T cells, where it acted as a non-store-operated Ca(2+) channel and contributed to T cell antigen receptor (TCR)-induced Ca(2+) influx, TCR signaling and T cell activation. In models of T cell-mediated colitis, TRPV1 promoted colitogenic T cell responses and intestinal inflammation. Furthermore, genetic and pharmacological inhibition of TRPV1 in human CD4(+) T cells recapitulated the phenotype of mouse Trpv1(-/-) CD4(+) T cells. Our findings suggest that inhibition of TRPV1 could represent a new therapeutic strategy for restraining proinflammatory T cell responses.
Collapse
Affiliation(s)
- Samuel Bertin
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yukari Aoki-Nonaka
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, 5274 Gakkocho 2-ban-cho, Chuo-ku, Niigata 951-8514, Japan
| | - Petrus Rudolf de Jong
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lilian L. Nohara
- Michael Smith Laboratories; Centre for Blood Research; The Brain Research Centre; Department of Medical Genetics; Department of Microbiology and Immunology; Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Hongjian Xu
- Michael Smith Laboratories; Centre for Blood Research; The Brain Research Centre; Department of Medical Genetics; Department of Microbiology and Immunology; Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Shawna R. Stanwood
- Michael Smith Laboratories; Centre for Blood Research; The Brain Research Centre; Department of Medical Genetics; Department of Microbiology and Immunology; Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jihyung Lee
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Keith To
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lior Abramson
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Timothy Yu
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Tiffany Han
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ranim Touma
- Department of Pediatrics University of California, San Diego, La Jolla, CA 92093, USA
| | - Xiangli Li
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Scott Herdman
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Guo Fu
- Department of Immunology and Microbial Science, IMM1, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Fujian 361102, China
| | - Hui Dong
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alessandra Franco
- Department of Pediatrics University of California, San Diego, La Jolla, CA 92093, USA
| | - Wilfred A. Jefferies
- Michael Smith Laboratories; Centre for Blood Research; The Brain Research Centre; Department of Medical Genetics; Department of Microbiology and Immunology; Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Eyal Raz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
34
|
Nilius B, Szallasi A. Transient Receptor Potential Channels as Drug Targets: From the Science of Basic Research to the Art of Medicine. Pharmacol Rev 2014; 66:676-814. [DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
35
|
Physiological role of Kv1.3 channel in T lymphocyte cell investigated quantitatively by kinetic modeling. PLoS One 2014; 9:e89975. [PMID: 24594979 PMCID: PMC3940720 DOI: 10.1371/journal.pone.0089975] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/23/2014] [Indexed: 11/30/2022] Open
Abstract
Kv1.3 channel is a delayed rectifier channel abundant in human T lymphocytes. Chronic inflammatory and autoimmune disorders lead to the over-expression of Kv1.3 in T cells. To quantitatively study the regulatory mechanism and physiological function of Kv1.3 in T cells, it is necessary to have a precise kinetic model of Kv1.3. In this study, we firstly established a kinetic model capable to precisely replicate all the kinetic features for Kv1.3 channels, and then constructed a T-cell model composed of ion channels including Ca2+-release activated calcium (CRAC) channel, intermediate K+ (IK) channel, TASK channel and Kv1.3 channel for quantitatively simulating the changes in membrane potentials and local Ca2+ signaling messengers during activation of T cells. Based on the experimental data from current-clamp recordings, we successfully demonstrated that Kv1.3 dominated the membrane potential of T cells to manipulate the Ca2+ influx via CRAC channel. Our results revealed that the deficient expression of Kv1.3 channel would cause the less Ca2+ signal, leading to the less efficiency in secretion. This was the first successful attempt to simulate membrane potential in non-excitable cells, which laid a solid basis for quantitatively studying the regulatory mechanism and physiological role of channels in non-excitable cells.
Collapse
|
36
|
Abstract
TRPC3 represents one of the first identified mammalian relative of the Drosophila trp gene product. Despite extensive biochemical and biophysical characterization as well as ambitious attempts to uncover its physiological role in native cell systems, the channel protein still represents a rather enigmatic member of the TRP superfamily. TRPC3 is significantly expressed in the brain and heart and appears of (patho)physiological importance in both non-excitable and excitable cells, being potentially involved in a wide spectrum of Ca(2+) signaling mechanisms. TRPC3 cation channels display unique gating and regulatory properties that allow for recognition and integration of multiple input stimuli including lipid mediators, cellular Ca(2+) gradients, as well as redox signals. Physiological/pathophysiological functions of this highly versatile cation channel protein are as yet incompletely understood. Its ability to associate in a dynamic manner with a variety of partner proteins enables TRPC3 to serve coordination of multiple downstream signaling pathways and control of divergent cellular functions. Here, we summarize current knowledge on ion channel features as well as possible signaling functions of TRPC3 and discuss the potential biological relevance of this signaling molecule.
Collapse
Affiliation(s)
- Michaela Lichtenegger
- Institute of Pharmaceutical Sciences - Pharmacology and Toxicology, University of Graz, A-8010, Graz, Austria
| | | |
Collapse
|
37
|
Saul S, Stanisz H, Backes CS, Schwarz EC, Hoth M. How ORAI and TRP channels interfere with each other: interaction models and examples from the immune system and the skin. Eur J Pharmacol 2013; 739:49-59. [PMID: 24291108 DOI: 10.1016/j.ejphar.2013.10.071] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/08/2013] [Accepted: 10/17/2013] [Indexed: 11/16/2022]
Abstract
Four types of Ca(2+) selective ion channels are known, ten voltage gated Ca(2+) (CaV) channels, four CatSper channels, three store operated CRAC channels (ORAI channels) and at least two members of the TRPV subfamily (TRPV5, TRPV6). Some of the other TRP channels also show some Ca(2+) selectivity like certain splice variants of TRPM3. In addition to Ca(2+) selective channels, various cation channels play an important role for Ca(2+) entry and furthermore, they may also regulate Ca(2+) entry through other channels by modulating the membrane potential or other means as outlined in this review. Of the different types of cation channels, TRP channels form one of the most prominent families of non-selective cation channels with functional relevance in electrically non-excitable and electrically excitable cell types. Among these, the seven channels of the TRPC subfamily are rather non-selective with very modest Ca(2+) selectivity, whereas in the other subfamilies, cation selectivity ranges from monovalent selectivity (i.e. TRPM4, TRPM5) to divalent selectivity (i.e. TRPM6, TRPM7) or Ca(2+) selectivity (i.e. TRPV5, TRPV6). Rather than discussing the heavily reviewed individual functions of ORAI or TRP channels, we summarize data and present models how TRP and ORAI may functionally interact to guide cellular functions. We focus on T lymphocytes representing a more ORAI-dominated tissue and skin as model system in which both ORAI and TRP channel have been reported to control relevant functions. We present several interaction models how ORAI and TRP may interfere with each other's function.
Collapse
Affiliation(s)
- Stephanie Saul
- Department of Biophysics, School of Medicine, Saarland University, Homburg, Germany
| | - Hedwig Stanisz
- Department of Dermatology, School of Medicine, Saarland University, Homburg, Germany
| | - Christian S Backes
- Department of Biophysics, School of Medicine, Saarland University, Homburg, Germany
| | - Eva C Schwarz
- Department of Biophysics, School of Medicine, Saarland University, Homburg, Germany
| | - Markus Hoth
- Department of Biophysics, School of Medicine, Saarland University, Homburg, Germany.
| |
Collapse
|
38
|
Abstract
TRP channels constitute a large superfamily of cation channel forming proteins, all related to the gene product of the transient receptor potential (trp) locus in Drosophila. In mammals, 28 different TRP channel genes have been identified, which exhibit a large variety of functional properties and play diverse cellular and physiological roles. In this article, we provide a brief and systematic summary of expression, function, and (patho)physiological role of the mammalian TRP channels.
Collapse
Affiliation(s)
- Maarten Gees
- Laboratory Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Campus Gasthuisberg, Leuven, Belgium
| | | | | | | |
Collapse
|
39
|
Badou A, Jha MK, Matza D, Flavell RA. Emerging roles of L-type voltage-gated and other calcium channels in T lymphocytes. Front Immunol 2013; 4:243. [PMID: 24009608 PMCID: PMC3757574 DOI: 10.3389/fimmu.2013.00243] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 08/05/2013] [Indexed: 11/16/2022] Open
Abstract
In T lymphocytes, calcium ion controls a variety of biological processes including development, survival, proliferation, and effector functions. These distinct and specific roles are regulated by different calcium signals, which are generated by various plasma membrane calcium channels. The repertoire of calcium-conducting proteins in T lymphocytes includes store-operated CRAC channels, transient receptor potential channels, P2X channels, and L-type voltage-gated calcium (Cav1) channels. In this paper, we will focus mainly on the role of the Cav1 channels found expressed by T lymphocytes, where these channels appear to operate in a T cell receptor stimulation-dependent and voltage sensor independent manner. We will review their expression profile at various differentiation stages of CD4 and CD8 T lymphocytes. Then, we will present crucial genetic evidence in favor of a role of these Cav1 channels and related regulatory proteins in both CD4 and CD8 T cell functions such as proliferation, survival, cytokine production, and cytolysis. Finally, we will provide evidence and speculate on how these voltage-gated channels might function in the T lymphocyte, a non-excitable cell.
Collapse
Affiliation(s)
- Abdallah Badou
- Equipe de recherche Environnement et Santé, Faculté Polydisciplinaire de Safi, Université Cadi Ayyad , Safi , Morocco
| | | | | | | |
Collapse
|
40
|
Omilusik KD, Nohara LL, Stanwood S, Jefferies WA. Weft, warp, and weave: the intricate tapestry of calcium channels regulating T lymphocyte function. Front Immunol 2013; 4:164. [PMID: 23805141 PMCID: PMC3690356 DOI: 10.3389/fimmu.2013.00164] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/11/2013] [Indexed: 12/20/2022] Open
Abstract
Calcium (Ca(2+)) is a universal second messenger important for T lymphocyte homeostasis, activation, proliferation, differentiation, and apoptosis. The events surrounding Ca(2+) mobilization in lymphocytes are tightly regulated and involve the coordination of diverse ion channels, membrane receptors, and signaling molecules. A mechanism termed store-operated Ca(2+) entry (SOCE), causes depletion of endoplasmic reticulum (ER) Ca(2+) stores following T cell receptor (TCR) engagement and triggers a sustained influx of extracellular Ca(2+) through Ca(2+) release-activated Ca(2+) (CRAC) channels in the plasma membrane. The ER Ca(2+) sensing molecule, stromal interaction molecule 1 (STIM1), and a pore-forming plasma membrane protein, ORAI1, have been identified as important mediators of SOCE. Here, we review the role of several additional families of Ca(2+) channels expressed on the plasma membrane of T cells that likely contribute to Ca(2+) influx following TCR engagement, particularly highlighting an important role for voltage-dependent Ca(2+) channels (CaV) in T lymphocyte biology.
Collapse
Affiliation(s)
- Kyla D Omilusik
- Michael Smith Laboratories, University of British Columbia , Vancouver, BC , Canada ; Centre for Blood Research, University of British Columbia , Vancouver, BC , Canada ; The Brain Research Centre, University of British Columbia , Vancouver, BC , Canada ; Department of Microbiology and Immunology, University of British Columbia , Vancouver, BC , Canada ; Department of Medical Genetics, University of British Columbia , Vancouver, BC , Canada ; Department of Zoology, University of British Columbia , Vancouver, BC , Canada
| | | | | | | |
Collapse
|
41
|
Harada M, Luo X, Qi XY, Tadevosyan A, Maguy A, Ordog B, Ledoux J, Kato T, Naud P, Voigt N, Shi Y, Kamiya K, Murohara T, Kodama I, Tardif JC, Schotten U, Van Wagoner DR, Dobrev D, Nattel S. Transient receptor potential canonical-3 channel-dependent fibroblast regulation in atrial fibrillation. Circulation 2012; 126:2051-64. [PMID: 22992321 DOI: 10.1161/circulationaha.112.121830] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Fibroblast proliferation and differentiation are central in atrial fibrillation (AF)-promoting remodeling. Here, we investigated fibroblast regulation by Ca(2+)-permeable transient receptor potential canonical-3 (TRPC3) channels. METHODS AND RESULTS Freshly isolated rat cardiac fibroblasts abundantly expressed TRPC3 and had appreciable nonselective cation currents (I(NSC)) sensitive to a selective TPRC3 channel blocker, pyrazole-3 (3 μmol/L). Pyrazole-3 suppressed angiotensin II-induced Ca(2+) influx, proliferation, and α-smooth muscle actin protein expression in fibroblasts. Ca(2+) removal and TRPC3 blockade suppressed extracellular signal-regulated kinase phosphorylation, and extracellular signal-regulated kinase phosphorylation inhibition reduced fibroblast proliferation. TRPC3 expression was upregulated in atria from AF patients, goats with electrically maintained AF, and dogs with tachypacing-induced heart failure. TRPC3 knockdown (based on short hairpin RNA [shRNA]) decreased canine atrial fibroblast proliferation. In left atrial fibroblasts freshly isolated from dogs kept in AF for 1 week by atrial tachypacing, TRPC3 protein expression, currents, extracellular signal-regulated kinase phosphorylation, and extracellular matrix gene expression were all significantly increased. In cultured left atrial fibroblasts from AF dogs, proliferation rates, α-smooth muscle actin expression, and extracellular signal-regulated kinase phosphorylation were increased and were suppressed by pyrazole-3. MicroRNA-26 was downregulated in canine AF atria; experimental microRNA-26 knockdown reproduced AF-induced TRPC3 upregulation and fibroblast activation. MicroRNA-26 has NFAT (nuclear factor of activated T cells) binding sites in the 5' promoter region. NFAT activation increased in AF fibroblasts, and NFAT negatively regulated microRNA-26 transcription. In vivo pyrazole-3 administration suppressed AF while decreasing fibroblast proliferation and extracellular matrix gene expression. CONCLUSIONS TRPC3 channels regulate cardiac fibroblast proliferation and differentiation, likely by controlling the Ca(2+) influx that activates extracellular signal-regulated kinase signaling. AF increases TRPC3 channel expression by causing NFAT-mediated downregulation of microRNA-26 and causes TRPC3-dependent enhancement of fibroblast proliferation and differentiation. In vivo, TRPC3 blockade prevents AF substrate development in a dog model of electrically maintained AF. TRPC3 likely plays an important role in AF by promoting fibroblast pathophysiology and is a novel potential therapeutic target.
Collapse
Affiliation(s)
- Masahide Harada
- Department of Medicine and Research Center, Montreal Heart Institute and Universite´ de Montre´al, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Frühwald J, Camacho Londoño J, Dembla S, Mannebach S, Lis A, Drews A, Wissenbach U, Oberwinkler J, Philipp SE. Alternative splicing of a protein domain indispensable for function of transient receptor potential melastatin 3 (TRPM3) ion channels. J Biol Chem 2012; 287:36663-72. [PMID: 22961981 DOI: 10.1074/jbc.m112.396663] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TRPM3 channels form ionotropic steroid receptors in the plasma membrane of pancreatic β and dorsal root ganglion cells and link steroid hormone signaling to insulin release and pain perception, respectively. We identified and compared the function of a number of TRPM3 splice variants present in mouse, rat and human tissues. We found that variants lacking a region of 18 amino acid residues display neither Ca(2+) entry nor ionic currents when expressed alone. Hence, splicing removes a region that is indispensable for channel function, which is called the ICF region. TRPM3 variants devoid of this region (TRPM3ΔICF), are ubiquitously present in different tissues and cell types where their transcripts constitute up to 15% of the TRPM3 isoforms. The ICF region is conserved throughout the TRPM family, and its presence in TRPM8 proteins is also necessary for function. Within the ICF region, 10 amino acid residues form a domain essential for the formation of operative TRPM3 channels. TRPM3ΔICF variants showed reduced interaction with other TRPM3 isoforms, and their occurrence at the cell membrane was diminished. Correspondingly, coexpression of ΔICF proteins with functional TRPM3 subunits not only reduced the number of channels but also impaired TRPM3-mediated Ca(2+) entry. We conclude that TRPM3ΔICF variants are regulatory channel subunits fine-tuning TRPM3 channel activity.
Collapse
Affiliation(s)
- Julia Frühwald
- From the Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Differential pathways for calcium influx activated by concanavalin A and CD3 stimulation in Jurkat T cells. Pflugers Arch 2011; 463:309-18. [DOI: 10.1007/s00424-011-1039-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 10/03/2011] [Accepted: 10/05/2011] [Indexed: 01/31/2023]
|
44
|
Bogeski I, Gulaboski R, Kappl R, Mirceski V, Stefova M, Petreska J, Hoth M. Calcium binding and transport by coenzyme Q. J Am Chem Soc 2011; 133:9293-303. [PMID: 21548646 DOI: 10.1021/ja110190t] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Coenzyme Q10 (CoQ10) is one of the essential components of the mitochondrial electron-transport chain (ETC) with the primary function to transfer electrons along and protons across the inner mitochondrial membrane (IMM). The concomitant proton gradient across the IMM is essential for the process of oxidative phosphorylation and consequently ATP production. Cytochrome P450 (CYP450) monoxygenase enzymes are known to induce structural changes in a variety of compounds and are expressed in the IMM. However, it is unknown if CYP450 interacts with CoQ10 and how such an interaction would affect mitochondrial function. Using voltammetry, UV-vis spectrometry, electron paramagnetic resonance (EPR), nuclear magnetic resonance (NMR), fluorescence microscopy and high performance liquid chromatography-mass spectrometry (HPLC-MS), we show that both CoQ10 and its analogue CoQ1, when exposed to CYP450 or alkaline media, undergo structural changes through a complex reaction pathway and form quinone structures with distinct properties. Hereby, one or both methoxy groups at positions 2 and 3 on the quinone ring are replaced by hydroxyl groups in a time-dependent manner. In comparison with the native forms, the electrochemically reduced forms of the new hydroxylated CoQs have higher antioxidative potential and are also now able to bind and transport Ca(2+) across artificial biomimetic membranes. Our results open new perspectives on the physiological importance of CoQ10 and its analogues, not only as electron and proton transporters, but also as potential regulators of mitochondrial Ca(2+) and redox homeostasis.
Collapse
Affiliation(s)
- Ivan Bogeski
- Department of Biophysics, School of Medicine, Saarland University, 66421 Homburg, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Wenning AS, Neblung K, Strauß B, Wolfs MJ, Sappok A, Hoth M, Schwarz EC. TRP expression pattern and the functional importance of TRPC3 in primary human T-cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:412-23. [DOI: 10.1016/j.bbamcr.2010.12.022] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 12/22/2010] [Accepted: 12/23/2010] [Indexed: 11/16/2022]
|
46
|
Lambert S, Drews A, Rizun O, Wagner TFJ, Lis A, Mannebach S, Plant S, Portz M, Meissner M, Philipp SE, Oberwinkler J. Transient receptor potential melastatin 1 (TRPM1) is an ion-conducting plasma membrane channel inhibited by zinc ions. J Biol Chem 2011; 286:12221-33. [PMID: 21278253 DOI: 10.1074/jbc.m110.202945] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
TRPM1 is the founding member of the melastatin subgroup of transient receptor potential (TRP) proteins, but it has not yet been firmly established that TRPM1 proteins form ion channels. Consequently, the biophysical and pharmacological properties of these proteins are largely unknown. Here we show that heterologous expression of TRPM1 proteins induces ionic conductances that can be activated by extracellular steroid application. However the current amplitudes observed were too small to enable a reliable biophysical characterization. We overcame this limitation by modifying TRPM1 channels in several independent ways that increased the similarity to the closely related TRPM3 channels. The resulting constructs produced considerably larger currents after overexpression. We also demonstrate that unmodified TRPM1 and TRPM3 proteins form functional heteromultimeric channels. With these approaches, we measured the divalent permeability profile and found that channels containing the pore of TRPM1 are inhibited by extracellular zinc ions at physiological concentrations, in contrast to channels containing only the pore of TRPM3. Applying these findings to pancreatic β cells, we found that TRPM1 proteins do not play a major role in steroid-activated currents of these cells. The inhibition of TRPM1 by zinc ions is primarily due to a short stretch of seven amino acids present only in the pore region of TRPM1 but not of TRPM3. Combined, our data demonstrate that TRPM1 proteins are bona fide ion-conducting plasma membrane channels. Their distinct biophysical properties allow a reliable identification of endogenous TRPM1-mediated currents.
Collapse
Affiliation(s)
- Sachar Lambert
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Woo JS, Cho CH, Kim DH, Lee EH. TRPC3 cation channel plays an important role in proliferation and differentiation of skeletal muscle myoblasts. Exp Mol Med 2011; 42:614-27. [PMID: 20644344 DOI: 10.3858/emm.2010.42.9.061] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
During membrane depolarization associated with skeletal excitation-contraction (EC) coupling, dihydropyridine receptor [DHPR, a L-type Ca(2+) channel in the transverse (t)-tubule membrane] undergoes conformational changes that are transmitted to ryanodine receptor 1 [RyR1, an internal Ca(2+)-release channel in the sarcoplasmic reticulum (SR) membrane] causing Ca(2+) release from the SR. Canonical-type transient receptor potential cation channel 3 (TRPC3), an extracellular Ca(2+)-entry channel in the t-tubule and plasma membrane, is required for full-gain of skeletal EC coupling. To examine additional role(s) for TRPC3 in skeletal muscle other than mediation of EC coupling, in the present study, we created a stable myoblast line with reduced TRPC3 expression and without alpha1((S))DHPR (MDG/TRPC3 KD myoblast) by knock-down of TRPC3 in alpha1((S))DHPR-null muscular dysgenic (MDG) myoblasts using retrovirus-delivered small interference RNAs in order to eliminate any DHPR-associated EC coupling-related events. Unlike wild-type or alpha1((S))DHPR-null MDG myoblasts, MDG/TRPC3 KD myoblasts exhibited dramatic changes in cellular morphology (e.g., unusual expansion of both cell volume and the plasma membrane, and multi-nuclei) and failed to differentiate into myotubes possibly due to increased Ca(2+) content in the SR. These results suggest that TRPC3 plays an important role in the maintenance of skeletal muscle myoblasts and myotubes.
Collapse
Affiliation(s)
- Jin Seok Woo
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | |
Collapse
|
48
|
Kumar PG, Shoeb M. The Role of TRP Ion Channels in Testicular Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:881-908. [DOI: 10.1007/978-94-007-0265-3_46] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
Gees M, Colsoul B, Nilius B. The role of transient receptor potential cation channels in Ca2+ signaling. Cold Spring Harb Perspect Biol 2010; 2:a003962. [PMID: 20861159 DOI: 10.1101/cshperspect.a003962] [Citation(s) in RCA: 310] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The 28 mammalian members of the super-family of transient receptor potential (TRP) channels are cation channels, mostly permeable to both monovalent and divalent cations, and can be subdivided into six main subfamilies: the TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin), TRPML (mucolipin), and the TRPA (ankyrin) groups. TRP channels are widely expressed in a large number of different tissues and cell types, and their biological roles appear to be equally diverse. In general, considered as polymodal cell sensors, they play a much more diverse role than anticipated. Functionally, TRP channels, when activated, cause cell depolarization, which may trigger a plethora of voltage-dependent ion channels. Upon stimulation, Ca2+ permeable TRP channels generate changes in the intracellular Ca2+ concentration, [Ca2+]i, by Ca2+ entry via the plasma membrane. However, more and more evidence is arising that TRP channels are also located in intracellular organelles and serve as intracellular Ca2+ release channels. This review focuses on three major tasks of TRP channels: (1) the function of TRP channels as Ca2+ entry channels; (2) the electrogenic actions of TRPs; and (3) TRPs as Ca2+ release channels in intracellular organelles.
Collapse
Affiliation(s)
- Maarten Gees
- KU Leuven, Department of Molecular Cell Biology, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, bus 802, Leuven, Belgium
| | | | | |
Collapse
|
50
|
Hogan PG, Lewis RS, Rao A. Molecular basis of calcium signaling in lymphocytes: STIM and ORAI. Annu Rev Immunol 2010; 28:491-533. [PMID: 20307213 DOI: 10.1146/annurev.immunol.021908.132550] [Citation(s) in RCA: 601] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ca(2+) entry into cells of the peripheral immune system occurs through highly Ca(2+)-selective channels known as CRAC (calcium release-activated calcium) channels. CRAC channels are a very well-characterized example of store-operated Ca(2+) channels, so designated because they open when the endoplasmic reticulum (ER) Ca(2+) store becomes depleted. Physiologically, Ca(2+) is released from the ER lumen into the cytoplasm when activated receptors couple to phospholipase C and trigger production of the second messenger inositol 1,4,5-trisphosphate (IP(3)). IP(3) binds to IP(3) receptors in the ER membrane and activates Ca(2+) release. The proteins STIM and ORAI were discovered through limited and genome-wide RNAi screens, respectively, performed in Drosophila cells and focused on identifying modulators of store-operated Ca(2+) entry. STIM1 and STIM2 sense the depletion of ER Ca(2+) stores, whereas ORAI1 is a pore subunit of the CRAC channel. In this review, we discuss selected aspects of Ca(2+) signaling in cells of the immune system, focusing on the roles of STIM and ORAI proteins in store-operated Ca(2+) entry.
Collapse
Affiliation(s)
- Patrick G Hogan
- Department of Pathology, Harvard Medical School, Immune Disease Institute, Children's Hospital Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|