1
|
Rubaiy HN. Transient Receptor Potential Canonical Channels in Cardiovascular Pathology and Their Modulators. J Cardiovasc Pharmacol 2025; 85:21-34. [PMID: 39405561 DOI: 10.1097/fjc.0000000000001643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/05/2024] [Indexed: 01/18/2025]
Abstract
ABSTRACT Ion channels play a crucial role in various aspects of cardiac function, such as regulating rhythm and contractility. As a result, they serve as key targets for therapeutic interventions in cardiovascular diseases. Cell function is substantially influenced by the concentration of free cytosolic calcium (Ca 2+ ) and the voltage across the plasma membrane. These characteristics are known to be regulated by Ca 2+ -permeable nonselective cationic channels, although our knowledge of these channels is still inadequate. The transient receptor potential (TRP) superfamily comprises of many nonselective cation channels with diverse Ca 2+ permeability. Canonical or classical TRP (TRPC) channels are a subgroup of the TRP superfamily that are expressed ubiquitously in mammalian cells. TRPC channels are multidimensional signaling protein complexes that play essential roles in a variety of physiological and pathological processes in humans, including cancer, neurological disorders, cardiovascular diseases, and others. The objective of this article was to focus on the role that TRPC channels play in the cardiovascular system. The role of TRPC channels will be deeply discussed in cardiovascular pathology. Together, a critical element in developing novel treatments that target TRPC channels is comprehending the molecular mechanisms and regulatory pathways of TRPC channels in related cardiovascular diseases and conditions.
Collapse
Affiliation(s)
- Hussein N Rubaiy
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
2
|
Groschner K. TRPC3: how current mechanistic understanding provides a basis for therapeutic targeting. Expert Opin Ther Targets 2024; 28:953-961. [PMID: 39543956 DOI: 10.1080/14728222.2024.2430520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/21/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Intensive and detailed investigations of the molecular function and cellular role of mammalian transient receptor potential canonical (TRPC) channels started back in the early 90th of the past century. Initial TRPC research was fueled by high hopes to resolve fundamental questions of cellular Ca2+ signaling. To date, we have learned important lessons in TRPC channel biology and biophysics, while little progress has been made in terms of therapeutic concepts. AREAS COVERED This is a critical account of recent progress in building a solid mechanistic basis for therapeutic interventions utilizing TRPC3 as a target. I focus on hurdles and key issues to be resolved, thereby drafting a list of essential scientific 'to-dos' on the way toward drugging of TRPC3. EXPERT OPINION Recent advances in the molecular physiology of TRPC3 include unveiling its lipid sensing machinery and the channels´ ability to serve spatiotemporally diverse Ca2+ signaling in a cell type- and context-dependent manner. The ongoing development of technology for high-precision manipulation of the channel opens up a view on novel therapeutic strategies. It is now to unravel the complex role of TRPC3 in human physiopathology, to pinpoint the channels´ therapeutic relevance, and to further refine technologies for targeted interventions.
Collapse
Affiliation(s)
- Klaus Groschner
- Gottfried Schatz Research Center, Medical Physics and Biophysics, Medical University of Graz, Graz, Austria
| |
Collapse
|
3
|
Chen J, Liu T, Cui H, Na Q, Liu S. MiRNA-26a-5p inhibits preterm labor initiation by targeting and regulating TRPC3 ion channel protein expression. ENVIRONMENTAL TOXICOLOGY 2024; 39:357-366. [PMID: 37755144 DOI: 10.1002/tox.23972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/03/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023]
Abstract
The incidence of preterm birth (PTB) is increasing annually worldwide, leading to various health problems or even fetal deaths. Our previous work demonstrated the activation of transient receptor potential cation channel subfamily C 3 (TRPC3) in mice with PTB, and its activation could promote inward flow of calcium ions and uterine smooth muscle (USM) contraction via regulation of Cav3.2, Cav3.1, and Cav1.2. However, the upstream regulators of TRPC3 and its mechanisms remain unknown. In the present study, the binding of miR-26a-5p to the 3' untranslated region of TRPC3 was predicted by bioinformatics databases (TargetScanHuman and starBase v3.0) and confirmed by a dual-luciferase assay. MiR-26a-5p was downregulated, while TRPC3 was upregulated in the USM tissues of patients with PTB compared to people without PTB. The results showed that miR-26a-5p mimic transfection markedly reduced TRPC3 expression in LPS-stimulated USM cells. Additionally, miR-26a-5p regulated intracellular Ca2+ levels in USM cells by targeting TRPC3. Furthermore, miR-26a-5p inhibited the CPI17/PKC/PLCγ signaling pathway and reduced the expression of Cav3.2, Cav3.1, and Cav1.2. In conclusion, miR-26a-5p regulated the initiation of PTB by targeting TRPC3 and regulating intracellular Ca2+ levels. This study provides a promising diagnostic biomarker and therapeutic target for PTB.
Collapse
Affiliation(s)
- Jing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Tong Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Hong Cui
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Quan Na
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Sishi Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
4
|
Savadipour A, Nims RJ, Katz DB, Guilak F. Regulation of chondrocyte biosynthetic activity by dynamic hydrostatic pressure: the role of TRP channels. Connect Tissue Res 2022; 63:69-81. [PMID: 33494617 PMCID: PMC10061443 DOI: 10.1080/03008207.2020.1871475] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Chondrocytes perceive and respond to mechanical loading as signals that regulate their metabolism. Joint loading exposes chondrocytes to multiple modes of mechanical stress, including hydrostatic pressure; however, the mechanisms by which chondrocytes sense physiologically relevant levels of hydrostatic pressure are not well understood. We hypothesized that hydrostatic pressure is transduced to an intracellular signal through mechanosensitive membrane ion channels of chondrocytes. The goals of this study were to examine the effect of hydrostatic loading on the development of engineered cartilage tissue and the contribution of mechanosensitive ion channels on these hydrostatic loading effects. METHODS Using a 3D model of porcine chondrocytes in agarose, we applied specific chemical inhibitors to determine the role of transient receptor potential (TRP) ion channels TRPV1, TRPV4, TRPC3, and TRPC1 in transducing hydrostatic pressure. RESULTS Hydrostatic loading caused a frequency and magnitude-dependent decrease in sulfated glycosaminoglycans (S-GAG), without changes in DNA content. Inhibiting TRPC3 and TRPV4 decreased S-GAG content; however, only the inhibition of TRPV1 partially attenuated the hydrostatic loading-induced reduction in S-GAG content. CONCLUSIONS Our findings indicate that TRPV1 may serve as a transducer of hydrostatic pressure in chondrocytes, and provide further support for the role of TRPV4 in regulating chondrocyte anabolism, as well as initial evidence implicating TRPC3 in chondrogenesis. These findings add to our further understanding of the chondrocyte "channelome" and suggest that a range of ion channels mediate the transduction of different biophysical stimuli such as hydrostatic pressure, membrane stretch, or osmotic stress.
Collapse
Affiliation(s)
- Alireza Savadipour
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, USA
| | - Robert J Nims
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dakota B Katz
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| |
Collapse
|
5
|
Yang PL, Li XH, Wang J, Ma XF, Zhou BY, Jiao YF, Wang WH, Cao P, Zhu MX, Li PW, Xiao ZH, Li CZ, Guo CR, Lei YT, Yu Y. GSK1702934A and M085 directly activate TRPC6 via a mechanism of stimulating the extracellular cavity formed by the pore helix and transmembrane helix S6. J Biol Chem 2021; 297:101125. [PMID: 34461094 PMCID: PMC8458982 DOI: 10.1016/j.jbc.2021.101125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/22/2021] [Accepted: 08/26/2021] [Indexed: 01/20/2023] Open
Abstract
Transient receptor potential canonical (TRPC) channels, as important membrane proteins regulating intracellular calcium (Ca2+i) signaling, are involved in a variety of physiological and pathological processes. Activation and regulation of TRPC are more dependent on membrane or intracellular signals. However, how extracellular signals regulate TRPC6 function remains to be further investigated. Here, we suggest that two distinct small molecules, M085 and GSK1702934A, directly activate TRPC6, both through a mechanism of stimulation of extracellular sites formed by the pore helix (PH) and transmembrane (TM) helix S6. In silico docking scanning of TRPC6 identified three extracellular sites that can bind small molecules, of which only mutations on residues of PH and S6 helix significantly reduced the apparent affinity of M085 and GSK1702934A and attenuated the maximal response of TRPC6 to these two chemicals by altering channel gating of TRPC6. Combing metadynamics, molecular dynamics simulations, and mutagenesis, we revealed that W679, E671, E672, and K675 in the PH and N701 and Y704 in the S6 helix constitute an orthosteric site for the recognition of these two agonists. The importance of this site was further confirmed by covalent modification of amino acid residing at the interface of the PH and S6 helix. Given that three structurally distinct agonists M085, GSK1702934A, and AM-0883, act at this site, as well as the occupancy of lipid molecules at this position found in other TRP subfamilies, it is suggested that the cavity formed by the PH and S6 has an important role in the regulation of TRP channel function by extracellular signals.
Collapse
Affiliation(s)
- Pei-Lin Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xing-Hua Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jin Wang
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xue-Fei Ma
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Bo-Ying Zhou
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuan-Feng Jiao
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wen-Hui Wang
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Peng Cao
- Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Michael Xi Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Pei-Wang Li
- State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Changsha, China
| | - Zhi-Hong Xiao
- State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Changsha, China
| | - Chang-Zhu Li
- State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Changsha, China
| | - Chang-Run Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Yun-Tao Lei
- School of Science, China Pharmaceutical University, Nanjing, China.
| | - Ye Yu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
6
|
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: 0.8] [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
|
7
|
Lu M, Fang XX, Shi DD, Liu R, Ding Y, Zhang QF, Wang HQ, Tang JM, He XJ. A Selective TRPC3 Inhibitor Pyr3 Attenuates Myocardial Ischemia/Reperfusion Injury in Mice. Curr Med Sci 2021; 40:1107-1113. [PMID: 33428139 DOI: 10.1007/s11596-020-2293-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/21/2020] [Indexed: 11/29/2022]
Abstract
An emerging body of evidence indicates that transient receptor potential TRP channels act as important mediators for a wide variety of physiological functions and are potential targets for drug discovery. Our previous study has identified transient receptor potential channel 3 (TRPC3) and TRPC6 as cation channels through which most of the damaging calcium enters, aggravates pathological changes in vivo and increases ischemia/reperfusion (I/R) injury in mice. This study aimed to verify the effects of TRPC3 inhibitor Pyr3 on myocardial I/R injury in mice. C57BL/6J wild-type male mice (8 to 12 weeks old) were anesthetized with 3.3% chloral hydrate. A murine I (30 min)/R (24 h) injury model was established by temporary occlusion of the left anterior descending (LAD) coronary artery. Pyr3 was administered at concentrations of 0, 2.5, 5, or 10 mg/kg via the right jugular vein 5 min before reperfusion. We observed that the selective TRPC3 inhibitor, 10 mg/kg Pyr3, significantly decreased the infarct size of left ventricle, and reduced the myocardial cell apoptosis rate and inflammatory response in mice. In a conclusion, TRPC3 can function as a candidate target for I/R injury prevention, and Pyr3 may directly bind to TRPC3 channel protein, inhibit TRPC3 channel activity, and improve TRPC3-related myocardial I/R injury. Pyr3 may be used for clarification of TRPC3 functions and for treatments of TRPC3-mediated diseases.
Collapse
Affiliation(s)
- Min Lu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Xiao-Xia Fang
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Dan-Dan Shi
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Rui Liu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Qiu-Fang Zhang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Han-Qin Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Jun-Ming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Xi-Ju He
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China. .,Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| |
Collapse
|
8
|
Chen X, Sooch G, Demaree IS, White FA, Obukhov AG. Transient Receptor Potential Canonical (TRPC) Channels: Then and Now. Cells 2020; 9:E1983. [PMID: 32872338 PMCID: PMC7565274 DOI: 10.3390/cells9091983] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Twenty-five years ago, the first mammalian Transient Receptor Potential Canonical (TRPC) channel was cloned, opening the vast horizon of the TRPC field. Today, we know that there are seven TRPC channels (TRPC1-7). TRPCs exhibit the highest protein sequence similarity to the Drosophila melanogaster TRP channels. Similar to Drosophila TRPs, TRPCs are localized to the plasma membrane and are activated in a G-protein-coupled receptor-phospholipase C-dependent manner. TRPCs may also be stimulated in a store-operated manner, via receptor tyrosine kinases, or by lysophospholipids, hypoosmotic solutions, and mechanical stimuli. Activated TRPCs allow the influx of Ca2+ and monovalent alkali cations into the cytosol of cells, leading to cell depolarization and rising intracellular Ca2+ concentration. TRPCs are involved in the continually growing number of cell functions. Furthermore, mutations in the TRPC6 gene are associated with hereditary diseases, such as focal segmental glomerulosclerosis. The most important recent breakthrough in TRPC research was the solving of cryo-EM structures of TRPC3, TRPC4, TRPC5, and TRPC6. These structural data shed light on the molecular mechanisms underlying TRPCs' functional properties and propelled the development of new modulators of the channels. This review provides a historical overview of the major advances in the TRPC field focusing on the role of gene knockouts and pharmacological tools.
Collapse
Affiliation(s)
- Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Gagandeep Sooch
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
| | - Isaac S. Demaree
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
| | - Fletcher A. White
- The Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G. Obukhov
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
TRPC and TRPV Channels' Role in Vascular Remodeling and Disease. Int J Mol Sci 2020; 21:ijms21176125. [PMID: 32854408 PMCID: PMC7503586 DOI: 10.3390/ijms21176125] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/19/2020] [Accepted: 08/23/2020] [Indexed: 12/15/2022] Open
Abstract
Transient receptor potentials (TRPs) are non-selective cation channels that are widely expressed in vascular beds. They contribute to the Ca2+ influx evoked by a wide spectrum of chemical and physical stimuli, both in endothelial and vascular smooth muscle cells. Within the superfamily of TRP channels, different isoforms of TRPC (canonical) and TRPV (vanilloid) have emerged as important regulators of vascular tone and blood flow pressure. Additionally, several lines of evidence derived from animal models, and even from human subjects, highlighted the role of TRPC and TRPV in vascular remodeling and disease. Dysregulation in the function and/or expression of TRPC and TRPV isoforms likely regulates vascular smooth muscle cells switching from a contractile to a synthetic phenotype. This process contributes to the development and progression of vascular disorders, such as systemic and pulmonary arterial hypertension, atherosclerosis and restenosis. In this review, we provide an overview of the current knowledge on the implication of TRPC and TRPV in the physiological and pathological processes of some frequent vascular diseases.
Collapse
|
10
|
Graziani A, Bacsa B, Krivic D, Wiedner P, Curcic S, Schindl R, Tiapko O, Groschner K. Light-Mediated Control over TRPC3-Mediated NFAT Signaling. Cells 2020; 9:E556. [PMID: 32120825 PMCID: PMC7140526 DOI: 10.3390/cells9030556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 01/20/2023] Open
Abstract
Canonical transient receptor potential (TRPC) channels were identified as key players in maladaptive remodeling, with nuclear factor of activated T-cells (NFAT) transcription factors serving as downstream targets of TRPC-triggered Ca2+ entry in these pathological processes. Strikingly, the reconstitution of TRPC-NFAT signaling by heterologous expression yielded controversial results. Specifically, nuclear translocation of NFAT1 was found barely responsive to recombinant TRPC3, presumably based on the requirement of certain spatiotemporal signaling features. Here, we report efficient control of NFAT1 nuclear translocation in human embryonic kidney 293 (HEK293) cells by light, using a new photochromic TRPC benzimidazole activator (OptoBI-1) and a TRPC3 mutant with modified activator sensitivity. NFAT1 nuclear translocation was measured along with an all-optical protocol to record local and global Ca2+ pattern generated during light-mediated activation/deactivation cycling of TRPC3. Our results unveil the ability of wild-type TRPC3 to produce constitutive NFAT nuclear translocation. Moreover, we demonstrate that TRPC3 mutant that lacks basal activity enables spatiotemporally precise control over NFAT1 activity by photopharmacology. Our results suggest tight linkage between TRPC3 activity and NFAT1 nuclear translocation based on global cellular Ca2+ signals.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Oleksandra Tiapko
- Gottfried-Schatz-Research-Center – Biophysics; Medical University of Graz, Neue Stiftingtalstrasse 6/D04, 8010 Graz, Austria; (A.G.); (B.B.); (D.K.); (P.W.); (S.C.); (R.S.)
| | - Klaus Groschner
- Gottfried-Schatz-Research-Center – Biophysics; Medical University of Graz, Neue Stiftingtalstrasse 6/D04, 8010 Graz, Austria; (A.G.); (B.B.); (D.K.); (P.W.); (S.C.); (R.S.)
| |
Collapse
|
11
|
Wang H, Cheng X, Tian J, Xiao Y, Tian T, Xu F, Hong X, Zhu MX. TRPC channels: Structure, function, regulation and recent advances in small molecular probes. Pharmacol Ther 2020; 209:107497. [PMID: 32004513 DOI: 10.1016/j.pharmthera.2020.107497] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/14/2020] [Indexed: 02/08/2023]
Abstract
Transient receptor potential canonical (TRPC) channels constitute a group of receptor-operated calcium-permeable nonselective cation channels of the TRP superfamily. The seven mammalian TRPC members, which can be further divided into four subgroups (TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7) based on their amino acid sequences and functional similarities, contribute to a broad spectrum of cellular functions and physiological roles. Studies have revealed complexity of their regulation involving several components of the phospholipase C pathway, Gi and Go proteins, and internal Ca2+ stores. Recent advances in cryogenic electron microscopy have provided several high-resolution structures of TRPC channels. Growing evidence demonstrates the involvement of TRPC channels in diseases, particularly the link between genetic mutations of TRPC6 and familial focal segmental glomerulosclerosis. Because TRPCs were discovered by the molecular identity first, their pharmacology had lagged behind. This is rapidly changing in recent years owning to great efforts from both academia and industry. A number of potent tool compounds from both synthetic and natural products that selective target different subtypes of TRPC channels have been discovered, including some preclinical drug candidates. This review will cover recent advancements in the understanding of TRPC channel regulation, structure, and discovery of novel TRPC small molecular probes over the past few years, with the goal of facilitating drug discovery for the study of TRPCs and therapeutic development.
Collapse
Affiliation(s)
- Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaoding Cheng
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yuling Xiao
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Tian Tian
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Fuchun Xu
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Xuechuan Hong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China; Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China.
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Lambert M, Capuano V, Olschewski A, Sabourin J, Nagaraj C, Girerd B, Weatherald J, Humbert M, Antigny F. Ion Channels in Pulmonary Hypertension: A Therapeutic Interest? Int J Mol Sci 2018; 19:ijms19103162. [PMID: 30322215 PMCID: PMC6214085 DOI: 10.3390/ijms19103162] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial and severe disease without curative therapies. PAH pathobiology involves altered pulmonary arterial tone, endothelial dysfunction, distal pulmonary vessel remodeling, and inflammation, which could all depend on ion channel activities (K⁺, Ca2+, Na⁺ and Cl-). This review focuses on ion channels in the pulmonary vasculature and discusses their pathophysiological contribution to PAH as well as their therapeutic potential in PAH.
Collapse
Affiliation(s)
- Mélanie Lambert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Véronique Capuano
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
- Department of Physiology, Medical University Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria.
| | - Jessica Sabourin
- Signalisation et Physiopathologie Cardiovasculaire, UMRS 1180, Univ. Paris-Sud, INSERM, Université Paris-Saclay, 92296 Châtenay-Malabry, France.
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
| | - Barbara Girerd
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Jason Weatherald
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
- Division of Respirology, Department of Medicine, University of Calgary, Calgary, AB T1Y 6J4, Canada.
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB T1Y 6J4, Canada.
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Fabrice Antigny
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| |
Collapse
|
13
|
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: 42] [Impact Index Per Article: 6.0] [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
|
14
|
Xiao X, Liu HX, Shen K, Cao W, Li XQ. Canonical Transient Receptor Potential Channels and Their Link with Cardio/Cerebro-Vascular Diseases. Biomol Ther (Seoul) 2017; 25:471-481. [PMID: 28274093 PMCID: PMC5590790 DOI: 10.4062/biomolther.2016.096] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 12/04/2016] [Accepted: 12/27/2016] [Indexed: 12/29/2022] Open
Abstract
The canonical transient receptor potential channels (TRPCs) constitute a series of nonselective cation channels with variable degrees of Ca2+ selectivity. TRPCs consist of seven mammalian members, TRPC1, TRPC2, TRPC3, TRPC4, TRPC5, TRPC6, and TRPC7, which are further divided into four subtypes, TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7. These channels take charge of various essential cell functions such as contraction, relaxation, proliferation, and dysfunction. This review, organized into seven main sections, will provide an overview of current knowledge about the underlying pathogenesis of TRPCs in cardio/cerebrovascular diseases, including hypertension, pulmonary arterial hypertension, cardiac hypertrophy, atherosclerosis, arrhythmia, and cerebrovascular ischemia reperfusion injury. Collectively, TRPCs could become a group of drug targets with important physiological functions for the therapy of human cardio/cerebro-vascular diseases.
Collapse
Affiliation(s)
- Xiong Xiao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Hui-Xia Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.,Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Kuo Shen
- Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Cao
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
15
|
Soni H, Peixoto-Neves D, Buddington RK, Adebiyi A. Adenosine A 1 receptor-operated calcium entry in renal afferent arterioles is dependent on postnatal maturation of TRPC3 channels. Am J Physiol Renal Physiol 2017; 313:F1216-F1222. [PMID: 28855189 DOI: 10.1152/ajprenal.00335.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/14/2017] [Accepted: 08/22/2017] [Indexed: 12/13/2022] Open
Abstract
Adenosine, a regulator of cardiovascular development and renal function, constricts renal afferent arterioles by inducing intracellular Ca2+ concentration ([Ca2+]i) elevation in smooth muscle cells (SMCs) via activation of its cognate A1 receptors (A1Rs). Mechanisms that underlie A1R-dependent [Ca2+]i elevation in renal vascular SMCs are not fully resolved. Whether A1R expression and function in preglomerular microvessels are dependent on postnatal kidney maturation is also unclear. In this study, we show that selective activation of A1Rs by 2-chloro-N6-cyclopentyladenosine (CCPA) does not stimulate store-operated Ca2+ entry in afferent arterioles isolated from neonatal pigs. However, CCPA-induced [Ca2+]i elevation is dependent on phospholipase C and transient receptor potential cation channel, subfamily C, member 3 (TRPC3). Basal [Ca2+]i was unchanged in afferent arterioles isolated from newborn (0-day-old) pigs compared with their 20-day-old counterparts. By contrast, CCPA treatment resulted in significantly larger [Ca2+]i in afferent arterioles from 20-day-old pigs. A1R protein expression levels in the kidneys and afferent arterioles were unaltered in 0- vs. 20-day-old pigs. However, the TRPC3 channel protein expression level was ~92 and 78% higher in 20-day-old pig kidneys and afferent arterioles, respectively. These data suggest that activation of A1Rs elicits receptor-operated Ca2+ entry in porcine afferent arterioles, the level of which is dependent on postnatal maturation of TRPC3 channels. We propose that TRPC3 channels may contribute to the physiology and pathophysiology of A1Rs.
Collapse
Affiliation(s)
- Hitesh Soni
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee; and
| | - Dieniffer Peixoto-Neves
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee; and
| | - Randal K Buddington
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee; and.,School of Health Studies, University of Memphis, Memphis, Tennessee
| | - Adebowale Adebiyi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee; and
| |
Collapse
|
16
|
Alonso-Carbajo L, Kecskes M, Jacobs G, Pironet A, Syam N, Talavera K, Vennekens R. Muscling in on TRP channels in vascular smooth muscle cells and cardiomyocytes. Cell Calcium 2017; 66:48-61. [PMID: 28807149 DOI: 10.1016/j.ceca.2017.06.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023]
Abstract
The human TRP protein family comprises a family of 27 cation channels with diverse permeation and gating properties. The common theme is that they are very important regulators of intracellular Ca2+ signaling in diverse cell types, either by providing a Ca2+ influx pathway, or by depolarising the membrane potential, which on one hand triggers the activation of voltage-gated Ca2+ channels, and on the other limits the driving force for Ca2+ entry. Here we focus on the role of these TRP channels in vascular smooth muscle and cardiac striated muscle. We give an overview of highlights from the recent literature, and highlight the important and diverse roles of TRP channels in the pathophysiology of the cardiovascular system. The discovery of the superfamily of Transient Receptor Potential (TRP) channels has significantly enhanced our knowledge of multiple signal transduction mechanisms in cardiac muscle and vascular smooth muscle cells (VSMC). In recent years, multiple studies have provided evidence for the involvement of these channels, not only in the regulation of contraction, but also in cell proliferation and remodeling in pathological conditions. The mammalian family of TRP cation channels is composed by 28 genes which can be divided into 6 subfamilies groups based on sequence similarity: TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipins), TRPV (Vanilloid), TRPP (Policystin) and TRPA (Ankyrin-rich protein). Functional TRP channels are believed to form four-unit complexes in the plasma, each of them expressed with six transmembrane domain and intracellular N and C termini. Here we review the current knowledge on the expression of TRP channels in both muscle types, and discuss their functional properties and role in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Lucía Alonso-Carbajo
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Miklos Kecskes
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Griet Jacobs
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Andy Pironet
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Ninda Syam
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Karel Talavera
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
17
|
Groschner K, Shrestha N, Fameli N. Cardiovascular and Hemostatic Disorders: SOCE in Cardiovascular Cells: Emerging Targets for Therapeutic Intervention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:473-503. [PMID: 28900929 DOI: 10.1007/978-3-319-57732-6_24] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The discovery of the store-operated Ca2+ entry (SOCE) phenomenon is tightly associated with its recognition as a pathway of high (patho)physiological significance in the cardiovascular system. Early on, SOCE has been investigated primarily in non-excitable cell types, and the vascular endothelium received particular attention, while a role of SOCE in excitable cells, specifically cardiac myocytes and pacemakers, was initially ignored and remains largely enigmatic even to date. With the recent gain in knowledge on the molecular components of SOCE as well as their cellular organization within nanodomains, potential tissue/cell type-dependent heterogeneity of the SOCE machinery along with high specificity of linkage to downstream signaling pathways emerged for cardiovascular cells. The basis of precise decoding of cellular Ca2+ signals was recently uncovered to involve correct spatiotemporal organization of signaling components, and even minor disturbances in these assemblies trigger cardiovascular pathologies. With this chapter, we wish to provide an overview on current concepts of cellular organization of SOCE signaling complexes in cardiovascular cells with particular focus on the spatiotemporal aspects of coupling to downstream signaling and the potential disturbance of these mechanisms by pathogenic factors. The significance of these mechanistic concepts for the development of novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Klaus Groschner
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria.
| | - Niroj Shrestha
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria
| | - Nicola Fameli
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Álvarez-Miguel I, Cidad P, Pérez-García MT, López-López JR. Differences in TRPC3 and TRPC6 channels assembly in mesenteric vascular smooth muscle cells in essential hypertension. J Physiol 2016; 595:1497-1513. [PMID: 27861908 DOI: 10.1113/jp273327] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/07/2016] [Indexed: 01/29/2023] Open
Abstract
KEY POINTS Canonical transient receptor potential (TRPC)3 and TRPC6 channels of vascular smooth muscle cells (VSMCs) mediate stretch- or agonist-induced cationic fluxes, contributing to membrane potential and vascular tone. Native TRPC3/C6 channels can form homo- or heterotetrameric complexes, which can hinder individual TRPC channel properties. The possibility that the differences in their association pattern may change their contribution to vascular tone in hypertension is unexplored. Functional characterization of heterologously expressed channels showed that TRPC6-containing complexes exhibited Pyr3/Pyr10-sensitive currents, whereas TRPC3-mediated currents were blocked by anti-TRPC3 antibodies. VSMCs from hypertensive (blood pressure high; BPH) mice have larger cationic basal currents insensitive to Pyr10 and sensitive to anti-TRPC3 antibodies. Consistently, myography studies showed a larger Pyr3/10-induced vasodilatation in BPN (blood pressure normal) mesenteric arteries. We conclude that the increased TRPC3 channel expression in BPH VSMCs leads to changes in TRPC3/C6 heteromultimeric assembly, with a higher TRPC3 channel contribution favouring depolarization of hypertensive VSMCs. ABSTRACT Increased vascular tone in essential hypertension involves a sustained rise in total peripheral resistance. A model has been proposed in which the combination of membrane depolarization and higher L-type Ca2+ channel activity generates augmented Ca2+ influx into vascular smooth muscle cells (VSMCs), contraction and vasoconstriction. The search for culprit ion channels responsible for membrane depolarization has provided several candidates, including members of the canonical transient receptor potential (TRPC) family. TRPC3 and TRPC6 are diacylglycerol-activated, non-selective cationic channels contributing to stretch- or agonist-induced depolarization. Conflicting information exists regarding changes in TRPC3/TRPC6 functional expression in hypertension. However, although TRPC3-TRPC6 channels can heteromultimerize, the possibility that differences in their association pattern may change their functional contribution to vascular tone is largely unexplored. We probe this hypothesis using a model of essential hypertension (BPH mice; blood pressure high) and its normotensive control (BPN mice; blood pressure normal). First, non-selective cationic currents through homo- and heterotetramers recorded from transfected Chinese hamster ovary cells indicated that TRPC currents were sensitive to the selective antagonist Pyr10 only when TRPC6 was present, whereas intracellular anti-TRPC3 antibody selectively blocked TRPC3-mediated currents. In mesenteric VSMCs, basal and agonist-induced currents were more sensitive to Pyr3 and Pyr10 in BPN cells. Consistently, myography studies showed a larger Pyr3/10-induced vasodilatation in BPN mesenteric arteries. mRNA and protein expression data supported changes in TRPC3 and TRPC6 proportions and assembly, with a higher TRPC3 channel contribution in BPH VSMCs that could favour cell depolarization. These differences in functional and pharmacological properties of TRPC3 and TRPC6 channels, depending on their assembly, could represent novel therapeutical opportunities.
Collapse
Affiliation(s)
- Inés Álvarez-Miguel
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - M Teresa Pérez-García
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | - José Ramón López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología e Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| |
Collapse
|
19
|
Wang L, Li J, Zhang J, He Q, Weng X, Huang Y, Guan M, Qiu C. Inhibition of TRPC3 downregulates airway hyperresponsiveness, remodeling of OVA-sensitized mouse. Biochem Biophys Res Commun 2016; 484:209-217. [PMID: 28034747 DOI: 10.1016/j.bbrc.2016.12.138] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 12/21/2016] [Indexed: 12/31/2022]
Abstract
Airway hyperresponsiveness (AHR), airway remodeling and inflammation are the fundamental pathological alterations that occur in asthma. Transient receptor potential canonical 3 (TRPC3) has been implicated in diverse functions of airway smooth muscle cells (ASMCs) in asthma. However, the underlying mechanisms remain incompletely understood. We investigated the mRNA and protein expression of TRPC3 in ASMCs from normal and OVA-sensitized mouse. And the effects of inhibition or knockdown of TRPC3 with Ethyl-1- (4- (2,3,3-trichloroacrylamide) phenyl) -5 - (trifluoromethyl) -1H -pyrazole -4-carboxylate (Pyr3) and lentiviral shRNA on OVA-sensitized mouse AHR, airway remodeling, circulating inflammatory cytokines, cell proliferation and migration. We found that TRPC3 mRNA and protein expression levels were significantly increased in ASMCs from OVA-sensitized mouse. Inhibiting TRPC3 with continuous subcutaneous administration of Pyr3 decreased enhanced pause (Penh) of OVA-sensitized mouse. Meanwhile, both Pyr3 and lentiviral shRNA treatment of ASMCs in OVA-sensitized mouse significantly decreased their proliferation and migration. These results suggest that TRPC3 plays a critical role in asthma and represents a promising new target for asthma treatment.
Collapse
Affiliation(s)
- Lingwei Wang
- Department of Respiratory Diseases, Second Clinical medical college (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Jie Li
- Department of Respiratory Diseases, Second Clinical medical college (Shenzhen People's Hospital), Jinan University, Shenzhen, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Jian Zhang
- Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qi He
- Department of Respiratory Diseases, Second Clinical medical college (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Xuanwen Weng
- Department of Respiratory Diseases, Second Clinical medical college (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Yanmei Huang
- Department of Respiratory Diseases, Second Clinical medical college (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Minjie Guan
- Department of Respiratory Diseases, Second Clinical medical college (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Chen Qiu
- Department of Respiratory Diseases, Second Clinical medical college (Shenzhen People's Hospital), Jinan University, Shenzhen, China.
| |
Collapse
|
20
|
Chen XX, Zhang JH, Pan BH, Ren HL, Feng XL, Wang JL, Xiao JH. TRPC3-mediated Ca2+ entry contributes to mouse airway smooth muscle cell proliferation induced by lipopolysaccharide. Cell Calcium 2016; 60:273-81. [DOI: 10.1016/j.ceca.2016.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/17/2016] [Accepted: 06/18/2016] [Indexed: 01/03/2023]
|
21
|
Min SJ, Kang TC. Positive feedback role of TRPC3 in TNF-α-mediated vasogenic edema formation induced by status epilepticus independent of ET B receptor activation. Neuroscience 2016; 337:37-47. [PMID: 27623392 DOI: 10.1016/j.neuroscience.2016.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 08/26/2016] [Accepted: 09/05/2016] [Indexed: 02/06/2023]
Abstract
Brain-blood barrier (BBB) disruption results in vasogenic edema, which is involved in the pathogenesis of epilepsy. Following status epilepticus (SE), up-regulated transient receptor potential canonical channel-3 (TRPC3), a Ca2+-permeable cation channels in endothelial cells, is relevant to vasogenic edema formation in the rat piriform cortex. In addition, pyrazole-3 (Pyr-3, a TRPC3 inhibitor) attenuated SE-induced vasogenic edema. However, the upstream regulators of TRPC3 expression in vasogenic edema formation have been unclear. In the present study, soluble tumor necrosis factor p55 receptor (sTNFp55R, a TNF-α inhibitor), SN50 (a nuclear factor-κB (NFκB) inhibitor), BQ-788 (an endothelin B (ETB) receptor inhibitor) and Pyr-3 effectively prevented vasogenic edema following SE. sTNFp55R and SN50 (but not BQ-788) attenuated SE-induced up-regulation of endothelial TRPC3 expression. Pyr-3 ameliorated SE-induced NFκB p65-Thr435 phosphorylation and ETB receptor expression. In addition, Pyr-3 mitigated NFκB p65-Thr435 phosphorylation induced by recombinant TNF-α. These findings indicate that TNF-α-mediated NFκB p65-Thr435 phosphorylation may up-regulate TRPC3 expression, which participates in vasogenic edema formation via increasing endothelial nitric oxide synthase expression following SE, independent of ETB receptor activation. Therefore, we suggest that TRPC3 may be involved in a positive feedback loop of NFκB/ETB receptor signaling pathway.
Collapse
Affiliation(s)
- Su-Ji Min
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, South Korea.
| |
Collapse
|
22
|
Abdoul-Azize S, Buquet C, Vannier JP, Dubus I. Pyr3, a TRPC3 channel blocker, potentiates dexamethasone sensitivity and apoptosis in acute lymphoblastic leukemia cells by disturbing Ca(2+) signaling, mitochondrial membrane potential changes and reactive oxygen species production. Eur J Pharmacol 2016; 784:90-8. [PMID: 27179991 DOI: 10.1016/j.ejphar.2016.05.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 01/02/2023]
Abstract
Dexamethasone (Dex) is used as a chemotherapeutic drug in the treatment of acute lymphoblastic leukemia (ALL) because of its capacity to induce apoptosis. However, some ALL patients acquire resistance to glucocorticoids (GC). Thus, it is important to explore new agents to overcome GC resistance. The aim of the present work was to assess the ability of Pyr3, a selective inhibitor of transient receptor potential canonical 3 (TRPC3), to sensitize human ALL cells to Dex. We show here, for the first time, that Pyr3 enhances Dex sensitivity through the distraction of Dex-mediated Ca(2+) signaling in ALL cells (in vitro) and primary blasts (ex vivo) associated with mitochondrial-mediated reactive oxygen species production in ALL cells. Pyr3 alone induced Ca(2+) signaling via only endoplasmic reticulum-released Ca(2+) and exerted inhibitory effect on store-operated Ca(2+) entry in dose-dependent manner in ALL cell lines. Pre-incubation of cells with Pyr3 significantly curtailed the thapsigargin- and Dex-evoked Ca(2+) signaling in ALL cell lines. Pyr3 synergistically potentiated Dex lethality, as shown by the induction of cell mortality, G2/M cell cycle arrest and apoptosis in ALL cell lines. Moreover, Pyr3 disrupted Dex-mediated Ca(2+) signaling and increased the sensitivity of Dex-induced cell death in primary blasts from ALL patients. Additional analysis showed that co-treatment with Dex and Pyr3 results in mitochondrial membrane potential depolarization and reactive oxygen species production in ALL cells. Together, Pyr3 exhibited potential therapeutic benefit in combination with Dex to inverse glucocorticoid resistance in human ALL and probably in other lymphoid malignancies.
Collapse
Affiliation(s)
- Souleymane Abdoul-Azize
- Groupe de Recherche "Micro-Environnement et Renouvellement Cellulaire Intégré" MERCI UPRES EA 3829, Faculté de Médecine et Pharmacie, Université de Rouen, 76183 Rouen Cedex, France.
| | - Catherine Buquet
- Groupe de Recherche "Micro-Environnement et Renouvellement Cellulaire Intégré" MERCI UPRES EA 3829, Faculté de Médecine et Pharmacie, Université de Rouen, 76183 Rouen Cedex, France
| | - Jean-Pierre Vannier
- Groupe de Recherche "Micro-Environnement et Renouvellement Cellulaire Intégré" MERCI UPRES EA 3829, Faculté de Médecine et Pharmacie, Université de Rouen, 76183 Rouen Cedex, France; Service Immuno-Hémato-Oncologie Pédiatrique, CHU Charles Nicolle, 76031 Rouen Cedex, France
| | - Isabelle Dubus
- Groupe de Recherche "Micro-Environnement et Renouvellement Cellulaire Intégré" MERCI UPRES EA 3829, Faculté de Médecine et Pharmacie, Université de Rouen, 76183 Rouen Cedex, France
| |
Collapse
|
23
|
Hagimori M, Murakami T, Shimizu K, Nishida M, Ohshima T, Mukai T. Synthesis of radioiodinated probes to evaluate the biodistribution of a potent TRPC3 inhibitor. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00023a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The transient receptor potential canonical 3 (TRPC3) channel is a member of the TRPC family that contributes to the entry of Ca2+through the plasma membrane or modulates the driving force for Ca2+entry channels.
Collapse
Affiliation(s)
| | | | - Kinue Shimizu
- Graduate School of Pharmaceutical Sciences
- Kyushu University
- Fukuoka 812-8582
- Japan
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences
- Kyushu University
- Fukuoka 812-8582
- Japan
- Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences)
| | - Takashi Ohshima
- Graduate School of Pharmaceutical Sciences
- Kyushu University
- Fukuoka 812-8582
- Japan
| | | |
Collapse
|
24
|
Saliba Y, Karam R, Smayra V, Aftimos G, Abramowitz J, Birnbaumer L, Farès N. Evidence of a Role for Fibroblast Transient Receptor Potential Canonical 3 Ca2+ Channel in Renal Fibrosis. J Am Soc Nephrol 2015; 26:1855-76. [PMID: 25479966 PMCID: PMC4520158 DOI: 10.1681/asn.2014010065] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 09/23/2014] [Indexed: 01/04/2023] Open
Abstract
Transient receptor potential canonical (TRPC) Ca(2+)-permeant channels, especially TRPC3, are increasingly implicated in cardiorenal diseases. We studied the possible role of fibroblast TRPC3 in the development of renal fibrosis. In vitro, a macromolecular complex formed by TRPC1/TRPC3/TRPC6 existed in isolated cultured rat renal fibroblasts. However, specific blockade of TRPC3 with the pharmacologic inhibitor pyr3 was sufficient to inhibit both angiotensin II- and 1-oleoyl-2-acetyl-sn-glycerol-induced Ca(2+) entry in these cells, which was detected by fura-2 Ca(2+) imaging. TRPC3 blockade or Ca(2+) removal inhibited fibroblast proliferation and myofibroblast differentiation by suppressing the phosphorylation of extracellular signal-regulated kinase (ERK1/2). In addition, pyr3 inhibited fibrosis and inflammation-associated markers in a noncytotoxic manner. Furthermore, TRPC3 knockdown by siRNA confirmed these pharmacologic findings. In adult male Wistar rats or wild-type mice subjected to unilateral ureteral obstruction, TRPC3 expression increased in the fibroblasts of obstructed kidneys and was associated with increased Ca(2+) entry, ERK1/2 phosphorylation, and fibroblast proliferation. Both TRPC3 blockade in rats and TRPC3 knockout in mice inhibited ERK1/2 phosphorylation and fibroblast activation as well as myofibroblast differentiation and extracellular matrix remodeling in obstructed kidneys, thus ameliorating tubulointerstitial damage and renal fibrosis. In conclusion, TRPC3 channels are present in renal fibroblasts and control fibroblast proliferation, differentiation, and activation through Ca(2+)-mediated ERK signaling. TRPC3 channels might constitute important therapeutic targets for improving renal remodeling in kidney disease.
Collapse
Affiliation(s)
- Youakim Saliba
- Physiology and Pathophysiology Research Laboratory, Pole of Technology and Health, Faculty of Medicine and
| | - Ralph Karam
- Physiology and Pathophysiology Research Laboratory, Pole of Technology and Health, Faculty of Medicine and
| | - Viviane Smayra
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Georges Aftimos
- Department of Anatomopathology, National Institute of Pathology, Baabda, Lebanon; and
| | - Joel Abramowitz
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Lutz Birnbaumer
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Nassim Farès
- Physiology and Pathophysiology Research Laboratory, Pole of Technology and Health, Faculty of Medicine and
| |
Collapse
|
25
|
Smani T, Shapovalov G, Skryma R, Prevarskaya N, Rosado JA. Functional and physiopathological implications of TRP channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1772-82. [DOI: 10.1016/j.bbamcr.2015.04.016] [Citation(s) in RCA: 289] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 04/22/2015] [Accepted: 04/24/2015] [Indexed: 10/23/2022]
|
26
|
Earley S, Brayden JE. Transient receptor potential channels in the vasculature. Physiol Rev 2015; 95:645-90. [PMID: 25834234 DOI: 10.1152/physrev.00026.2014] [Citation(s) in RCA: 319] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The mammalian genome encodes 28 distinct members of the transient receptor potential (TRP) superfamily of cation channels, which exhibit varying degrees of selectivity for different ionic species. Multiple TRP channels are present in all cells and are involved in diverse aspects of cellular function, including sensory perception and signal transduction. Notably, TRP channels are involved in regulating vascular function and pathophysiology, the focus of this review. TRP channels in vascular smooth muscle cells participate in regulating contractility and proliferation, whereas endothelial TRP channel activity is an important contributor to endothelium-dependent vasodilation, vascular wall permeability, and angiogenesis. TRP channels are also present in perivascular sensory neurons and astrocytic endfeet proximal to cerebral arterioles, where they participate in the regulation of vascular tone. Almost all of these functions are mediated by changes in global intracellular Ca(2+) levels or subcellular Ca(2+) signaling events. In addition to directly mediating Ca(2+) entry, TRP channels influence intracellular Ca(2+) dynamics through membrane depolarization associated with the influx of cations or through receptor- or store-operated mechanisms. Dysregulation of TRP channels is associated with vascular-related pathologies, including hypertension, neointimal injury, ischemia-reperfusion injury, pulmonary edema, and neurogenic inflammation. In this review, we briefly consider general aspects of TRP channel biology and provide an in-depth discussion of the functions of TRP channels in vascular smooth muscle cells, endothelial cells, and perivascular cells under normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Scott Earley
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and Department of Pharmacology, University of Vermont College of Medicine, Burlington, Vermont
| | - Joseph E Brayden
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada; and Department of Pharmacology, University of Vermont College of Medicine, Burlington, Vermont
| |
Collapse
|
27
|
Guo BC, Wei J, Su KH, Chiang AN, Zhao JF, Chen HY, Shyue SK, Lee TS. Transient receptor potential vanilloid type 1 is vital for (-)-epigallocatechin-3-gallate mediated activation of endothelial nitric oxide synthase. Mol Nutr Food Res 2015; 59:646-57. [PMID: 25581901 DOI: 10.1002/mnfr.201400699] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/31/2014] [Accepted: 01/02/2015] [Indexed: 11/06/2022]
Abstract
SCOPE Epigallocatechin-3-gallate (EGCG), the most abundant catechin of green tea, has beneficial effects on physiological functions of endothelial cells (ECs), yet the detailed mechanisms are not fully understood. In this study, we investigated the role of transient receptor potential vanilloid type 1 (TRPV1), a ligand-gated nonselective calcium channel, in EGCG-mediated endothelial nitric oxide (NO) synthase (eNOS) activation and angiogenesis. METHODS AND RESULTS In ECs, treatment with EGCG time-dependently increased the intracellular level of Ca(2+) . Removal of extracellular calcium (Ca(2+) ) by EGTA or EDTA or inhibition of TRPV1 by capsazepine or SB366791 abrogated EGCG-increased intracellular Ca(2+) level in ECs or TRPV1-transfected HEK293 cells. Additionally, EGCG increased the phsophorylation of eNOS at Ser635 and Ser1179, Akt at Ser473, calmodulin-dependent protein kinase II (CaMKII) at Thr286 and AMP-activated protein kinase (AMPK) at Thr172, all abolished by the TRPV1 antagonist capsazepine. EGCG-induced NO production was diminished by pretreatment with LY294002 (an Akt inhibitor), KN62 (a CaMKII inhibitor), and compound C (an AMPK inhibitor). Moreover, blocking TRPV1 activation prevented EGCG-induced EC proliferation, migration, and tube formation, as well as angiogenesis in Matrigel plugs in mice. CONCLUSION EGCG may trigger activation of TRPV1-Ca(2+) signaling, which leads to phosphorylation of Akt, AMPK, and CaMKII; eNOS activation; NO production; and, ultimately, angiogenesis in ECs.
Collapse
Affiliation(s)
- Bei-Chia Guo
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Yue Z, Xie J, Yu AS, Stock J, Du J, Yue L. Role of TRP channels in the cardiovascular system. Am J Physiol Heart Circ Physiol 2015; 308:H157-82. [PMID: 25416190 PMCID: PMC4312948 DOI: 10.1152/ajpheart.00457.2014] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/14/2014] [Indexed: 12/12/2022]
Abstract
The transient receptor potential (TRP) superfamily consists of a large number of nonselective cation channels with variable degree of Ca(2+)-permeability. The 28 mammalian TRP channel proteins can be grouped into six subfamilies: canonical, vanilloid, melastatin, ankyrin, polycystic, and mucolipin TRPs. The majority of these TRP channels are expressed in different cell types including both excitable and nonexcitable cells of the cardiovascular system. Unlike voltage-gated ion channels, TRP channels do not have a typical voltage sensor, but instead can sense a variety of other stimuli including pressure, shear stress, mechanical stretch, oxidative stress, lipid environment alterations, hypertrophic signals, and inflammation products. By integrating multiple stimuli and transducing their activity to downstream cellular signal pathways via Ca(2+) entry and/or membrane depolarization, TRP channels play an essential role in regulating fundamental cell functions such as contraction, relaxation, proliferation, differentiation, and cell death. With the use of targeted deletion and transgenic mouse models, recent studies have revealed that TRP channels are involved in numerous cellular functions and play an important role in the pathophysiology of many diseases in the cardiovascular system. Moreover, several TRP channels are involved in inherited diseases of the cardiovascular system. This review presents an overview of current knowledge concerning the physiological functions of TRP channels in the cardiovascular system and their contributions to cardiovascular diseases. Ultimately, TRP channels may become potential therapeutic targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Zhichao Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jia Xie
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Albert S Yu
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jonathan Stock
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jianyang Du
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Lixia Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
29
|
Itsuki K, Imai Y, Hase H, Okamura Y, Inoue R, Mori MX. PLC-mediated PI(4,5)P2 hydrolysis regulates activation and inactivation of TRPC6/7 channels. ACTA ACUST UNITED AC 2014; 143:183-201. [PMID: 24470487 PMCID: PMC4001779 DOI: 10.1085/jgp.201311033] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transient receptor potential classical (or canonical) (TRPC)3, TRPC6, and TRPC7 are a subfamily of TRPC channels activated by diacylglycerol (DAG) produced through the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) by phospholipase C (PLC). PI(4,5)P2 depletion by a heterologously expressed phosphatase inhibits TRPC3, TRPC6, and TRPC7 activity independently of DAG; however, the physiological role of PI(4,5)P2 reduction on channel activity remains unclear. We used Förster resonance energy transfer (FRET) to measure PI(4,5)P2 or DAG dynamics concurrently with TRPC6 or TRPC7 currents after agonist stimulation of receptors that couple to Gq and thereby activate PLC. Measurements made at different levels of receptor activation revealed a correlation between the kinetics of PI(4,5)P2 reduction and those of receptor-operated TRPC6 and TRPC7 current activation and inactivation. In contrast, DAG production correlated with channel activation but not inactivation; moreover, the time course of channel inactivation was unchanged in protein kinase C-insensitive mutants. These results suggest that inactivation of receptor-operated TRPC currents is primarily mediated by the dissociation of PI(4,5)P2. We determined the functional dissociation constant of PI(4,5)P2 to TRPC channels using FRET of the PLCδ Pleckstrin homology domain (PHd), which binds PI(4,5)P2, and used this constant to fit our experimental data to a model in which channel gating is controlled by PI(4,5)P2 and DAG. This model predicted similar FRET dynamics of the PHd to measured FRET in either human embryonic kidney cells or smooth muscle cells, whereas a model lacking PI(4,5)P2 regulation failed to reproduce the experimental data, confirming the inhibitory role of PI(4,5)P2 depletion on TRPC currents. Our model also explains various PLC-dependent characteristics of channel activity, including limitation of maximum open probability, shortening of the peak time, and the bell-shaped response of total current. In conclusion, our studies demonstrate a fundamental role for PI(4,5)P2 in regulating TRPC6 and TRPC7 activity triggered by PLC-coupled receptor stimulation.
Collapse
Affiliation(s)
- Kyohei Itsuki
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Chen KH, Liu H, Yang L, Jin MW, Li GR. SKF-96365 strongly inhibits voltage-gated sodium current in rat ventricular myocytes. Pflugers Arch 2014; 467:1227-36. [PMID: 25017106 DOI: 10.1007/s00424-014-1565-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/19/2014] [Accepted: 06/23/2014] [Indexed: 12/11/2022]
Abstract
SKF-96365 (1-(beta-[3-(4-methoxy-phenyl) propoxy]-4-methoxyphenethyl)-1H-imidazole hydrochloride) is a general TRPC channel antagonist commonly used to characterize the potential functions of TRPC channels in cardiovascular system. Recent reports showed that SKF-96365 induced a reduction in cardiac conduction. The present study investigates whether the reduced cardiac conduction caused by SKF-96365 is related to the blockade of voltage-gated sodium current (I Na) in rat ventricular myocytes using the whole-cell patch voltage-clamp technique. It was found that SKF-96365 inhibited I Na in rat ventricular myocytes in a concentration-dependent manner. The compound (1 μM) negatively shifted the potential of I Na availability by 9.5 mV, increased the closed-state inactivation of I Na, and slowed the recovery of I Na from inactivation. The inhibition of cardiac I Na by SKF-96365 was use-dependent and frequency-dependent, and the IC₅₀ was decreased from 1.36 μM at 0.5 Hz to 1.03, 0.81, 0.61, 0.56 μM at 1, 2, 5, 10 Hz, respectively. However, the selective TRPC3 antagonist Pyr3 decreased cardiac I Na by 8.5% at 10 μM with a weak use and frequency dependence. These results demonstrate that the TRPC channel antagonist SKF-96365 strongly blocks cardiac I Na in use-dependent and frequency-dependent manners. Caution should be taken for interpreting the alteration of cardiac electrical activity when SKF-96365 is used in native cells as a TRPC antagonist.
Collapse
Affiliation(s)
- Kui-Hao Chen
- Department of Physiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong, China
| | | | | | | | | |
Collapse
|
31
|
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. [PMID: 24951385 DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 377] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The large Trp gene family encodes transient receptor potential (TRP) proteins that form novel cation-selective ion channels. In mammals, 28 Trp channel genes have been identified. TRP proteins exhibit diverse permeation and gating properties and are involved in a plethora of physiologic functions with a strong impact on cellular sensing and signaling pathways. Indeed, mutations in human genes encoding TRP channels, the so-called "TRP channelopathies," are responsible for a number of hereditary diseases that affect the musculoskeletal, cardiovascular, genitourinary, and nervous systems. This review gives an overview of the functional properties of mammalian TRP channels, describes their roles in acquired and hereditary diseases, and discusses their potential as drug targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bernd Nilius
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| | - Arpad Szallasi
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| |
Collapse
|
32
|
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
|
33
|
König S, Browne S, Doleschal B, Schernthaner M, Poteser M, Mächler H, Wittchow E, Braune M, Muik M, Romanin C, Groschner K. Inhibition of Orai1-mediated Ca(2+) entry is a key mechanism of the antiproliferative action of sirolimus in human arterial smooth muscle. Am J Physiol Heart Circ Physiol 2013; 305:H1646-57. [PMID: 24056904 DOI: 10.1152/ajpheart.00365.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sirolimus (rapamycin) is used in drug-eluting stent strategies and proved clearly superior in this application compared with other immunomodulators such as pimecrolimus. The molecular basis of this action of sirolimus in the vascular system is still incompletely understood. Measurements of cell proliferation in human coronary artery smooth muscle cells (hCASM) demonstrated a higher antiproliferative activity of sirolimus compared with pimecrolimus. Although sirolimus lacks inhibitory effects on calcineurin, nuclear factor of activated T-cell activation in hCASM was suppressed to a similar extent by both drugs at 10 μM. Sirolimus, but not pimecrolimus, inhibited agonist-induced and store-operated Ca(2+) entry as well as cAMP response element binding protein (CREB) phosphorylation in human arterial smooth muscle, suggesting the existence of an as-yet unrecognized inhibitory effect of sirolimus on Ca(2+) signaling and Ca(2+)-dependent gene transcription. Electrophysiological experiments revealed that only sirolimus but not pimecrolimus significantly blocked the classical stromal interaction molecule/Orai-mediated, store-operated Ca(2+) current reconstituted in human embryonic kidney cells (HEK293). A link between Orai function and proliferation was confirmed by dominant-negative knockout of Orai in hCASM. Analysis of the effects of sirolimus on cell proliferation and CREB activation in an in vitro model of arterial intervention using human aorta corroborated the ability of sirolimus to suppress stent implantation-induced CREB activation in human arteries. We suggest inhibition of store-operated Ca(2+) entry based on Orai channels and the resulting suppression of Ca(2+) transcription coupling as a key mechanism underlying the antiproliferative activity of sirolimus in human arteries. This mechanism of action is specific for sirolimus and not a general feature of drugs interacting with FK506-binding proteins.
Collapse
Affiliation(s)
- Sarah König
- Institute of Biophysics, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Transient receptor potential canonical type 3 channels--their evolving role in hypertension and its related complications. J Cardiovasc Pharmacol 2013; 61:455-60. [PMID: 23364606 DOI: 10.1097/fjc.0b013e31828748a1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
: Recent studies indicate that transient receptor potential canonical type 3 (TRPC3) channels contribute to the regulation of blood pressure and vascular and renal function. Several studies show that TRPC3 dysfunction is associated with hypertension, atherosclerosis, cardiac hypertrophy, and cerebrovascular events. In this review, we summarize the role of TRPC3 channels in the cardiovascular system, and we focus on their pathophysiological role in hypertension and related target organ damages. We provide new insight into the involvement of TRPC3 channels in the development of hypertension and its related complications.
Collapse
|
35
|
Senadheera S, Bertrand PP, Grayson TH, Leader L, Tare M, Murphy TV, Sandow SL. Enhanced contractility in pregnancy is associated with augmented TRPC3, L-type, and T-type voltage-dependent calcium channel function in rat uterine radial artery. Am J Physiol Regul Integr Comp Physiol 2013; 305:R917-26. [DOI: 10.1152/ajpregu.00225.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In pregnancy, α-adrenoceptor-mediated vasoconstriction is augmented in uterine radial arteries and is accompanied by underlying changes in smooth muscle (SM) Ca2+ activity. This study aims to determine the Ca2+ entry channels associated with altered vasoconstriction in pregnancy, with the hypothesis that augmented vasoconstriction involves transient receptor potential canonical type-3 (TRPC3) and L- and T-type voltage-dependent Ca2+ channels. Immunohistochemistry showed TRPC3, L-type Cav1.2 (as the α1C subunit), T-type Cav3.1 (α1G), and Cav3.2 (α1H) localization to the uterine radial artery SM. Fluorescence intensity of TRPC3, Cav1.2, and Cav3.2 was increased, and Cav3.1 decreased in radial artery SM from pregnant rats. Western blot analysis confirmed increased TRPC3 protein expression in the radial artery from pregnant rats. Pressure myography incorporating pharmacological intervention to examine the role of these channels in uterine radial arteries showed an attenuation of phenylephrine (PE)-induced constriction with Pyr3 {1-[4-[(2,3,3-trichloro-1-oxo-2-propen-1-yl)amino]phenyl]-5-(trifluoromethyl)-1 H-pyrazole-4-carboxylic acid}-mediated TRPC3 inhibition or with nifedipine-mediated L-type channel block alone in vessels from pregnant rats; both effects of which were diminished in radial arteries from nonpregnant rats. Combined TRPC3 and L-type inhibition attenuated PE-induced constriction in radial arteries, and the residual vasoconstriction was reduced and abolished with T-type channel block with NNC 55-0396 in arteries from nonpregnant and pregnant rats, respectively. With SM Ca2+ stores depleted and in the presence of PE, nifedipine, and NNC 55-0396, blockade of TRPC3 reversed PE-induced constriction. These data suggest that TRPC3 channels act synergistically with L- and T-type channels to modulate radial artery vasoconstriction, with the mechanism being augmented in pregnancy.
Collapse
Affiliation(s)
- Sevvandi Senadheera
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Paul P. Bertrand
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - T. Hilton Grayson
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Leo Leader
- Leo Leader, School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Marianne Tare
- Department of Physiology, Monash University, Melbourne, Australia; and
| | - Timothy V. Murphy
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Shaun L. Sandow
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydoore, Australia
| |
Collapse
|
36
|
Ryu HJ, Kim JE, Kim YJ, Kim JY, Kim WIL, Choi SY, Kim MJ, Kang TC. Endothelial transient receptor potential conical channel (TRPC)-3 activation induces vasogenic edema formation in the rat piriform cortex following status epilepticus. Cell Mol Neurobiol 2013; 33:575-85. [PMID: 23529532 PMCID: PMC11497918 DOI: 10.1007/s10571-013-9931-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 03/18/2013] [Indexed: 11/29/2022]
Abstract
Transient receptor potential canonical channel (TRPC) is a nonselective cation channel permeable to Ca(2+), which express in many cell types, including neurons. However the alterations in TRPC receptor expressions in response to status epilepticus (SE) have not been explored. Therefore, the present study was designated to elucidate the roles of TRPC3 in neuronal death and vasogenic edema within the rat piriform cortex (PC) following SE. In non-SE animals, TRPC3 immunoreactivity was abundantly detected in the PC. Following SE, TRPC3 immunoreactivity was increased in neurons. Furthermore, TRPC3 expression was detected in endothelial cells that did not contain it in non-SE animals. Loss of SMI-71 (a blood-brain barrier antigen) immunoreactivity was also observed in TRPC3 positive endothelial cells. In addition, FJB positive neurons and vasogenic edema were noticeably detected in the PC. To directly determine whether TRPC3 activation is correlated to SE-induced vasogenic edema formation and neuronal damages in the PC, the effect of Pyr-3 (a TRPC3 antagonist) on SE-induced insults were investigated. Pyr-3 infusion effectively attenuated vasogenic edema in the PC as compared to the vehicle. Therefore, our findings indicate that TRPC3 activation/overexpression induced by SE may involve BBB disruption and neuronal damages in the rat PC following SE. Therefore, the present study was TRPC3 may play an important role in SE-induced vasogenic edema formation through BBB disruptions in the rat PC.
Collapse
Affiliation(s)
- Hea Jin Ryu
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
| | - Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
| | - Yeon-Joo Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
| | - Ji-Yang Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
| | - Won IL Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
| | - So-Yeon Choi
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
| | - Min-Ju Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon, 200-702 South Korea
| |
Collapse
|