1
|
Silva A, Prior R, D'Antonio M, Swinnen JV, Van Den Bosch L. Lipid metabolism alterations in peripheral neuropathies. Neuron 2025:S0896-6273(25)00262-4. [PMID: 40311611 DOI: 10.1016/j.neuron.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
Alterations in lipid metabolism are increasingly recognized as central pathological hallmarks of inherited and acquired peripheral neuropathies. Correct lipid balance is critical for cellular homeostasis. However, the mechanisms linking lipid disturbances to cellular dysfunction and whether these changes are primary drivers or secondary effects of disease remain unresolved. This is particularly relevant in the peripheral nervous system, where the lipid-rich myelin integrity is critical for axonal function, and even subtle perturbations can cause widespread effects. This review explores the role of lipids as structural components as well as signaling molecules, emphasizing their metabolic role in peripheral neurons and Schwann cells. Additionally, we explore the genetic and environmental connections in both inherited and acquired peripheral neuropathies, respectively, which are known to affect lipid metabolism in peripheral neurons or Schwann cells. Overall, we highlight how understanding lipid-centric mechanisms could advance biomarker discovery and therapeutic interventions for peripheral nerve disorders.
Collapse
Affiliation(s)
- Alessio Silva
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| | - Robert Prior
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium; Department of Ophthalmology, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Maurizio D'Antonio
- Biology of Myelin Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| |
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
|
Silva-Cardoso GK, Boda VK, Li W, N'Gouemo P. Inhibition of TRPC3 channels suppresses seizure susceptibility in the genetically-epilepsy prone rats. Eur J Pharmacol 2024; 977:176722. [PMID: 38851562 PMCID: PMC11295865 DOI: 10.1016/j.ejphar.2024.176722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/27/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Transient receptor potential canonical 3 (TRPC3) channels are important in regulating Ca2+ homeostasis and have been implicated in the pathophysiology of chemically induced seizures. Inherited seizure susceptibility in genetically epilepsy-prone rats (GEPR-3s) has been linked to increased voltage-gated Ca2+ channel currents in the inferior colliculus neurons, which can affect intraneuronal Ca2+ homeostasis. However, whether TRPC3 channels also contribute to inherited seizure susceptibility in GEPR-3s is unclear. This study investigated the effects of JW-65, a potent and selective inhibitor of TRPC3 channels, on acoustically evoked seizure susceptibility in adult male and female GEPR-3s. These seizures consisted of wild running seizures (WRSs) that evolved into generalized tonic-clonic seizures (GTCSs). The results showed that acute administration of low doses of JW-65 significantly decreased by 55-89% the occurrence of WRSs and GTCSs and the seizure severity in both male and female GEPR-3s. This antiseizure effect was accompanied by increased seizure latency and decreased seizure duration. Additionally, female GEPR-3s were more responsive to JW-65's antiseizure effects than males. Moreover, JW-65 treatment for five consecutive days completely suppressed acoustically evoked seizures in male and female GEPR-3s. These findings suggest that inhibiting TRPC3 channels could be a promising antiseizure strategy targeting Ca2+ signaling mechanisms in inherited generalized tonic-clonic epilepsy.
Collapse
Affiliation(s)
- Gleice K Silva-Cardoso
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC, 20059, USA
| | - Vijay K Boda
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Wei Li
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Prosper N'Gouemo
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC, 20059, USA.
| |
Collapse
|
4
|
Presence of TRPA1 Modifies CD4+/CD8+ T Lymphocyte Ratio and Activation. Pharmaceuticals (Basel) 2022; 15:ph15010057. [PMID: 35056114 PMCID: PMC8781558 DOI: 10.3390/ph15010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/10/2022] Open
Abstract
Transient Receptor Potential Ankyrin 1 (TRPA1) has been reported to influence neuroinflammation and lymphocyte function. We analysed the immune phenotype and activation characteristics of TRPA1-deficient mice (knockout—KO) generated by targeted deletion of the pore-loop domain of the ion channel. We compared TRPA1 mRNA and protein expression in monocyte and lymphocyte subpopulations isolated from primary and secondary lymphatic organs of wild type (WT) and KO mice. qRT-PCR and flow cytometric studies indicated a higher level of TRPA1 in monocytes than in lymphocytes, but both were orders of magnitude lower than in sensory neurons. We found lower CD4+/CD8+ thymocyte ratios, diminished CD4/CD8 rates, and B cell numbers in the KO mice. Early activation marker CD69 was lower in CD4+ T cells of KO, while the level of CD8+/CD25+ cells was higher. In vitro TcR-mediated activation did not result in significant differences in CD69 level between WT and KO splenocytes, but lower cytokine (IL-1β, IL-6, TNF-α, IL-17A, IL-22, and RANTES) secretion was observed in KO splenocytes. Basal intracellular Ca2+ level and TcR-induced Ca2+ signal in T lymphocytes did not differ significantly, but interestingly, imiquimod-induced Ca2+ level in KO thymocytes was higher. Our results support the role of TRPA1 in the regulation of activation, cytokine production, and T and B lymphocytes composition in mice.
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
|
Xie R, Wang Z, Liu T, Xiao R, Lv K, Wu C, Luo Y, Cai Y, Fan X. AAV Delivery of shRNA Against TRPC6 in Mouse Hippocampus Impairs Cognitive Function. Front Cell Dev Biol 2021; 9:688655. [PMID: 34327201 PMCID: PMC8313999 DOI: 10.3389/fcell.2021.688655] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/22/2021] [Indexed: 11/13/2022] Open
Abstract
Transient Receptor Potential Canonical 6 (TRPC6) has been suggested to be involved in synapse function and contribute to hippocampal-dependent cognitive processes. Gene silencing of TRPC6 was performed by injecting adeno-associated virus (AAV) expressing TRPC6-specific shRNA (shRNA-TRPC6) into the hippocampal dentate gyrus (DG). Spatial learning, working memory and social recognition memory were impaired in the shRNA-TRPC6 treated mice compared to control mice after 4 weeks. In addition, gene ontology (GO) analysis of RNA-sequencing revealed that viral intervention of TRPC6 expression in DG resulted in the enrichment of the process of synaptic transmission and cellular compartment of synaptic structure. KEGG analysis showed PI3K-Akt signaling pathway were significantly down-regulated. Furthermore, the shRNA-TRPC6 treatment reduced dendritic spines of DG granule neurons, in terms of spine loss, the thin and mushroom types predominated. Accompanying the spine loss, the levels of PSD95, pAkt and CREB in the hippocampus were decreased in the shRNA-TRPC6 treated animals. Taken together, our results suggest that knocking down TRPC6 in the DG have a disadvantageous effect on cognitive processes.
Collapse
Affiliation(s)
- Ruxin Xie
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhongke Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rui Xiao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Keyi Lv
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuan Wu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yun Cai
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
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
|
Canonical transient receptor potential 6 channel deficiency promotes smooth muscle cells dedifferentiation and increased proliferation after arterial injury. JVS Vasc Sci 2020; 1:136-150. [PMID: 33554153 PMCID: PMC7861475 DOI: 10.1016/j.jvssci.2020.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Objective Previous studies showed the benefit of canonical transient receptor potential 6 (TRPC6) channel deficiency in promoting endothelial healing of arterial injuries in hypercholesterolemic animals. Long-term studies utilizing a carotid wire-injury model were undertaken in wild-type (WT) and TRPC6-/- mice to determine the effects of TRPC6 on phenotypic modulation of vascular smooth muscle cells (SMC) and neointimal hyperplasia. We hypothesized that TRPC6 was essential in the maintenance or reexpression of a differentiated SMC phenotype and minimized luminal stenosis following arterial injury. Methods The common carotid arteries (CCA) of WT and TRPC6-/- mice were evaluated at baseline and 4 weeks after wire injury. At baseline, CCA of TRPC6-/- mice had reduced staining of MYH11 and SM22, fewer elastin lamina, luminal dilation, and wall thinning. After carotid wire injury, TRPC6-/- mice developed significantly more pronounced luminal stenosis compared with WT mice. Injured TRPC6-/- CCA demonstrated increased medial/intimal cell number and active cell proliferation when compared with WT CCA. Immunohistochemistry suggested that expression of contractile biomarkers in medial SMC were essentially at baseline levels in WT CCA at 28 days after wire injury. By contrast, at 28 days after injury medial SMC from TRPC6-/- CCA showed a significant decrease in the expression of contractile biomarkers relative to baseline levels. To assess the role of TRPC6 in systemic arterial SMC phenotype modulation, SMC were harvested from thoracic aortae of WT and TRPC6-/- mice and were characterized. TRPC6-/- SMC showed enhanced proliferation and migration in response to serum stimulation. Expression of contractile phenotype biomarkers, MYH11 and SM22, was attenuated in TRPC6-/- SMC. siRNA-mediated TRPC6 deficiency inhibited contractile biomarker expression in a mouse SMC line. Conclusions These results suggest that TRPC6 contributes to the restoration or maintenance of arterial SMC contractile phenotype following injury. Understanding the role of TRPC6 in phenotypic modulation may lead to mechanism-based therapies for attenuation of IH. After endovascular intervention and open vascular surgery, vascular smooth muscle cells (VSMC) undergo a coordinated reprogramming of gene expression to facilitate arterial healing. Down regulation of VSMC-specific contractile biomarkers (eg, SM22 and MYH11) and induction of pathways that promote cell proliferation, migration, and matrix synthesis are hallmarks of this phenotypic switch. Dysregulated phenotypic switching leads to the development of neointimal hyperplasia and vascular restenosis. Identifying pathways that regulate or constrain VSMC phenotypic modulation, therefore, has the potential to decrease neointimal hyperplasia and improve outcomes after vascular intervention. In this study, we demonstrate that depletion of the non-voltage-gated cation channel TRPC6 promotes phenotypic switching and loss of contractile biomarkers in systemic arterial VSMC. TRPC6-/- mice developed significantly more pronounced luminal stenosis compared with wild-type mice after carotid wire injury. These results suggest that TRPC6 contributes to the restoration or maintenance of contractile phenotype in VSMC after injury. Understanding the role of TRPC6 in phenotypic switching may lead to mechanism-based therapies to mitigate restenosis.
Collapse
|
9
|
TASK channels: channelopathies, trafficking, and receptor-mediated inhibition. Pflugers Arch 2020; 472:911-922. [DOI: 10.1007/s00424-020-02403-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 01/06/2023]
|
10
|
Fiedler S, Storch U, Erdogmus S, Gudermann T, Mederos Y Schnitzler M, Dietrich A. Small Fluorescein Arsenical Hairpin-Based Förster Resonance Energy Transfer Analysis Reveals Changes in Amino- to Carboxyl-Terminal Interactions upon OAG Activation of Classical Transient Receptor Potential 6. Mol Pharmacol 2019; 96:90-98. [PMID: 31171574 DOI: 10.1124/mol.119.115949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/02/2019] [Indexed: 01/19/2023] Open
Abstract
Although the overall structure of many classical transient receptor potential proteins (TRPC), including human and murine TRPC6, were recently resolved by cryoelectron microscopy analysis, structural changes during channel activation by 1-oleoyl-1-acetyl-sn-glycerol (OAG), the membrane-permeable analog of diacylglycerol, were not defined. Moreover, data on carboxyl- and amino-terminal interactions were not provided, as the amino-terminal regions of murine and human TRPC6 were not resolved. Therefore, we employed a Förster resonance energy transfer (FRET) approach using a small fluorescein arsenical hairpin (FlAsH) targeted to a short tetracysteine sequence at the unresolved amino-terminus and cerulean, a cyan fluorescent protein, as a tag at the carboxyl-terminus of the murine TRPC6 protein. After OAG as well as GSK-1702934A activation, FRET efficiency was simultaneously and significantly reduced, indicating a decreased interaction between the amino to carboxyl termini in the functional tagged murine TRPC6 tetramer (TRPC6 WT) heterologously expressed in human embryonic kidney 293T cells. There was a significant reduction in the FRET signal obtained from analysis of murine TRPC6 FRET constructs with homologous amino-terminal mutations (M131T, G108S) that had been identified in human patients with inherited focal segmental glomerulosclerosis, a condition that can lead to end-stage renal disease. A novel, designed loss-of-function TRPC6 mutation (N109A) in the amino-terminus in close proximity to the carboxyl-terminus produced similar FRET ratios. SIGNIFICANCE STATEMENT: Our data show for the first time that FlAsH-tagging of ion channels is a promising tool to study conformational changes after channel opening and may significantly advance the analysis of ion channel activation as well as their mutants involved in channelopathies.
Collapse
Affiliation(s)
- Susanne Fiedler
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Ursula Storch
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Serap Erdogmus
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Thomas Gudermann
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Michael Mederos Y Schnitzler
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Alexander Dietrich
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| |
Collapse
|
11
|
Ibeh CL, Yiu AJ, Kanaras YL, Paal E, Birnbaumer L, Jose PA, Bandyopadhyay BC. Evidence for a regulated Ca 2+ entry in proximal tubular cells and its implication in calcium stone formation. J Cell Sci 2019; 132:jcs.225268. [PMID: 30910829 DOI: 10.1242/jcs.225268] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 03/14/2019] [Indexed: 12/14/2022] Open
Abstract
Calcium phosphate (CaP) crystals, which begin to form in the early segments of the loop of Henle (LOH), are known to act as precursors for calcium stone formation. The proximal tubule (PT), which is just upstream of the LOH and is a major site for Ca2+ reabsorption, could be a regulator of such CaP crystal formation. However, PT Ca2+ reabsorption is mostly described as being paracellular. Here, we show the existence of a regulated transcellular Ca2+ entry pathway in luminal membrane PT cells induced by Ca2+-sensing receptor (CSR, also known as CASR)-mediated activation of transient receptor potential canonical 3 (TRPC3) channels. In support of this idea, we found that both CSR and TRPC3 are physically and functionally coupled at the luminal membrane of PT cells. More importantly, TRPC3-deficient mice presented with a deficiency in PT Ca2+ entry/transport, elevated urinary [Ca2+], microcalcifications in LOH and urine microcrystals formations. Taken together, these data suggest that a signaling complex comprising CSR and TRPC3 exists in the PT and can mediate transcellular Ca2+ transport, which could be critical in maintaining the PT luminal [Ca2+] to mitigate formation of the CaP crystals in LOH and subsequent formation of calcium stones.
Collapse
Affiliation(s)
- Cliff-Lawrence Ibeh
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA
| | - Allen J Yiu
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA.,Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University, Washington DC, DC 20037, USA
| | - Yianni L Kanaras
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA
| | - Edina Paal
- Pathology and Laboratory Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA
| | - Lutz Birnbaumer
- Division of Intramural Research, NIEHS, Research Triangle Park, Durham, NC 27709, USA.,Institute for Biomedical Research (BIOMED), Catholic University of Argentina, C1107AFF Buenos Aires, Argentina
| | - Pedro A Jose
- Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University, Washington DC, DC 20037, USA.,Department of Pharmacology and Physiology, The George Washington University, Washington DC, DC 20037, USA
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA .,Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University, Washington DC, DC 20037, USA.,Department of Pharmacology and Physiology, The George Washington University, Washington DC, DC 20037, USA
| |
Collapse
|
12
|
Spires D, Manis AD, Staruschenko A. Ion channels and transporters in diabetic kidney disease. CURRENT TOPICS IN MEMBRANES 2019; 83:353-396. [PMID: 31196609 PMCID: PMC6815098 DOI: 10.1016/bs.ctm.2019.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Type 1 and 2 diabetes mellitus are major medical epidemics affecting millions of patients worldwide. Diabetes mellitus is the leading cause of diabetic kidney disease (DKD), which is the most common cause of end-stage renal disease (ESRD). DKD is associated with significant changes in renal hemodynamics and electrolyte transport. Alterations in renal ion transport triggered by pathophysiological conditions in diabetes can exacerbate hypertension, accelerate renal injury, and are integral to the development of DKD. Renal ion transporters and electrolyte homeostasis play a fundamental role in functional changes and injury to the kidney during DKD. With the large number of ion transporters involved in DKD, understanding the roles of individual transporters as well as the complex cascades through which they interact is essential in the development of effective treatments for patients suffering from this disease. This chapter aims to gather current knowledge of the major renal ion transporters with altered expression and activity under diabetic conditions, and provide a comprehensive overview of their interactions and collective functions in DKD.
Collapse
Affiliation(s)
- Denisha Spires
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anna D Manis
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States; Clement J. Zablocki VA Medical Center, Milwaukee, WI, United States.
| |
Collapse
|
13
|
Modulators of Transient Receptor Potential (TRP) Channels as Therapeutic Options in Lung Disease. Pharmaceuticals (Basel) 2019; 12:ph12010023. [PMID: 30717260 PMCID: PMC6469169 DOI: 10.3390/ph12010023] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/25/2022] Open
Abstract
The lungs are essential for gas exchange and serve as the gateways of our body to the external environment. They are easily accessible for drugs from both sides, the airways and the vasculature. Recent literature provides evidence for a role of Transient Receptor Potential (TRP) channels as chemosensors and essential members of signal transduction cascades in stress-induced cellular responses. This review will focus on TRP channels (TRPA1, TRPC6, TRPV1, and TRPV4), predominantly expressed in non-neuronal lung tissues and their involvement in pathways associated with diseases like asthma, cystic fibrosis, chronic obstructive pulmonary disease (COPD), lung fibrosis, and edema formation. Recently identified specific modulators of these channels and their potential as new therapeutic options as well as strategies for a causal treatment based on the mechanistic understanding of molecular events will also be evaluated.
Collapse
|
14
|
Reyes RV, Castillo-Galán S, Hernandez I, Herrera EA, Ebensperger G, Llanos AJ. Revisiting the Role of TRP, Orai, and ASIC Channels in the Pulmonary Arterial Response to Hypoxia. Front Physiol 2018; 9:486. [PMID: 29867539 PMCID: PMC5949889 DOI: 10.3389/fphys.2018.00486] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/16/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary arteries are exquisitely responsive to oxygen changes. They rapidly and proportionally contract as arterial PO2 decrease, and they relax as arterial PO2 is re-established. The hypoxic pulmonary vasoconstriction (HPV) is intrinsic since it does not require neural or endocrine factors, as evidenced in isolated vessels. On the other hand, pulmonary arteries also respond to sustained hypoxia with structural and functional remodeling, involving growth of smooth muscle medial layer and later recruitment of adventitial fibroblasts, secreted mitogens from endothelium and changes in the response to vasoconstrictor and vasodilator stimuli. Hypoxic pulmonary arterial vasoconstriction and remodeling are relevant biological responses both under physiological and pathological conditions, to explain matching between ventilation and perfusion, fetal to neonatal transition of pulmonary circulation and pulmonary artery over-constriction and thickening in pulmonary hypertension. Store operated channels (SOC) and receptor operated channels (ROC) are plasma membrane cationic channels that mediate calcium influx in response to depletion of internal calcium stores or receptor activation, respectively. They are involved in both HPV and pathological remodeling since their pharmacological blockade or genetic suppression of several of the Stim, Orai, TRP, or ASIC proteins in SOC or ROC complexes attenuate the calcium increase, the tension development, the pulmonary artery smooth muscle proliferation, and pulmonary arterial hypertension. In this Mini Review, we discussed the evidence obtained in in vivo animal models, at the level of isolated organ or cells of pulmonary arteries, and we identified and discussed the questions for future research needed to validate these signaling complexes as targets against pulmonary hypertension.
Collapse
Affiliation(s)
- Roberto V Reyes
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Sebastián Castillo-Galán
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ismael Hernandez
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Emilio A Herrera
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Germán Ebensperger
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Aníbal J Llanos
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| |
Collapse
|
15
|
Dong P, Guo C, Huang S, Ma M, Liu Q, Luo W. TRPC3 Is Dispensable for β-Alanine Triggered Acute Itch. Sci Rep 2017; 7:13869. [PMID: 29066740 PMCID: PMC5654773 DOI: 10.1038/s41598-017-12770-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/14/2017] [Indexed: 01/08/2023] Open
Abstract
The detection of pruritic (itchy) stimuli is mediated by a variety of receptors and channels expressed by primary sensory neurons. The G protein-coupled receptor (GPCR) MRGPRD is selectively expressed by a subset of mouse non-peptidergic nociceptors and functions as the molecular receptor for the itch-inducing chemical β-alanine. However, the channels responsible for generating electrical signals downstream of MRGPRD remain unclear. Here, we found that a member of the canonical TRP channel family, TRPC3, is highly expressed in MRGPRD+ non-peptidergic nociceptors, raising the possibility of whether TRPC3 functions as a downstream channel in the MRGPRD signaling pathway. We tested TrpC3 null mice for β-alanine induced itch, and found that these mice exhibit normal responses to β-alanine. At the cellular level, calcium influx triggered by β-alanine is also unchanged in cultured DRG neurons from TrpC3 null mice compared to wild type. Together, our results demonstrate that mouse TrpC3 is dispensable for β-alanine-induced acute itch.
Collapse
Affiliation(s)
- Peter Dong
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Changxiong Guo
- Department of Anesthesiology and the Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Shengxiang Huang
- Department of Anesthesiology and the Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Wuhan University, Wuhan, Hubei, China
| | - Minghong Ma
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qin Liu
- Department of Anesthesiology and the Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
16
|
Malczyk M, Erb A, Veith C, Ghofrani HA, Schermuly RT, Gudermann T, Dietrich A, Weissmann N, Sydykov A. The Role of Transient Receptor Potential Channel 6 Channels in the Pulmonary Vasculature. Front Immunol 2017; 8:707. [PMID: 28670316 PMCID: PMC5472666 DOI: 10.3389/fimmu.2017.00707] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/31/2017] [Indexed: 01/21/2023] Open
Abstract
Canonical or classical transient receptor potential channel 6 (TRPC6) is a Ca2+-permeable non-selective cation channel that is widely expressed in the heart, lung, and vascular tissues. The use of TRPC6-deficient (“knockout”) mice has provided important insights into the role of TRPC6 in normal physiology and disease states of the pulmonary vasculature. Evidence indicates that TRPC6 is a key regulator of acute hypoxic pulmonary vasoconstriction. Moreover, several studies implicated TRPC6 in the pathogenesis of pulmonary hypertension. Furthermore, a unique genetic variation in the TRPC6 gene promoter has been identified, which might link the inflammatory response to the upregulation of TRPC6 expression and ultimate development of pulmonary vascular abnormalities in idiopathic pulmonary arterial hypertension. Additionally, TRPC6 is critically involved in the regulation of pulmonary vascular permeability and lung edema formation during endotoxin or ischemia/reperfusion-induced acute lung injury. In this review, we will summarize latest findings on the role of TRPC6 in the pulmonary vasculature.
Collapse
Affiliation(s)
- Monika Malczyk
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Alexandra Erb
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Christine Veith
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Hossein Ardeschir Ghofrani
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Thomas Gudermann
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig Maximilian University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Alexander Dietrich
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig Maximilian University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| |
Collapse
|
17
|
Transient receptor potential canonical type 3 channels: Interactions, role and relevance - A vascular focus. Pharmacol Ther 2017; 174:79-96. [DOI: 10.1016/j.pharmthera.2017.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Leonhardt J, Villela DC, Teichmann A, Münter LM, Mayer MC, Mardahl M, Kirsch S, Namsolleck P, Lucht K, Benz V, Alenina N, Daniell N, Horiuchi M, Iwai M, Multhaup G, Schülein R, Bader M, Santos RA, Unger T, Steckelings UM. Evidence for Heterodimerization and Functional Interaction of the Angiotensin Type 2 Receptor and the Receptor MAS. Hypertension 2017; 69:1128-1135. [PMID: 28461604 DOI: 10.1161/hypertensionaha.116.08814] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/21/2016] [Accepted: 04/06/2017] [Indexed: 11/16/2022]
Abstract
The angiotensin type 2 receptor (AT2R) and the receptor MAS are receptors of the protective arm of the renin-angiotensin system. They mediate strikingly similar actions. Moreover, in various studies, AT2R antagonists blocked the effects of MAS agonists and vice versa. Such cross-inhibition may indicate heterodimerization of these receptors. Therefore, this study investigated the molecular and functional interplay between MAS and the AT2R. Molecular interactions were assessed by fluorescence resonance energy transfer and by cross correlation spectroscopy in human embryonic kidney-293 cells transfected with vectors encoding fluorophore-tagged MAS or AT2R. Functional interaction of AT2R and MAS was studied in astrocytes with CX3C chemokine receptor-1 messenger RNA expression as readout. Coexpression of fluorophore-tagged AT2R and MAS resulted in a fluorescence resonance energy transfer efficiency of 10.8 ± 0.8%, indicating that AT2R and MAS are capable to form heterodimers. Heterodimerization was verified by competition experiments using untagged AT2R and MAS. Specificity of dimerization of AT2R and MAS was supported by lack of dimerization with the transient receptor potential cation channel, subfamily C-member 6. Dimerization of the AT2R was abolished when it was mutated at cysteine residue 35. AT2R and MAS stimulation with the respective agonists, Compound 21 or angiotensin-(1-7), significantly induced CX3C chemokine receptor-1 messenger RNA expression. Effects of each agonist were blocked by an AT2R antagonist (PD123319) and also by a MAS antagonist (A-779). Knockout of a single of these receptors made astrocytes unresponsive for both agonists. Our results suggest that MAS and the AT2R form heterodimers and that-at least in astrocytes-both receptors functionally depend on each other.
Collapse
Affiliation(s)
- Julia Leonhardt
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Daniel C Villela
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Anke Teichmann
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Lisa-Marie Münter
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Magnus C Mayer
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Maibritt Mardahl
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Sebastian Kirsch
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Pawel Namsolleck
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Kristin Lucht
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Verena Benz
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Natalia Alenina
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Nicholas Daniell
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Masatsugu Horiuchi
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Masaru Iwai
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Gerhard Multhaup
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Ralf Schülein
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Michael Bader
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Robson A Santos
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Thomas Unger
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Ulrike Muscha Steckelings
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.).
| |
Collapse
|
19
|
Dietrich A, Steinritz D, Gudermann T. Transient receptor potential (TRP) channels as molecular targets in lung toxicology and associated diseases. Cell Calcium 2017; 67:123-137. [PMID: 28499580 DOI: 10.1016/j.ceca.2017.04.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/24/2022]
Abstract
The lungs as the gateways of our body to the external environment are essential for gas exchange. They are also exposed to toxicants from two sides, the airways and the vasculature. Apart from naturally produced toxic agents, millions of human made chemicals were produced since the beginning of the industrial revolution whose toxicity still needs to be determined. While the knowledge about toxic substances is increasing only slowly, a paradigm shift regarding the proposed mechanisms of toxicity at the plasma membrane emerged. According to their broad-range chemical reactivity, the mechanism of lung injury evoked by these agents has long been described as rather unspecific. Consequently, therapeutic options are still restricted to symptomatic treatment. The identification of molecular down-stream effectors in cells was a major step forward in the mechanistic understanding of the action of toxic chemicals and will pave the way for more causal and specific toxicity testing as well as therapeutic options. In this context, the involvement of Transient Receptor Potential (TRP) channels as chemosensors involved in the detection and effectors of toxicant action is an attractive concept intensively discussed in the scientific community. In this review we will summarize recent evidence for an involvement of TRP channels (TRPA1, TRPC4, TRPC6, TRPV1, TRPV4, TRPM2 and TRPM8) expressed in the lung in pathways of toxin sensing and as mediators of lung inflammation and associated diseases like asthma, COPD, lung fibrosis and edema formation. Specific modulators of these channels may offer new therapeutic options in the future and will endorse strategies for a causal, specifically tailored treatment based on the mechanistic understanding of molecular events induced by lung-toxic agents.
Collapse
Affiliation(s)
- Alexander Dietrich
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), LMU Munich, Germany.
| | - Dirk Steinritz
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), LMU Munich, Germany; Bundeswehr-Institute of Pharmacology and Toxicology, Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), LMU Munich, Germany
| |
Collapse
|
20
|
Wang M, Tang YB, Ma MM, Chen JH, Hu CP, Zhao SP, Peng DQ, Zhou JG, Guan YY, Zhang Z. TRPC3 channel confers cerebrovascular remodelling during hypertension via transactivation of EGF receptor signalling. Cardiovasc Res 2015; 109:34-43. [DOI: 10.1093/cvr/cvv246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 10/11/2015] [Indexed: 01/07/2023] Open
|
21
|
Ambrus L, Oláh A, Oláh T, Balla G, Saleem MA, Orosz P, Zsuga J, Bíró K, Csernoch L, Bíró T, Szabó T. Inhibition of TRPC6 by protein kinase C isoforms in cultured human podocytes. J Cell Mol Med 2015; 19:2771-9. [PMID: 26404773 PMCID: PMC4687697 DOI: 10.1111/jcmm.12660] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 06/23/2015] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential canonical‐6 (TRPC6) ion channels, expressed at high levels in podocytes of the filtration barrier, are recently implicated in the pathogenesis of various forms of proteinuric kidney diseases. Indeed, inherited or acquired up‐regulation of TRPC6 activities are suggested to play a role in podocytopathies. Yet, we possess limited information about the regulation of TRPC6 in human podocytes. Therefore, in this study, we aimed at defining how the protein kinase C (PKC) system, one of the key intracellular signalling pathways, regulates TRPC6 function and expression. On human differentiated podocytes, we identified the molecular expressions of both TRPC6 and several PKC isoforms. We also showed that TRPC6 channels are functional since the TRPC6 activator 1‐oleoyl‐2‐acetyl‐sn‐glycerol (OAG) induced Ca2+‐influx to the cells. By assessing the regulatory roles of the PKCs, we found that inhibitors of the endogenous activities of classical and novel PKC isoforms markedly augmented TRPC6 activities. In contrast, activation of the PKC system by phorbol 12‐myristate 13‐acetate (PMA) exerted inhibitory actions on TRPC6 and suppressed its expression. Importantly, PMA treatment markedly down‐regulated the expression levels of PKCα, PKCβ, and PKCη reflecting their activation. Taken together, these results indicate that the PKC system exhibits a ‘tonic’ inhibition on TRPC6 activity in human podocytes suggesting that pathological conditions altering the expression and/or activation patterns of podocyte‐expressed PKCs may influence TRPC6 activity and hence podocyte functions. Therefore, it is proposed that targeted manipulation of certain PKC isoforms might be beneficial in certain proteinuric kidney diseases with altered TRPC6 functions.
Collapse
Affiliation(s)
- Lídia Ambrus
- DE-MTA "Lendület" Cellular Physiology Research Group, Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Attila Oláh
- DE-MTA "Lendület" Cellular Physiology Research Group, Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Tamás Oláh
- Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - György Balla
- Department of Pediatrics, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Moin A Saleem
- Renal Academic Unit, University of Bristol, Bristol, UK
| | - Petronella Orosz
- Department of Pediatrics, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Judit Zsuga
- Department of Health Systems Management and Quality Management for Health Care, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Klára Bíró
- Department of Health Systems Management and Quality Management for Health Care, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - László Csernoch
- Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Tamás Bíró
- DE-MTA "Lendület" Cellular Physiology Research Group, Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary.,Department of Immunology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Medical Faculty, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
22
|
Zang Z, Li S, Zhang W, Chen X, Zheng D, Shu H, Guo W, Zhao B, Shen K, Wei Y, Zheng X, Liu S, Yang H. Expression Patterns of TRPC1 in Cortical Lesions from Patients with Focal Cortical Dysplasia. J Mol Neurosci 2015; 57:265-72. [PMID: 26280213 DOI: 10.1007/s12031-015-0615-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 06/30/2015] [Indexed: 02/05/2023]
Abstract
Focal cortical dysplasia (FCD) is known as a common cause of chronic refractory epilepsy, but the underlying mechanisms of the factors that lead to FCD-related epilepsy are unclear. Previous studies have shown that canonical transient receptor potential channels (TRPCs) might be involved in the process of epileptogenesis. Canonical transient receptor potential channel 1 (TRPC1), which is ubiquitously expressed in the brain, has been shown to be involved in epileptiform bust firing in knockout mice. In this study, we examined the expression of TRPC1 in FCD type Ia (FCDIa), FCD type IIa (FCDIIa), and FCD type IIb (FCDIIb) surgical specimens from patients and age-matched autopsy control samples. Real-time quantitative PCR and western blotting indicated that TRPC1 mRNA and protein levels were increased in FCDIa, FCDIIa, and FCDIIb samples compared to control samples. Immunohistochemistry results revealed that TRPC1 was mainly distributed in microcolumns, dysmorphic neurons, and balloon cells. Further double immunofluorescent staining showed that TRPC1 was co-localized with glutamatergic and GABAergic markers. Taken together, our results demonstrate that the overexpression and specific cellular location of TRPC1 might be related to the epileptogenesis of FCD.
Collapse
Affiliation(s)
- Zhenle Zang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Wei Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Xin Chen
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Dahai Zheng
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Haifeng Shu
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.,Department of Neurosurgery, Chengdu Military General Hospital of PLA, Chengdu, Sichuan, 610083, China
| | - Wei Guo
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Bangyun Zhao
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Kaifeng Shen
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - YuJia Wei
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Xin Zheng
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Shiyong Liu
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| |
Collapse
|
23
|
Grubb DR, Crook B, Ma Y, Luo J, Qian HW, Gao XM, Kiriazis H, Du XJ, Gregorevic P, Woodcock EA. The atypical 'b' splice variant of phospholipase Cβ1 promotes cardiac contractile dysfunction. J Mol Cell Cardiol 2015; 84:95-103. [PMID: 25918049 DOI: 10.1016/j.yjmcc.2015.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/16/2015] [Accepted: 04/20/2015] [Indexed: 10/23/2022]
Abstract
The activity of the early signaling enzyme, phospholipase Cβ1b (PLCβ1b), is selectively elevated in diseased myocardium and activity increases with disease progression. We aimed to establish the contribution of heightened PLCβ1b activity to cardiac pathology. PLCβ1b, the alternative splice variant, PLCβ1a, and a blank virus were expressed in mouse hearts using adeno-associated viral vectors (rAAV6-FLAG-PLCβ1b, rAAV6-FLAG-PLCβ1a, or rAAV6-blank) delivered intravenously (IV). Following viral delivery, FLAG-PLCβ1b was expressed in all of the chambers of the mouse heart and was localized to the sarcolemma. Heightened PLCβ1b expression caused a rapid loss of contractility, 4-6 weeks, that was fully reversed, within 5 days, by inhibition of protein kinase Cα (PKCα). PLCβ1a did not localize to the sarcolemma and did not affect contractile function. Expression of PLCβ1b, but not PLCβ1a, caused downstream dephosphorylation of phospholamban and depletion of the Ca(2+) stores of the sarcoplasmic reticulum. We conclude that heightened PLCβ1b activity observed in diseased myocardium contributes to pathology by PKCα-mediated contractile dysfunction. PLCβ1b is a cardiac-specific signaling system, and thus provides a potential therapeutic target for the development of well-tolerated inotropic agents for use in failing myocardium.
Collapse
Affiliation(s)
- David R Grubb
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Bryony Crook
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Yi Ma
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Jieting Luo
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Hong Wei Qian
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Paul Gregorevic
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia
| | - Elizabeth A Woodcock
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 Victoria, Australia.
| |
Collapse
|
24
|
Veit F, Pak O, Brandes RP, Weissmann N. Hypoxia-dependent reactive oxygen species signaling in the pulmonary circulation: focus on ion channels. Antioxid Redox Signal 2015; 22:537-52. [PMID: 25545236 PMCID: PMC4322788 DOI: 10.1089/ars.2014.6234] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE An acute lack of oxygen in the lung causes hypoxic pulmonary vasoconstriction, which optimizes gas exchange. In contrast, chronic hypoxia triggers a pathological vascular remodeling causing pulmonary hypertension, and ischemia can cause vascular damage culminating in lung edema. RECENT ADVANCES Regulation of ion channel expression and gating by cellular redox state is a widely accepted mechanism; however, it remains a matter of debate whether an increase or a decrease in reactive oxygen species (ROS) occurs under hypoxic conditions. Ion channel redox regulation has been described in detail for some ion channels, such as Kv channels or TRPC6. However, in general, information on ion channel redox regulation remains scant. CRITICAL ISSUES AND FUTURE DIRECTIONS In addition to the debate of increased versus decreased ROS production during hypoxia, we aim here at describing and deciphering why different oxidants, under different conditions, can cause both activation and inhibition of channel activity. While the upstream pathways affecting channel gating are often well described, we need a better understanding of redox protein modifications to be able to determine the complexity of ion channel redox regulation. Against this background, we summarize the current knowledge on hypoxia-induced ROS-mediated ion channel signaling in the pulmonary circulation.
Collapse
Affiliation(s)
- Florian Veit
- 1 Excellence Cluster Cardiopulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL) , Giessen, Germany
| | | | | | | |
Collapse
|
25
|
Bista P, Pawlowski M, Cerina M, Ehling P, Leist M, Meuth P, Aissaoui A, Borsotto M, Heurteaux C, Decher N, Pape HC, Oliver D, Meuth SG, Budde T. Differential phospholipase C-dependent modulation of TASK and TREK two-pore domain K+ channels in rat thalamocortical relay neurons. J Physiol 2014; 593:127-44. [PMID: 25556792 DOI: 10.1113/jphysiol.2014.276527] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 09/23/2014] [Indexed: 01/10/2023] Open
Abstract
KEY POINTS During the behavioural states of sleep and wakefulness thalamocortical relay neurons fire action potentials in high frequency bursts or tonic sequences, respectively. The modulation of specific K(+) channel types, termed TASK and TREK, allows these neurons to switch between the two modes of activity. In this study we show that the signalling lipids phosphatidylinositol 4,5-bisphosphate (PIP2) and diacylglycerol (DAG), which are components of their membrane environment, switch on and shut off TREK and TASK channels, respectively. These channel modulations contribute to a better understanding of the molecular basis of the effects of neurotransmitters such as ACh which are released by the brainstem arousal system. The present report introduces PIP2 and DAG as new elements of signal transduction in the thalamus. The activity of two-pore domain potassium channels (K2P ) regulates the excitability and firing modes of thalamocortical (TC) neurons. In particular, the inhibition of two-pore domain weakly inwardly rectifying K(+) channel (TWIK)-related acid-sensitive K(+) (TASK) channels and TWIK-related K(+) (TREK) channels, as a consequence of the stimulation of muscarinic ACh receptors (MAChRs) which are coupled to phosphoinositide-specific phospholipase C (PLCβ), induces a shift from burst to tonic firing. By using a whole cell patch-clamp approach, the contribution of the membrane-bound second messenger molecules phosphatidylinositol 4,5-bisphosphate (PIP2 ) and diacylglycerol (DAG) acting downstream of PLCβ was probed. The standing outward current (ISO ) was used to monitor the current through TASK and TREK channels in TC neurons. By exploiting different manoeuvres to change the intracellular PIP2 level in TC neurons, we here show that the scavenging of PIP2 (by neomycin) results in an increased muscarinic effect on ISO whereas increased availability of PIP2 (inclusion to the patch pipette; histone-based carrier) decreased muscarinic signalling. The degree of muscarinic inhibition specifically depends on phosphatidylinositol phosphate (PIP) and PIP2 but no other phospholipids (phosphatidic acid, phosphatidylserine). The use of specific blockers revealed that PIP2 is targeting TREK but not TASK channels. Furthermore, we demonstrate that the inhibition of TASK channels is induced by the application of the DAG analogue 1-oleoyl-2-acetyl-sn-glycerol (OAG). Under current clamp conditions the activation of MAChRs and PLCβ as well as the application of OAG resulted in membrane depolarization, while PIP2 application via histone carrier induced a hyperpolarization. These results demonstrate a differential role of PIP2 and DAG in K2P channel modulation in native neurons which allows a fine-tuned inhibition of TREK (via PIP2 depletion) and TASK (via DAG) channels following MAChR stimulation.
Collapse
Affiliation(s)
- Pawan Bista
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Robert-Koch-Straße 27a, D-48149, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhao L, Wang J, Wang L, Liang YT, Chen YQ, Lu WJ, Zhou WL. Remodeling of rat pulmonary artery induced by chronic smoking exposure. J Thorac Dis 2014; 6:818-28. [PMID: 24977008 DOI: 10.3978/j.issn.2072-1439.2014.03.31] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 03/25/2014] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To evaluate the dominant role in rat pulmonary artery (PA) remodeling induced by chronic smoking exposure (CSE). METHODS Thirty-five male Sprague-Dawley (SD) rats were exposed to 36 cigarettes per day, 6 days per week, for 1, 3, or 5 months. Another 35 SD rats were sham-exposed during the same period. Hemodynamic measurement, evaluation of the right ventricular hypertrophy index (RVHI) plus right ventricle-to-weight ratio, and hematoxylin eosin staining was performed. Wall thickness, artery radius, luminal area, and total area were measured morphometrically. Western blotting assessed expression of PPAR-γ BMP4, BMPR2, and TRPC1/4/6 in the artery and lung. Store-operated calcium entry (SOCE) and [Ca(2+)]i were measured using Fura-2 as dye. RESULTS Mean right ventricular pressure increased after 3 months of smoking exposure and continued to increase through 5 months. Right ventricular systolic pressure (RVSP) increased after 3 months of exposure and then stabilized. RVHI increased after 5 months; right ventricle-to-weight ratio was elevated after 3 months and further increased after 5 months. Wall thickness-to-radius ratio does-dependently increased after 3 months through 5 months, in parallel with the decreased luminal area/total area ratio after 5 months. Other changes included the development of inflammatory responses, enlargement of the alveolar spaces, and reductions in the endothelial lining of PAs, proliferative smooth muscle cells, fibroblasts, and adventitia. Moreover, BMP4 and TRPC1/4/6 expression increased to varying degrees in the arteries and lungs of smoking-exposed animals, whereas BMPR expression and SOCE increased only in the arteries, and PPAR-γ was downregulated in both the arteries and lungs. CONCLUSIONS In SD rats, smoking exposure induces pulmonary vascular remodeling. The consequences of increased SOCE include increase in TRPC1/4/6, probably via augmented BMP4 expression, which also contribute to inflammatory responses in the lung. Moreover, interactions between BMP4 and PPAR-γ may play a role in preventing inflammation under normal physiological conditions.
Collapse
Affiliation(s)
- Lei Zhao
- 1 Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou 510182, China ; 2 Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China ; 3 School of Life Science, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jian Wang
- 1 Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou 510182, China ; 2 Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China ; 3 School of Life Science, Sun Yat-Sen University, Guangzhou 510275, China
| | - Lu Wang
- 1 Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou 510182, China ; 2 Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China ; 3 School of Life Science, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu-Ting Liang
- 1 Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou 510182, China ; 2 Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China ; 3 School of Life Science, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu-Qin Chen
- 1 Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou 510182, China ; 2 Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China ; 3 School of Life Science, Sun Yat-Sen University, Guangzhou 510275, China
| | - Wen-Jun Lu
- 1 Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou 510182, China ; 2 Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China ; 3 School of Life Science, Sun Yat-Sen University, Guangzhou 510275, China
| | - Wen-Liang Zhou
- 1 Department of Physiology, School of Basic Science, Guangzhou Medical University, Guangzhou 510182, China ; 2 Guangzhou Institute of Respiratory Disease, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China ; 3 School of Life Science, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
27
|
Renal biopsy: use of biomarkers as a tool for the diagnosis of focal segmental glomerulosclerosis. DISEASE MARKERS 2014; 2014:192836. [PMID: 24719498 PMCID: PMC3955602 DOI: 10.1155/2014/192836] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 12/12/2022]
Abstract
Focal segmental glomerulosclerosis (FSGS) is a glomerulopathy associated with nephrotic syndrome and podocyte injury. FSGS occurs both in children and adults and it is considered the main idiopathic nephrotic syndrome nowadays. It is extremely difficult to establish a morphological diagnosis, since some biopsies lack a considerable quantifiable number of sclerotic glomeruli, given their focal aspect and the fact that FSGS occurs in less than half of the glomeruli. Therefore, many biological molecules have been evaluated as potential markers that would enhance the diagnosis of FSGS. Some of these molecules and receptors are associated with the pathogenesis of FSGS and have potential use in diagnosis.
Collapse
|
28
|
Spahn V, Stein C, Zöllner C. Modulation of transient receptor vanilloid 1 activity by transient receptor potential ankyrin 1. Mol Pharmacol 2014; 85:335-44. [PMID: 24275229 DOI: 10.1124/mol.113.088997] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a nonselective ligand-gated cation channel responding to noxious heat, protons, and chemicals such as capsaicin. TRPV1 is expressed in sensory neurons and plays a critical role in pain associated with tissue injury, inflammation, and nerve lesions. Transient receptor potential ankyrin 1 (TRPA1) is coexpressed with TRPV1. It is activated by compounds that cause a burning sensation (e.g., mustard oil) and, indirectly, by components of the inflammatory milieu eliciting nociceptor excitation and pain hypersensitivity. Previous studies indicate an interaction of TRPV1 and TRPA1 signaling pathways. Here we sought to examine the molecular mechanisms underlying such interactions in nociceptive neurons. We first excluded physical interactions of both channels using radioligand binding studies. By microfluorimetry, electrophysiological experiments, cAMP measurements, and site-directed mutagenesis we found a sensitization of TRPV1 after TRPA1 stimulation with mustard oil in a calcium and cAMP/protein kinase A (PKA)-dependent manner. TRPA1 stimulation enhanced TRPV1 phosphorylation via the putative PKA phosphorylation site serine 116. We also detected calcium-sensitive increased TRPV1 activity after TRPA1 activation in dorsal root ganglion neurons. The inhibition of TRPA1 by HC-030031 (1,2,3,6-tetrahydro-1,3-dimethyl-N-[4-(1-methylethyl)phenyl]-2,6-dioxo-7H-purine-7-acetamide, 2-(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl)-N-(4-isopropylphenyl)acetamide) after its initial stimulation (and the calcium-insensitive TRPA1 mutant D477A) still showed increased capsaicin-induced TRPV1 activity. This excludes a calcium-induced additive TRPA1 current after TRPV1 stimulation. Our study shows sensitization of TRPV1 via activation of TRPA1, which involves adenylyl cyclase, increased cAMP, subsequent translocation and activation of PKA, and phosphorylation of TRPV1 at PKA phosphorylation residues. This suggests that cross-sensitization of TRP channels contributes to enhanced pain sensitivity in inflamed tissues.
Collapse
Affiliation(s)
- Viola Spahn
- Charité, Universitätsmedizin Berlin, Klinik für Anästhesiologie und Operative Intensivmedizin, Berlin, Germany (V.S., C.S., C.Z.); and Universitätsklinikum Hamburg, Eppendorf, Klinik und Poliklinik für Anästhesiologie, Zentrum für Anästhesiologie und Intensivmedizin, Hamburg, Germany (C.Z.)
| | | | | |
Collapse
|
29
|
Zhang X, Trebak M. Transient receptor potential canonical 7: a diacylglycerol-activated non-selective cation channel. Handb Exp Pharmacol 2014; 222:189-204. [PMID: 24756707 DOI: 10.1007/978-3-642-54215-2_8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transient receptor potential canonical 7 (TRPC7) channel is the seventh member of the mammalian TRPC channel family. TRPC7 mRNA, protein, and channel activity have been detected in many tissues and organs from the mouse, rat, and human. TRPC7 has high sequence homology with TRPC3 and TRPC6, and all three channels are activated by membrane receptors that couple to isoforms of phospholipase C (PLC) and mediate non-selective cation currents. TRPC7, along with TRPC3 and TRPC6, can be activated by direct exogenous application of diacylglycerol (DAG) analogues and by pharmacological maneuvers that increase endogenous DAG in cells. TRPC7 shows distinct properties of activation, such as constitutive activity and susceptibility to negative regulation by extracellular Ca(2+) and by protein kinase C. TRPC7 can form heteromultimers with TRPC3 and TRPC6. Although TRPC7 remains one of the least studied TRPC channel, its role in various cell types and physiological and pathophysiological conditions is beginning to emerge.
Collapse
Affiliation(s)
- Xuexin Zhang
- Nanobioscience Constellation, The State University of New York (SUNY), College of Nanoscale Science and Engineering (CNSE), 257 Fuller Road, Albany, NY, 12203, USA
| | | |
Collapse
|
30
|
Zhou FW, Roper SN. TRPC3 mediates hyperexcitability and epileptiform activity in immature cortex and experimental cortical dysplasia. J Neurophysiol 2013; 111:1227-37. [PMID: 24353305 DOI: 10.1152/jn.00607.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuronal hyperexcitability plays an important role in epileptogenesis. Conditions of low extracellular calcium (Ca) or magnesium (Mg) can induce hyperexcitability and epileptiform activity with unclear mechanisms. Transient receptor potential canonical type 3 (TRPC3) channels play a pivotal role in neuronal excitability and are activated in low-Ca and/or low-Mg conditions to depolarize neurons. TRPC3 staining was highly enriched in immature, but very weak in mature, control cortex, whereas it was strong in dysplastic cortex at all ages. Depolarization and susceptibility to epileptiform activity increased with decreasing Ca and Mg. Combinations of low Ca and low Mg induced larger depolarization in pyramidal neurons and greater susceptibility to epileptiform activity in immature and dysplastic cortex than in mature and control cortex, respectively. Intracellular application of anti-TRPC3 antibody to block TRPC3 channels and bath application of the selective TRPC3 inhibitor Pyr3 greatly diminished depolarization in immature control and both immature and mature dysplastic cortex with strong TRPC3 expression. Epileptiform activity was initiated in low Ca and low Mg when synaptic activity was blocked, and Pyr3 completely suppressed this activity. In conclusion, TRPC3 primarily mediates low Ca- and low Mg-induced depolarization and epileptiform activity, and the enhanced expression of TRPC3 could make dysplastic and immature cortex more hyperexcitable and more susceptible to epileptiform activity.
Collapse
Affiliation(s)
- Fu-Wen Zhou
- Department of Neurosurgery and McKnight Brain Institute, University of Florida, Gainesville, Florida
| | | |
Collapse
|
31
|
Narayanan KL, Subramaniam S, Bengston CP, Irmady K, Unsicker K, von Bohlen und Halbach O. Role of transient receptor potential channel 1 (TRPC1) in glutamate-induced cell death in the hippocampal cell line HT22. J Mol Neurosci 2013; 52:425-33. [PMID: 24242951 DOI: 10.1007/s12031-013-0171-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/31/2013] [Indexed: 02/06/2023]
Abstract
Transient receptor potential channel 1 (TRPC1; a cation channel activated by store depletion and/or through an intracellular messenger) is expressed in a variety of tissues, including the brain. To study the physiological function of TRPC1, we investigated the role of endogenously expressed TRPC1 in glutamate-induced cell death, using the murine hippocampal cell line HT22. Knocking down TRPC1 mRNA using TRPC1-shRNA or blocking of TRPC channels using 2-APB (≥200 μM) robustly attenuated glutamate-induced cell death after 24 h of incubation with 5 mM glutamate. Glutamate toxicity in HT22 cells seems to involve metabotropic glutamate receptor mGluR5 since MPEP (2-methyl-6-(phenylethynyl)-pyridine), an mGluR5 antagonist (≥100 μM), abrogated glutamate toxicity. Furthermore, a direct activation of mGluR5 by CHPG [(RS)-chloro-5-hydroxyphenylglycine; 100 μM or 300 μM] promoted HT22 cell death. TRPC1 knock-down markedly reduced CHPG-induced cell death. These observations suggest that glutamate-induced cell death in HT22 cells activates mGluR5 receptors, which significantly increases Ca(2+) influx through TRPC1 channels. TRPC1 knock-down prevented glutamate- and CHPG-induced cell death, suggesting that glutamate-induced toxicity in HT22 cells is mediated through TRPC1 channels and an mGluR5-dependent pathway. Together, this work provides evidence for a novel receptor activation pathway of TRPC1 in glutamate-induced toxicity.
Collapse
Affiliation(s)
- K Lakshmi Narayanan
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, 02129-4404, USA
| | | | | | | | | | | |
Collapse
|
32
|
Stiber JA, Tang Y, Li T, Rosenberg PB. Cytoskeletal regulation of TRPC channels in the cardiorenal system. Curr Hypertens Rep 2013; 14:492-7. [PMID: 23054893 DOI: 10.1007/s11906-012-0313-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transient receptor potential canonical (TRPC) channels have been implicated in several aspects of cardiorenal physiology including regulation of blood pressure, vasoreactivity, vascular remodeling, and glomerular filtration. Gain and loss of function studies also support the role of TRPC channels in adverse remodeling associated with cardiac hypertrophy and heart failure. This review discusses TRP channels in the cardiovascular and glomerular filtration systems and their role in disease pathogenesis. We describe the regulation of gating of TRPC channels in the cardiorenal system as well as the influence on activation of these channels by the underlying cytoskeleton and scaffolding proteins. We then focus on the role of TRP channels in the pathogenesis of adverse cardiac remodeling and as potential therapeutic targets in the treatment of heart failure.
Collapse
Affiliation(s)
- Jonathan A Stiber
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
33
|
Sukumaran P, Löf C, Kemppainen K, Kankaanpää P, Pulli I, Näsman J, Viitanen T, Törnquist K. Canonical transient receptor potential channel 2 (TRPC2) as a major regulator of calcium homeostasis in rat thyroid FRTL-5 cells: importance of protein kinase C δ (PKCδ) and stromal interaction molecule 2 (STIM2). J Biol Chem 2012; 287:44345-60. [PMID: 23144458 DOI: 10.1074/jbc.m112.374348] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mammalian non-selective transient receptor potential cation channels (TRPCs) are important in the regulation of cellular calcium homeostasis. In thyroid cells, including rat thyroid FRTL-5 cells, calcium regulates a multitude of processes. RT-PCR screening of FRTL-5 cells revealed the presence of TRPC2 channels only. Knockdown of TRPC2 using shRNA (shTRPC2) resulted in decreased ATP-evoked calcium peak amplitude and inward current. In calcium-free buffer, there was no difference in the ATP-evoked calcium peak amplitude between control cells and shTRPC2 cells. Store-operated calcium entry was indistinguishable between the two cell lines. Basal calcium entry was enhanced in shTRPC2 cells, whereas the level of PKCβ1 and PKCδ, the activity of sarco/endoplasmic reticulum Ca(2+)-ATPase, and the calcium content in the endoplasmic reticulum were decreased. Stromal interaction molecule (STIM) 2, but not STIM1, was arranged in puncta in resting shTRPC2 cells but not in control cells. Phosphorylation site Orai1 S27A/S30A mutant and non-functional Orai1 R91W attenuated basal calcium entry in shTRPC2 cells. Knockdown of PKCδ with siRNA increased STIM2 punctum formation and enhanced basal calcium entry but decreased sarco/endoplasmic reticulum Ca(2+)-ATPase activity in wild-type cells. Transfection of a truncated, non-conducting mutant of TRPC2 evoked similar results. Thus, TRPC2 functions as a major regulator of calcium homeostasis in rat thyroid cells.
Collapse
Affiliation(s)
- Pramod Sukumaran
- Department of Biosciences, Åbo Akademi University, Tykistökatu 6A, 20520 Turku, Finland
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Harada M, Luo X, Qi XY, Tadevosyan A, Maguy A, Ordog B, Ledoux J, Kato T, Naud P, Voigt N, Shi Y, Kamiya K, Murohara T, Kodama I, Tardif JC, Schotten U, Van Wagoner DR, Dobrev D, Nattel S. Transient receptor potential canonical-3 channel-dependent fibroblast regulation in atrial fibrillation. Circulation 2012; 126:2051-64. [PMID: 22992321 DOI: 10.1161/circulationaha.112.121830] [Citation(s) in RCA: 207] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Fibroblast proliferation and differentiation are central in atrial fibrillation (AF)-promoting remodeling. Here, we investigated fibroblast regulation by Ca(2+)-permeable transient receptor potential canonical-3 (TRPC3) channels. METHODS AND RESULTS Freshly isolated rat cardiac fibroblasts abundantly expressed TRPC3 and had appreciable nonselective cation currents (I(NSC)) sensitive to a selective TPRC3 channel blocker, pyrazole-3 (3 μmol/L). Pyrazole-3 suppressed angiotensin II-induced Ca(2+) influx, proliferation, and α-smooth muscle actin protein expression in fibroblasts. Ca(2+) removal and TRPC3 blockade suppressed extracellular signal-regulated kinase phosphorylation, and extracellular signal-regulated kinase phosphorylation inhibition reduced fibroblast proliferation. TRPC3 expression was upregulated in atria from AF patients, goats with electrically maintained AF, and dogs with tachypacing-induced heart failure. TRPC3 knockdown (based on short hairpin RNA [shRNA]) decreased canine atrial fibroblast proliferation. In left atrial fibroblasts freshly isolated from dogs kept in AF for 1 week by atrial tachypacing, TRPC3 protein expression, currents, extracellular signal-regulated kinase phosphorylation, and extracellular matrix gene expression were all significantly increased. In cultured left atrial fibroblasts from AF dogs, proliferation rates, α-smooth muscle actin expression, and extracellular signal-regulated kinase phosphorylation were increased and were suppressed by pyrazole-3. MicroRNA-26 was downregulated in canine AF atria; experimental microRNA-26 knockdown reproduced AF-induced TRPC3 upregulation and fibroblast activation. MicroRNA-26 has NFAT (nuclear factor of activated T cells) binding sites in the 5' promoter region. NFAT activation increased in AF fibroblasts, and NFAT negatively regulated microRNA-26 transcription. In vivo pyrazole-3 administration suppressed AF while decreasing fibroblast proliferation and extracellular matrix gene expression. CONCLUSIONS TRPC3 channels regulate cardiac fibroblast proliferation and differentiation, likely by controlling the Ca(2+) influx that activates extracellular signal-regulated kinase signaling. AF increases TRPC3 channel expression by causing NFAT-mediated downregulation of microRNA-26 and causes TRPC3-dependent enhancement of fibroblast proliferation and differentiation. In vivo, TRPC3 blockade prevents AF substrate development in a dog model of electrically maintained AF. TRPC3 likely plays an important role in AF by promoting fibroblast pathophysiology and is a novel potential therapeutic target.
Collapse
Affiliation(s)
- Masahide Harada
- Department of Medicine and Research Center, Montreal Heart Institute and Universite´ de Montre´al, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Molnar T, Barabas P, Birnbaumer L, Punzo C, Kefalov V, Križaj D. Store-operated channels regulate intracellular calcium in mammalian rods. J Physiol 2012; 590:3465-81. [PMID: 22674725 DOI: 10.1113/jphysiol.2012.234641] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Exposure to daylight closes cyclic nucleotide-gated (CNG) and voltage-operated Ca(2+) -permeable channels in mammalian rods. The consequent lowering of the cytosolic calcium concentration ([Ca(2+)](i)), if protracted, can contribute to light-induced damage and apoptosis in these cells. We here report that mouse rods are protected against prolonged lowering of [Ca(2+)](i) by store-operated Ca(2+) entry (SOCE). Ca(2+) stores were depleted in Ca(2+)-free saline supplemented with the endoplasmic reticulum (ER) sequestration blocker cyclopiazonic acid. Store depletion elicited [Ca(2+)](i) signals that exceeded baseline [Ca(2+)](i) by 5.9 ± 0.7-fold and were antagonized by an inhibitory cocktail containing 2-APB, SKF 96365 and Gd(3+). Cation influx through SOCE channels was sufficient to elicit a secondary activation of L-type voltage-operated Ca2+ entry. We also found that TRPC1, the type 1 canonical mammalian homologue of the Drosophila photoreceptor TRP channel, is predominantly expressed within the outer nuclear layer of the retina. Rod loss in Pde6b(rdl) (rd1), Chx10/Kip1(-/-rdl) and Elovl4(TG2) dystrophic models was associated with ∼70% reduction in Trpc1 mRNA content whereas Trpc1 mRNA levels in rodless cone-full Nrl(-/-) retinas were decreased by ∼50%. Genetic ablation of TRPC1 channels, however, had no effect on SOCE, the sensitivity of the rod phototransduction cascade or synaptic transmission at rod and cone synapses. Thus, we localized two new mechanisms, SOCE and TRPC1, to mammalian rods and characterized the contribution of SOCE to Ca(2+) homeostasis. By preventing the cytosolic [Ca(2+)](i) from dropping too low under sustained saturating light conditions, these signalling pathways may protect Ca(2+)-dependent mechanisms within the ER and the cytosol without affecting normal rod function.
Collapse
Affiliation(s)
- Tünde Molnar
- Department of Ophthalmology & Visual Sciences, Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | | | | | | |
Collapse
|
36
|
Anfinogenova Y, Brett SE, Walsh MP, Harraz OF, Welsh DG. Do TRPC-like currents and G protein-coupled receptors interact to facilitate myogenic tone development? Am J Physiol Heart Circ Physiol 2011; 301:H1378-88. [DOI: 10.1152/ajpheart.00460.2011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The objective of this study was to determine whether Gq/11-coupled receptor activation can enhance the mechanosensitivity of a canonical transient receptor potential (TRPC)-like current and consequently the myogenic responsiveness of rat anterior cerebral arteries. Initial patch-clamp experiments revealed the presence of a basal cation current in isolated smooth muscle cells that displayed evidence of double rectification, which was blocked by trivalent cations (Gd3+ and La3+). PCR analysis identified the expression of TRPC1, 3, 6 and 7 mRNA and, characteristic of TRPC-like current, the whole-cell conductance was insensitive to a Na+-dependent transport (amiloride), TRP vanilloid (ruthenium red), and chloride channel (DIDS, niflumic acid, and flufenamate) inhibitors. One notable exception was tamoxifen, which elicited a dual effect, blocking or activating the TRPC-like current at 1 and 10 μM, respectively. This TRPC-like current was augmented by constrictor agonists (uridine 5′-triphosphate and U46619) or hyposmotic challenge (303 to 223 mOsm/l), a mechanical stimulus. Although each stimulus was effective alone, smooth muscle cells pretreated with agonist did not augment the whole-cell response to hyposmotic challenge. Consistent with these electrophysiological recordings, functional experiments revealed that neither UTP nor U46619 enhanced the sensitivity of intact cerebral arteries to hyposmotic challenge or elevated intravascular pressure. In summary, this study found no evidence that Gq/11-coupled receptor activation augments the mechanosensitivity of a TRPC-like current and consequently the myogenic responsiveness of anterior cerebral arteries.
Collapse
Affiliation(s)
| | | | - Michael P. Walsh
- Biochemistry and Molecular Biology, Hotchkiss Brain and Libin Cardiovascular Institutes, University of Calgary, Alberta, Canada
| | | | | |
Collapse
|
37
|
Meuchel LW, Stewart A, Smelter DF, Abcejo AJ, Thompson MA, Zaidi SIA, Martin RJ, Prakash YS. Neurokinin-neurotrophin interactions in airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2011; 301:L91-8. [PMID: 21515660 DOI: 10.1152/ajplung.00320.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Neurally derived tachykinins such as substance P (SP) play a key role in modulating airway contractility (especially with inflammation). Separately, the neurotrophin brain-derived neurotrophic factor (BDNF; potentially derived from nerves as well as airway smooth muscle; ASM) and its tropomyosin-related kinase receptor, TrkB, are involved in enhanced airway contractility. In this study, we hypothesized that neurokinins and neurotrophins are linked in enhancing intracellular Ca(2+) concentration ([Ca(2+)](i)) regulation in ASM. In rat ASM cells, 24 h exposure to 10 nM SP significantly increased BDNF and TrkB expression (P < 0.05). Furthermore, [Ca(2+)](i) responses to 1 μM ACh as well as BDNF (30 min) effects on [Ca(2+)](i) regulation were enhanced by prior SP exposure, largely via increased Ca(2+) influx (P < 0.05). The enhancing effect of SP on BDNF signaling was blunted by the neurokinin-2 receptor antagonist MEN-10376 (1 μM, P < 0.05) to a greater extent than the neurokinin-1 receptor antagonist RP-67580 (5 nM). Chelation of extracellular BDNF (chimeric TrkB-F(c); 1 μg/ml), as well as tyrosine kinase inhibition (100 nM K252a), substantially blunted SP effects (P < 0.05). Overnight (24 h) exposure of ASM cells to 50% oxygen increased BDNF and TrkB expression and potentiated both SP- and BDNF-induced enhancement of [Ca(2+)](i) (P < 0.05). These results suggest a novel interaction between SP and BDNF in regulating agonist-induced [Ca(2+)](i) regulation in ASM. The autocrine mechanism we present here represents a new area in the development of bronchoconstrictive reflex response and airway hyperreactive disorders.
Collapse
Affiliation(s)
- Lucas W Meuchel
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wenning AS, Neblung K, Strauß B, Wolfs MJ, Sappok A, Hoth M, Schwarz EC. TRP expression pattern and the functional importance of TRPC3 in primary human T-cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:412-23. [DOI: 10.1016/j.bbamcr.2010.12.022] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 12/22/2010] [Accepted: 12/23/2010] [Indexed: 11/16/2022]
|
39
|
TRP channels in the cardiopulmonary vasculature. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:781-810. [PMID: 21290327 DOI: 10.1007/978-94-007-0265-3_41] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transient receptor potential (TRP) channels are expressed in almost every human tissue, including the heart and the vasculature. They play unique roles not only in physiological functions but, if over-expressed, also in pathophysiological disease states. Cardiovascular diseases are the leading cause of death in the industrialized countries. Therefore, TRP channels are attractive drug targets for more effective pharmacological treatments of these diseases. This review focuses on three major cell types of the cardiovascular system: cardiomyocytes as well as smooth muscle cells and endothelial cells from the systemic and pulmonary circulation. TRP channels initiate multiple signals in all three cell types (e.g. contraction, migration) and are involved in gene transcription leading to cell proliferation or cell death. Identification of their genes has significantly improved our knowledge of multiple signal transduction pathways in these cells. Some TRP channels are important cellular sensors and are mostly permeable to Ca(2+), while most other TRP channels are receptor activated and allow for the entry of Na(+), Ca(2+) and Mg(2+). Physiological functions of TRPA, TRPC, TRPM, TRPP and TRPV channels in the cardiovascular system, dissected by down-regulating channel activity in isolated tissues or by the analysis of gene-deficient mouse models, are reviewed. The involvement of TRPs as homomeric or heteromeric channels in pathophysiological processes in the cardiovascular system like heart failure, cardiac hypertrophy, hypertension as well as edema formation by increased endothelial permeability will be discussed.
Collapse
|
40
|
Expression and physiological roles of TRP channels in smooth muscle cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:687-706. [PMID: 21290322 DOI: 10.1007/978-94-007-0265-3_36] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Smooth muscles are widely distributed in mammal body through various systems such as circulatory, respiratory, gastro-intestinal and urogenital systems. The smooth muscle cell (SMC) is not only a contractile cell but is able to perform other important functions such as migration, proliferation, production of cytokines, chemokines, extracellular matrix proteins, growth factors and cell surface adhesion molecules. Thus, SMC appears today as a fascinating cell with remarkable plasticity that contributes to its roles in physiology and disease. Most of the SMC functions are dependent on a key event: the increase in intracellular calcium concentration ([Ca(2+)](i)). Calcium entry from the extracellular space is a major step in the elevation of [Ca(2+)](i) in SMC and involves a variety of plasmalemmal calcium channels, among them is the superfamily of transient receptor potential (TRP) proteins. TRPC (canonical), TRPM (melastatin), TRPV (vanilloid) and TRPP (polycystin), are widely expressed in both visceral (airways, gastrointestinal tract, uterus) and vascular (systemic and pulmonary circulation) smooth muscles. Mainly, TRPC, TRPV and TRPM are implicated in a variety of physiological and pathophysiological processes such as: SMC contraction, relaxation, growth, migration and proliferation; control of blood pressure, arterial myogenic tone, pulmonary hypertension, intestinal motility, gastric acidity, uterine activity during parturition and labor. Thus it is becoming evident that TRP are major element of SMC calcium homeostasis and, thus, appear as novel drug targets for a better management of diseases originating from SMC dysfunction.
Collapse
|
41
|
Evidence for a supportive role of classical transient receptor potential 6 (TRPC6) in the exploration behavior of mice. Physiol Behav 2010; 102:245-50. [PMID: 21059368 DOI: 10.1016/j.physbeh.2010.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 10/11/2010] [Accepted: 11/01/2010] [Indexed: 11/21/2022]
Abstract
Non-selective classical transient receptor potential (TRPC) cation channels share important roles in processes of neuronal development and function. To test the influence of TRPC6 activity on behavior, we developed a TRPC6-deficient (TRPC6(-/-)) mouse model in a BALB/c genetic background. Both, TRPC6(-/-) and wild-type (WT) mice were analyzed first for their general health and reflex status (modified SHIRPA protocol) and then in three different behavioral tests (marble-burying test, square open field and elevated star maze). No abnormalities were detected in the SHIRPA protocol. Most interestingly, TRPC6(-/-) mice showed no significant differences in anxiety in a marble-burying test, but demonstrated reduced exploration in the square open field and the elevated star maze. Therefore, TRPC6 channel activity may play a yet unknown role for exploration behavior.
Collapse
|
42
|
Bomben VC, Sontheimer H. Disruption of transient receptor potential canonical channel 1 causes incomplete cytokinesis and slows the growth of human malignant gliomas. Glia 2010; 58:1145-56. [PMID: 20544850 DOI: 10.1002/glia.20994] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite decades of research, primary brain tumors, gliomas, lack effective treatment options and present a huge clinical challenge. Particularly, the most malignant subtype, Glioblastoma multiforme, proliferates extensively and cells often undergo incomplete cell divisions, resulting in multinucleated cells. We now present evidence that multinucleated glioma cells result from the functional loss of transient receptor potential canonical 1 (TRPC1) channels, plasma membrane proteins involved in agonist-induced calcium entry and reloading of intracellular Ca(2+) stores. Pharmacological inhibition or shRNA mediated suppression of TRPC1 causes loss of functional channels and store-operated calcium entry in D54MG glioma cells. This is associated with reduced cell proliferation and, frequently, with incomplete cell division. The resulting multinucleated cells are reminiscent of those found in patient biopsies. In a flank tumor model, tumor size was significantly decreased when TRPC1 expression was disrupted using a doxycycline inducible shRNA knockdown approach. These results suggest that TRPC1 channels play an important role in glioma cell division most likely by regulating calcium signaling during cytokinesis.
Collapse
Affiliation(s)
- Valerie C Bomben
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
43
|
Fuchs B, Sommer N, Dietrich A, Schermuly RT, Ghofrani HA, Grimminger F, Seeger W, Gudermann T, Weissmann N. Redox signaling and reactive oxygen species in hypoxic pulmonary vasoconstriction. Respir Physiol Neurobiol 2010; 174:282-91. [PMID: 20801235 DOI: 10.1016/j.resp.2010.08.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 08/20/2010] [Accepted: 08/20/2010] [Indexed: 11/16/2022]
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is an essential physiological mechanism of the lung that matches blood perfusion with alveolar ventilation to optimize gas exchange. Perturbations of HPV, as may occur in pneumonia or adult respiratory distress syndrome, can cause life-threatening hypoxemia. Despite intensive research for decades, the molecular mechanisms of HPV have not been fully elucidated. Reactive oxygen species (ROS) and changes in the cellular redox state are proposed to link O2 sensing and pulmonary arterial smooth muscle cell contraction underlying HPV. In this regard, mitochondria and NAD(P)H oxidases are discussed as sources of ROS. However, there is controversy whether ROS levels decrease or increase during hypoxia. With this background we summarize the current knowledge on the role of ROS and redox state in HPV.
Collapse
Affiliation(s)
- Beate Fuchs
- Excellence Cluster Cardio-Pulmonary System, University of Giessen Lung Center, Department of Internal Medicine II, Justus-Liebig-University Giessen, Aulweg 130, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Foster RR, Welsh GI, Satchell SC, Marlow RD, Wherlock MD, Pons D, Mathieson PW, Bates DO, Saleem MA. Functional distinctions in cytosolic calcium regulation between cells of the glomerular filtration barrier. Cell Calcium 2010; 48:44-53. [PMID: 20674014 PMCID: PMC2937225 DOI: 10.1016/j.ceca.2010.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 06/29/2010] [Accepted: 06/30/2010] [Indexed: 12/22/2022]
Abstract
The importance of intracellular calcium ([Ca(2+)]i) regulation in the glomerular filtration barrier (GFB) has recently been highlighted by mutations in the cation channel TRPC6, resulting in a renal-specific phenotype. We examined the effects of FFA, a tool that can activate TRPC6, on [Ca(2+)]i in human conditionally immortalised glomerular endothelial cells (ciGEnC) and human podocytes (ciPod) that form the GFB. Changes in [Ca(2+)]i stimulated by FFA were measured in Fura 2-AM loaded cells. In GEnC, cell activation by FFA was dependent on external Ca(2+), yet in ciPod it was not. Depletion of internal Ca(2+) stores with thapsigargin did not affect cell activation by FFA in ciGEnC, but inhibited it in ciPod in a nephrin-dependent manner, demonstrated using nephrin deficient (ND) ciPod in conjunction with nephrin rescue experiments. FFA induced [Ca(2+)]i store release in ciPod, but not in ciGEnC or ND ciPod. In parallel, there were differences in the localisation of overexpressed TRPC6 between ciGEnC and ciPod. Furthermore, co-transfection of nephrin with TRPC6 in HEK293 cells reduced the FFA-induced increase in [Ca(2+)]i and nephrin clustering altered TRPC6 distribution. In conclusion, cell activation by FFA in podocytes stimulates the opening of a Ca(2+) channel, probably TRPC6, in a nephrin-dependent manner with a different activation profile to GEnC.
Collapse
|
45
|
Bardell TK, Barker EL. Activation of TRPC6 channels promotes endocannabinoid biosynthesis in neuronal CAD cells. Neurochem Int 2010; 57:76-83. [PMID: 20466028 DOI: 10.1016/j.neuint.2010.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 04/29/2010] [Accepted: 05/01/2010] [Indexed: 12/13/2022]
Abstract
Calcium influx activates biosynthesis of the endogenous cannabinoids 2-arachidonyl glycerol (2-AG) and anandamide (AEA). The calcium channel involved with endocannabinoid synthesis and release in neurons is still unknown. The canonical TRP (TRPC) channels are calcium-permeable channels that are a homology-based subdivision of the broader class of TRP channels. TRPC3, 6, and 7 are G-protein-gated non-selective cation channels that have been localized to lipid rafts and shown to colocalize with caveolin 1. Because endocannabinoid synthesis has been found to occur "on demand" in a calcium-dependent manner and has been linked to lipid rafts, we explored the potential role of transient receptor potential (TRP) channels in this process. Previously, we observed that after metabolism AEA and arachidonic acid (ArA) can be recycled into new endocannabinoid molecules. Consistent with these previous findings, we found that Cath.a differentiated (CAD) cells pretreated with radiolabeled ArA exhibited a robust increase in 2-AG release in response to TRPC stimulation with the diacylglycerol (DAG) analogue, 1-oleoyl-2-acetyl-sn-glycerol (OAG). Furthermore, cells pretreated with [(3)H]AEA produced a significant amount of AEA and 2-AG upon stimulation of TRPC channels. This process was not mediated through protein kinase C activation. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis revealed that only TRPC6 was present in the CAD cells. siRNA-induced knockdown of TRPC6 in the CAD cells abolished OAG-stimulated production of the endocannabionids. This evidence suggests that TRPC6 may be capable of promoting endocannabinoid synthesis in neuronal cells.
Collapse
Affiliation(s)
- Tamera K Bardell
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University School of Pharmacy and Pharmaceutical Sciences, West Lafayette, IN 47907-2091, United States
| | | |
Collapse
|
46
|
Abstract
Many ion channels and transporters are involved in the filtration, secretion, and resorption of electrolytes by the kidney. In recent years, the superfamily of transient receptor potential (TRP) ion channels have received deserved attention because mutated TRP channels are linked to human kidney diseases. This review focuses on two TRP members--TRPC6 and TRPM6--and their functions in the kidney. Gain-of-function mutations in TRPC6 are the cause for progressive kidney failure with urinary protein loss such as FSGS. Thus, TRPC6 is an essential signaling component in a functional slit diaphragm formed by podocytes around the glomerular capillaries. Loss-of-function mutations in TRPM6 are a molecular cause of hypomagnesemia with secondary hypocalcemia, suggesting that TRPM6 is critically involved in transcellular Mg2+ transport in the kidney. Here, we highlight how recent studies analyzing function and expression of these channels in the kidney improve our mechanistic understanding of TRP channel function in general and pave the way to new, promising therapeutic strategies to target kidney diseases such as FSGS and hypomagnesemia with secondary hypocalcemia.
Collapse
Affiliation(s)
- Alexander Dietrich
- Institute of Pharmacology and Toxicology, School of Medicine, University of Marburg, Marburg, Germany
| | | | | |
Collapse
|
47
|
Fuchs B, Dietrich A, Gudermann T, Kalwa H, Grimminger F, Weissmann N. The role of classical transient receptor potential channels in the regulation of hypoxic pulmonary vasoconstriction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 661:187-200. [PMID: 20204731 DOI: 10.1007/978-1-60761-500-2_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is an essential mechanism of the lung matching blood perfusion to ventilation during local alveolar hypoxia. HPV thus optimizes pulmonary gas exchange. In contrast chronic and generalized hypoxia leads to pulmonary vascular remodeling with subsequent pulmonary hypertension and right heart hypertrophy. Among other non-selective cation channels, the family of classical transient receptor potential channels (TRPC) has been shown to be expressed in pulmonary arterial smooth muscle cells. Among this family, TRPC6 is essential for the regulation of acute HPV in mice. Against this background, in this chapter we give an overview about the TRPC family and their role in HPV.
Collapse
Affiliation(s)
- B Fuchs
- University of Giessen Lung Center (UGLC), Justus-Liebig-University Giessen, Giessen, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Hill MA, Meininger GA, Davis MJ, Laher I. Therapeutic potential of pharmacologically targeting arteriolar myogenic tone. Trends Pharmacol Sci 2009; 30:363-74. [PMID: 19541373 DOI: 10.1016/j.tips.2009.04.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 04/28/2009] [Accepted: 04/28/2009] [Indexed: 01/05/2023]
Abstract
The arteriolar myogenic response, which is defined as vasoconstriction to increases in intraluminal pressure and, conversely, dilation to a reduction in pressure, is key in the setting of vascular resistance, local control of microvascular blood flow through autoregulation, and in the control of capillary hydrostatic pressure. Although considerable progress has been made in the quest for understanding the underlying sensory apparatus and cellular mechanisms, fundamental questions remain - particularly if this pathway is to be considered as a target for novel strategies of pharmacological intervention. We propose that an ability to 're-set' myogenic tone would enable modification of systemic vascular resistance and pressure while at the same time preserving existing interactions with neurohumoral regulatory mechanisms. The challenge, therefore, is to identify steps unique to the myogenic signaling pathway to enable specific pharmacological targeting.
Collapse
Affiliation(s)
- Michael A Hill
- Dalton Cardiovascular Research Center and Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA.
| | | | | | | |
Collapse
|
49
|
Foster RR, Zadeh MAH, Welsh GI, Satchell SC, Ye Y, Mathieson PW, Bates DO, Saleem MA. Flufenamic acid is a tool for investigating TRPC6-mediated calcium signalling in human conditionally immortalised podocytes and HEK293 cells. Cell Calcium 2009; 45:384-90. [PMID: 19232718 DOI: 10.1016/j.ceca.2009.01.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 01/07/2009] [Accepted: 01/15/2009] [Indexed: 01/09/2023]
Abstract
Mutations in the cation channel TRPC6 result in a renal-specific phenotype of familial nephrotic syndrome, affecting intracellular calcium ([Ca(2+)](i)) signalling in the glomerular podocyte. Tools to study native TRPC6 activity are scarce, although there has been recent success with flufenamic acid (FFA). We confirm the specificity of FFA for TRPC6 both in an artificial expression system and in a human conditionally immortalised podocyte cell line (ciPod). Cells were loaded with fura-2AM and changes in intracellular calcium ([Ca(2+)](i)) were calculated. 200microM FFA induced an increase in [Ca(2+)](i) in HEK293 cells with native TRPC6 expression, which was enhanced by overexpression of TRPC6 and completely blocked in the absence of extracellular calcium. Expressed TRPC7 did not significantly affect the response to FFA whereas expressed TRPC3 reduced it. FFA also induced an increase ciPod in [Ca(2+)](i), which was inhibited using SKF96365 and 2-APB, but not indomethacin. In ciPod, adenovirus (Ad-v) wild type (WT) TRPC6 increased [Ca(2+)](i) activity to FFA compared to native TRPC6, whereas activity was significantly reduced with Ad-v dominant negative (DN) TRPC6. The niflumic acid (NFA) induced increase in [Ca(2+)](i) in ciPod was not affected by Ad-v TRPC6 DN, and in HEK293 cells was not affected by WT TRPC6. In conclusion, FFA activates TRPC6 [Ca(2+)](i) signalling in both ciPod and HEK293 cells independently of TRPC3 and TRPC7, and independently of properties of the fenamate family.
Collapse
|
50
|
Hirschler-Laszkiewicz I, Tong Q, Conrad K, Zhang W, Flint WW, Barber AJ, Barber DL, Cheung JY, Miller BA. TRPC3 activation by erythropoietin is modulated by TRPC6. J Biol Chem 2009; 284:4567-81. [PMID: 19074769 PMCID: PMC2640975 DOI: 10.1074/jbc.m804734200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 12/09/2008] [Indexed: 11/06/2022] Open
Abstract
Regulation of intracellular calcium ([Ca(2+)](i)) by erythropoietin (Epo) is an essential part of signaling pathways controlling proliferation and differentiation of erythroid progenitors, but regulatory mechanisms are largely unknown. TRPC3 and the homologous TRPC6 are two members of the transient receptor potential channel (TRPC) superfamily that are expressed on normal human erythroid precursors. Here we show that TRPC3 expression increases but TRPC6 decreases during erythroid differentiation. This is associated with a significantly greater increase in [Ca(2+)](i) in response to Epo stimulation, suggesting that the ratio of TRPC3/TRPC6 is physiologically important. In HEK 293T cells heterologously expressing TRPC and erythropoietin receptor (Epo-R), Epo stimulated an increase in [Ca(2+)](i) through TRPC3 but not TRPC6. Replacement of the C terminus of TRPC3 with the TRPC6 C terminus (TRPC3-C6C) resulted in loss of activation by Epo. In contrast, substitution of the C terminus of TRPC6 with that of TRPC3 (TRPC6-C3C) resulted in an increase in [Ca(2+)](i) in response to Epo. Substitution of the N termini had no effect. Domains in the TRPC3 C terminus between amino acids 671 and 746 are critical for the response to Epo. Epo-R and phospholipase Cgamma associated with TRPC3, and these interactions were significantly reduced with TRPC6 and TRPC3-C6C chimeras. TRPC3 and TRPC6 form heterotetramers. Coexpression of TRPC6 or C3/C6 chimeras with TRPC3 and Epo-R inhibited the Epo-stimulated increase in [Ca(2+)](i). In a heterologous expression system, Epo stimulation increased cell surface expression of TRPC3, which was inhibited by TRPC6. However, in primary erythroblasts, an increase in TRPC3 cell surface expression was not observed in erythroblasts in which Epo stimulated an increase in [Ca(2+)](i), demonstrating that increased membrane insertion of TRPC3 is not required. These data demonstrate that TRPC6 regulates TRPC3 activation by Epo. Endogenously, regulation of TRPC3 by TRPC6 may primarily be through modulation of signaling mechanisms, including reduced interaction of TRPC6 with phospholipase Cgamma and Epo-R.
Collapse
Affiliation(s)
- Iwona Hirschler-Laszkiewicz
- Department of Pediatrics, the Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|