1
|
Khare P, Chand J, Ptakova A, Liguori R, Ferrazzi F, Bishnoi M, Vlachova V, Zimmermann K. The TRPC5 receptor as pharmacological target for pain and metabolic disease. Pharmacol Ther 2024; 263:108727. [PMID: 39384022 DOI: 10.1016/j.pharmthera.2024.108727] [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: 06/06/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
The transient receptor potential canonical (TRPC) channels are a group of highly homologous nonselective cation channels from the larger TRP channel family. They have the ability to form homo- and heteromers with varying degrees of calcium (Ca2+) permeability and signalling properties. TRPC5 is the one cold-sensitive among them and likewise facilitates the influx of extracellular Ca2+ into cells to modulate neuronal depolarization and integrate various intracellular signalling pathways. Recent research with cryo-electron microscopy revealed its structure, along with clear insight into downstream signalling and protein-protein interaction sites. Investigations using global and conditional deficient mice revealed the involvement of TRPC5 in metabolic diseases, energy balance, thermosensation and conditions such as osteoarthritis, rheumatoid arthritis, and inflammatory pain including opioid-induced hyperalgesia and hyperalgesia following tooth decay and pulpitis. This review provides an update on recent advances in our understanding of the role of TRPC5 with focus on metabolic diseases and pain.
Collapse
Affiliation(s)
- Pragyanshu Khare
- Department of Anesthesiology, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany; Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Jagdish Chand
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Alexandra Ptakova
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Renato Liguori
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fulvia Ferrazzi
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mahendra Bishnoi
- TR(i)P for Health Laboratory Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector (Knowledge City), Punjab, India
| | - Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Katharina Zimmermann
- Department of Anesthesiology, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
2
|
Bernal L, Sotelo-Hitschfeld P, König C, Sinica V, Wyatt A, Winter Z, Hein A, Touska F, Reinhardt S, Tragl A, Kusuda R, Wartenberg P, Sclaroff A, Pfeifer JD, Ectors F, Dahl A, Freichel M, Vlachova V, Brauchi S, Roza C, Boehm U, Clapham DE, Lennerz JK, Zimmermann K. Odontoblast TRPC5 channels signal cold pain in teeth. SCIENCE ADVANCES 2021; 7:7/13/eabf5567. [PMID: 33771873 PMCID: PMC7997515 DOI: 10.1126/sciadv.abf5567] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/09/2021] [Indexed: 05/21/2023]
Abstract
Teeth are composed of many tissues, covered by an inflexible and obdurate enamel. Unlike most other tissues, teeth become extremely cold sensitive when inflamed. The mechanisms of this cold sensation are not understood. Here, we clarify the molecular and cellular components of the dental cold sensing system and show that sensory transduction of cold stimuli in teeth requires odontoblasts. TRPC5 is a cold sensor in healthy teeth and, with TRPA1, is sufficient for cold sensing. The odontoblast appears as the direct site of TRPC5 cold transduction and provides a mechanism for prolonged cold sensing via TRPC5's relative sensitivity to intracellular calcium and lack of desensitization. Our data provide concrete functional evidence that equipping odontoblasts with the cold-sensor TRPC5 expands traditional odontoblast functions and renders it a previously unknown integral cellular component of the dental cold sensing system.
Collapse
Affiliation(s)
- Laura Bernal
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
- Departamento de Biología de Sistemas, Facultad de Medicina, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Pamela Sotelo-Hitschfeld
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
- Institute of Physiology, Faculty of Medicine and Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Christine König
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Viktor Sinica
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Amanda Wyatt
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Zoltan Winter
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Alexander Hein
- HHMI, Cardiovascular Division, Boston Children's Hospital, and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Filip Touska
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Susanne Reinhardt
- Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Aaron Tragl
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Ricardo Kusuda
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Philipp Wartenberg
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Allen Sclaroff
- Department of Otolaryngology, Washington University School of Medicine, St Louis, MO, USA
| | - John D Pfeifer
- Department of Pathology, Washington University School of Medicine, St Louis, MO, USA
| | - Fabien Ectors
- FARAH Mammalian Transgenics Platform, Liège University, Liège, Belgium
| | - Andreas Dahl
- Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Marc Freichel
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Sebastian Brauchi
- Institute of Physiology, Faculty of Medicine and Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Millennium Nucleus of Ion Channel-associated Diseases (MiNICAD), Santiago, Chile
| | - Carolina Roza
- Departamento de Biología de Sistemas, Facultad de Medicina, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - David E Clapham
- HHMI, Cardiovascular Division, Boston Children's Hospital, and Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| | - Jochen K Lennerz
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
| | - Katharina Zimmermann
- Department of Anesthesiology, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany.
| |
Collapse
|
3
|
Chen X, Sooch G, Demaree IS, White FA, Obukhov AG. Transient Receptor Potential Canonical (TRPC) Channels: Then and Now. Cells 2020; 9:E1983. [PMID: 32872338 PMCID: PMC7565274 DOI: 10.3390/cells9091983] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Twenty-five years ago, the first mammalian Transient Receptor Potential Canonical (TRPC) channel was cloned, opening the vast horizon of the TRPC field. Today, we know that there are seven TRPC channels (TRPC1-7). TRPCs exhibit the highest protein sequence similarity to the Drosophila melanogaster TRP channels. Similar to Drosophila TRPs, TRPCs are localized to the plasma membrane and are activated in a G-protein-coupled receptor-phospholipase C-dependent manner. TRPCs may also be stimulated in a store-operated manner, via receptor tyrosine kinases, or by lysophospholipids, hypoosmotic solutions, and mechanical stimuli. Activated TRPCs allow the influx of Ca2+ and monovalent alkali cations into the cytosol of cells, leading to cell depolarization and rising intracellular Ca2+ concentration. TRPCs are involved in the continually growing number of cell functions. Furthermore, mutations in the TRPC6 gene are associated with hereditary diseases, such as focal segmental glomerulosclerosis. The most important recent breakthrough in TRPC research was the solving of cryo-EM structures of TRPC3, TRPC4, TRPC5, and TRPC6. These structural data shed light on the molecular mechanisms underlying TRPCs' functional properties and propelled the development of new modulators of the channels. This review provides a historical overview of the major advances in the TRPC field focusing on the role of gene knockouts and pharmacological tools.
Collapse
Affiliation(s)
- Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Gagandeep Sooch
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
| | - Isaac S. Demaree
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
| | - Fletcher A. White
- The Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G. Obukhov
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
4
|
Dattilo M, Penington NJ, Williams K. Regulation of TRPC5 currents by intracellular ATP: Single channel studies. J Cell Physiol 2020; 235:7056-7066. [PMID: 31994734 DOI: 10.1002/jcp.29602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/10/2020] [Indexed: 11/09/2022]
Abstract
TRPC5 channels are nonselective cation channels activated by G-protein-coupled receptors. It was previously found that recombinant TRPC5 currents are inhibited by intracellular ATP, when studied by whole-cell patch-clamp recording. In the present study, we investigated the mechanism of ATP inhibition at the single-channel level using patches from HEK-293 cells transiently transfected with TRPC5 and the M1 muscarinic receptor. In inside-out patches, application of ATP to the intracellular face of the membrane reduced TRPC5 channel activity at both positive and negative potentials without affecting the unitary current amplitude or open dwell time of the channel. The effect of ATP was rapidly reversible. These results suggest that ATP may bind to the channel protein and affect the ability of the channel to open or to remain in an open, nondesensitized state. The activity of TRPC5 channels may be influenced by cellular metabolism via changes in ATP levels.
Collapse
Affiliation(s)
- Michael Dattilo
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York.,Program in Neural and Behavioral Science and Robert F. Furchgott, Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - Nicholas J Penington
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York.,Program in Neural and Behavioral Science and Robert F. Furchgott, Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - Keith Williams
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York.,Program in Neural and Behavioral Science and Robert F. Furchgott, Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| |
Collapse
|
5
|
Wang H, Cheng X, Tian J, Xiao Y, Tian T, Xu F, Hong X, Zhu MX. TRPC channels: Structure, function, regulation and recent advances in small molecular probes. Pharmacol Ther 2020; 209:107497. [PMID: 32004513 DOI: 10.1016/j.pharmthera.2020.107497] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/14/2020] [Indexed: 02/08/2023]
Abstract
Transient receptor potential canonical (TRPC) channels constitute a group of receptor-operated calcium-permeable nonselective cation channels of the TRP superfamily. The seven mammalian TRPC members, which can be further divided into four subgroups (TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7) based on their amino acid sequences and functional similarities, contribute to a broad spectrum of cellular functions and physiological roles. Studies have revealed complexity of their regulation involving several components of the phospholipase C pathway, Gi and Go proteins, and internal Ca2+ stores. Recent advances in cryogenic electron microscopy have provided several high-resolution structures of TRPC channels. Growing evidence demonstrates the involvement of TRPC channels in diseases, particularly the link between genetic mutations of TRPC6 and familial focal segmental glomerulosclerosis. Because TRPCs were discovered by the molecular identity first, their pharmacology had lagged behind. This is rapidly changing in recent years owning to great efforts from both academia and industry. A number of potent tool compounds from both synthetic and natural products that selective target different subtypes of TRPC channels have been discovered, including some preclinical drug candidates. This review will cover recent advancements in the understanding of TRPC channel regulation, structure, and discovery of novel TRPC small molecular probes over the past few years, with the goal of facilitating drug discovery for the study of TRPCs and therapeutic development.
Collapse
Affiliation(s)
- Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaoding Cheng
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yuling Xiao
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Tian Tian
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Fuchun Xu
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Xuechuan Hong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China; Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China.
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
6
|
Schwarz Y, Oleinikov K, Schindeldecker B, Wyatt A, Weißgerber P, Flockerzi V, Boehm U, Freichel M, Bruns D. TRPC channels regulate Ca2+-signaling and short-term plasticity of fast glutamatergic synapses. PLoS Biol 2019; 17:e3000445. [PMID: 31536487 PMCID: PMC6773422 DOI: 10.1371/journal.pbio.3000445] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 10/01/2019] [Accepted: 08/29/2019] [Indexed: 11/18/2022] Open
Abstract
Transient receptor potential (TRP) proteins form Ca2+-permeable, nonselective cation channels, but their role in neuronal Ca2+ homeostasis is elusive. In the present paper, we show that TRPC channels potently regulate synaptic plasticity by changing the presynaptic Ca2+-homeostasis of hippocampal neurons. Specifically, loss of TRPC1/C4/C5 channels decreases basal-evoked secretion, reduces the pool size of readily releasable vesicles, and accelerates synaptic depression during high-frequency stimulation (HFS). In contrast, primary TRPC5 channel-expressing neurons, identified by a novel TRPC5–τ-green fluorescent protein (τGFP) knockin mouse line, show strong short-term enhancement (STE) of synaptic signaling during HFS, indicating a key role of TRPC5 in short-term plasticity. Lentiviral expression of either TRPC1 or TRPC5 turns classic synaptic depression of wild-type neurons into STE, demonstrating that TRPCs are instrumental in regulating synaptic plasticity. Presynaptic Ca2+ imaging shows that TRPC activity strongly boosts synaptic Ca2+ dynamics, showing that TRPC channels provide an additional presynaptic Ca2+ entry pathway, which efficiently regulates synaptic strength and plasticity. Transient receptor potential (TRP) proteins can form non-selective cation channels, but their role in synaptic transmission is poorly understood. This study shows that calcium-permeable TRPC channels provide an additional calcium entry pathway at presynaptic sites and are efficient regulators of synaptic strength and plasticity.
Collapse
Affiliation(s)
- Yvonne Schwarz
- Institute for Physiology, Saarland University, CIPMM, Homburg/Saar, Germany
| | | | | | - Amanda Wyatt
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, Homburg/Saar, Germany
| | - Petra Weißgerber
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, Homburg/Saar, Germany
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, Homburg/Saar, Germany
| | - Ulrich Boehm
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, Homburg/Saar, Germany
| | - Marc Freichel
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Dieter Bruns
- Institute for Physiology, Saarland University, CIPMM, Homburg/Saar, Germany
| |
Collapse
|
7
|
Emerging Roles of Diacylglycerol-Sensitive TRPC4/5 Channels. Cells 2018; 7:cells7110218. [PMID: 30463370 PMCID: PMC6262340 DOI: 10.3390/cells7110218] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 12/31/2022] Open
Abstract
Transient receptor potential classical or canonical 4 (TRPC4) and TRPC5 channels are members of the classical or canonical transient receptor potential (TRPC) channel family of non-selective cation channels. TRPC4 and TRPC5 channels are widely accepted as receptor-operated cation channels that are activated in a phospholipase C-dependent manner, following the Gq/11 protein-coupled receptor activation. However, their precise activation mechanism has remained largely elusive for a long time, as the TRPC4 and TRPC5 channels were considered as being insensitive to the second messenger diacylglycerol (DAG) in contrast to the other TRPC channels. Recent findings indicate that the C-terminal interactions with the scaffolding proteins Na+/H+ exchanger regulatory factor 1 and 2 (NHERF1 and NHERF2) dynamically regulate the DAG sensitivity of the TRPC4 and TRPC5 channels. Interestingly, the C-terminal NHERF binding suppresses, while the dissociation of NHERF enables, the DAG sensitivity of the TRPC4 and TRPC5 channels. This leads to the assumption that all of the TRPC channels are DAG sensitive. The identification of the regulatory function of the NHERF proteins in the TRPC4/5-NHERF protein complex offers a new starting point to get deeper insights into the molecular basis of TRPC channel activation. Future studies will have to unravel the physiological and pathophysiological functions of this multi-protein channel complex.
Collapse
|
8
|
Ding M, Wang H, Qu C, Xu F, Zhu Y, Lv G, Lu Y, Zhou Q, Zhou H, Zeng X, Zhang J, Yan C, Lin J, Luo HR, Deng Z, Xiao Y, Tian J, Zhu MX, Hong X. Pyrazolo[1,5-a]pyrimidine TRPC6 antagonists for the treatment of gastric cancer. Cancer Lett 2018; 432:47-55. [PMID: 29859875 PMCID: PMC6345172 DOI: 10.1016/j.canlet.2018.05.041] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/09/2018] [Accepted: 05/24/2018] [Indexed: 01/16/2023]
Abstract
Transient receptor potential canonical 6 (TRPC6) proteins form receptor-operated Ca2+-permeable channels, which have been thought to bring benefit to the treatment of diseases, including cancer. However, selective antagonists for TRPC channels are rare and none of them has been tested against gastric cancer. Compound 14a and analogs were synthesized by chemical elaboration of previously reported TRPC3/6/7 agonist 4o. 14a had very weak agonist activity at TRPC6 expressed in HEK293 cells but exhibited strong inhibition on both 4o-mediated and receptor-operated activation of TRPC6 with an IC50 of about 1 μM. When applied to the culture media, 14a suppressed proliferation of AGS and MKN45 cells with IC50 values of 17.1 ± 0.3 and 18.5 ± 1.0 μM, respectively, and inhibited tube formation and migration of cultured human endothelial cells. This anti-tumor effect on gastric cancer was further verified in xenograft models using nude mice. This study has found a new tool compound which shows excellent therapeutic potential against human gastric cancer most likely through targeting TRPC6 channels.
Collapse
Affiliation(s)
- Mingmin Ding
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Medical College, Tibet University, Lasa, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Chunrong Qu
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Medical College, Tibet University, Lasa, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Fuchun Xu
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Medical College, Tibet University, Lasa, China
| | - Yingmin Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Guangyao Lv
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Yungang Lu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qingjun Zhou
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Hui Zhou
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Xiaodong Zeng
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Jingwen Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Chunhong Yan
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Jiacheng Lin
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Huai-Rong Luo
- Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Zixing Deng
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Yuling Xiao
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xuechuan Hong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Medical College, Tibet University, Lasa, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China.
| |
Collapse
|
9
|
Reboreda A, Theissen FM, Valero-Aracama MJ, Arboit A, Corbu MA, Yoshida M. Do TRPC channels support working memory? Comparing modulations of TRPC channels and working memory through G-protein coupled receptors and neuromodulators. Behav Brain Res 2018; 354:64-83. [PMID: 29501506 DOI: 10.1016/j.bbr.2018.02.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
Working memory is a crucial ability we use in daily life. However, the cellular mechanisms supporting working memory still remain largely unclear. A key component of working memory is persistent neural firing which is believed to serve short-term (hundreds of milliseconds up to tens of seconds) maintenance of necessary information. In this review, we will focus on the role of transient receptor potential canonical (TRPC) channels as a mechanism underlying persistent firing. Many years of in vitro work have been suggesting a crucial role of TRPC channels in working memory and temporal association tasks. If TRPC channels are indeed a central mechanism for working memory, manipulations which impair or facilitate working memory should have a similar effect on TRPC channel modulation. However, modulations of working memory and TRPC channels were never systematically compared, and it remains unanswered whether TRPC channels indeed contribute to working memory in vivo or not. In this article, we review the effects of G-protein coupled receptors (GPCR) and neuromodulators, including acetylcholine, noradrenalin, serotonin and dopamine, on working memory and TRPC channels. Based on comparisons, we argue that GPCR and downstream signaling pathways that activate TRPC, generally support working memory, while those that suppress TRPC channels impair it. However, depending on the channel types, areas, and systems tested, this is not the case in all studies. Further work to clarify involvement of specific TRPC channels in working memory tasks and how they are affected by neuromodulators is still necessary in the future.
Collapse
Affiliation(s)
- Antonio Reboreda
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany.
| | - Frederik M Theissen
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Maria J Valero-Aracama
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 17, 91054 Erlangen, Germany
| | - Alberto Arboit
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Mihaela A Corbu
- Ruhr University Bochum (RUB), Universitätsstraße 150, 44801, Bochum, Germany
| | - Motoharu Yoshida
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany; Center for Behavioral Brain Sciences, 39106, Magdeburg, Germany.
| |
Collapse
|
10
|
Maddox JW, Khorsandi N, Gleason E. TRPC5 is required for the NO-dependent increase in dendritic Ca 2+ and GABA release from chick retinal amacrine cells. J Neurophysiol 2017; 119:262-273. [PMID: 28978766 DOI: 10.1152/jn.00500.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
GABAergic signaling from amacrine cells (ACs) is a fundamental aspect of visual signal processing in the inner retina. We have previously shown that nitric oxide (NO) can elicit release of GABA independently from activation of voltage-gated Ca2+ channels in cultured retinal ACs. This voltage-independent quantal GABA release relies on a Ca2+ influx mechanism with pharmacological characteristics consistent with the involvement of the transient receptor potential canonical (TRPC) channels TRPC4 and/or TRPC5. To determine the identity of these channels, we evaluated the ability of NO to elevate dendritic Ca2+ and to stimulate GABA release from cultured ACs under conditions known to alter the function of TRPC4 and 5. We found that these effects of NO are phospholipase C dependent, have a biphasic dependence on La3+, and are unaffected by moderate concentrations of the TRPC4-selective antagonist ML204. Together, these results suggest that NO promotes GABA release by activating TRPC5 channels in AC dendrites. To confirm a role for TRPC5, we knocked down the expression of TRPC5 using CRISPR/Cas9-mediated gene knockdown and found that both the NO-dependent Ca2+ elevations and increase in GABA release are dependent on the expression of TRPC5. These results demonstrate a novel NO-dependent mechanism for regulating neurotransmitter output from retinal ACs. NEW & NOTEWORTHY Elucidating the mechanisms regulating GABAergic synaptic transmission in the inner retina is key to understanding the flexibility of retinal ganglion cell output. Here, we demonstrate that nitric oxide (NO) can activate a transient receptor potential canonical 5 (TRPC5)-mediated Ca2+ influx, which is sufficient to drive vesicular GABA release from retinal amacrine cells. This NO-dependent mechanism can bypass the need for depolarization and may have an important role in processing the visual signal by enhancing retinal amacrine cell GABAergic inhibitory output.
Collapse
Affiliation(s)
- J Wesley Maddox
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Nikka Khorsandi
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Evanna Gleason
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| |
Collapse
|
11
|
Integrative Approach with Electrophysiological and Theoretical Methods Reveals a New Role of S4 Positively Charged Residues in PKD2L1 Channel Voltage-Sensing. Sci Rep 2017; 7:9760. [PMID: 28852171 PMCID: PMC5575081 DOI: 10.1038/s41598-017-10357-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/02/2017] [Indexed: 01/15/2023] Open
Abstract
Numerical model-based simulations provide important insights into ion channel gating when experimental limitations exist. Here, a novel strategy combining numerical simulations with patch clamp experiments was used to investigate the net positive charges in the putative transmembrane segment 4 (S4) of the atypical, positively-shifted voltage-dependence of polycystic kidney disease 2-like 1 (PKD2L1) channel. Charge-neutralising mutations (K452Q, K455Q and K461Q) in S4 reduced gating charges, positively shifted the Boltzmann-type activation curve [i.e., open probability (Popen)-V curve] and altered the time-courses of activation/deactivation of PKD2L1, indicating that this region constitutes part of a voltage sensor. Numerical reconstruction of wild-type (WT) and mutant PKD2L1-mediated currents necessitated, besides their voltage-dependent gating parameters, a scaling factor that describes the voltage-dependence of maximal conductance, Gmax. Subsequent single-channel conductance (γ) measurements revealed that voltage-dependence of Gmax in WT can be explained by the inward-rectifying property of γ, which is greatly changed in PKD2L1 mutants. Homology modelling based on PKD2 and NaVAb structures suggest that such voltage dependence of Popen and γ in PKD2L1 could both reflect the charged state of the S4 domain. The present conjunctive experimental and theoretical approaches provide a framework to explore the undetermined mechanism(s) regulating TRP channels that possess non-classical voltage-dependent properties.
Collapse
|
12
|
He Z. TRPC Channel Downstream Signaling Cascades. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 976:25-33. [PMID: 28508310 DOI: 10.1007/978-94-024-1088-4_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The family of TRP channel is comprised of a large group of cation-permeable channels, displaying as signaling integrators for sensing extracellular stimulus and initiating intracellular signaling cascades. This chapter offers a brief review of the signaling molecules related to TRPC channels, the first identified mammalian TRP family. Besides the signaling molecules involved in TRPC activation, I will focus on their upstream and downstream signaling cascades and the molecules involved in their intracellular trafficking.
Collapse
Affiliation(s)
- Zhuohao He
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
13
|
Kirschmer N, Bandleon S, von Ehrlich-Treuenstätt V, Hartmann S, Schaaf A, Lamprecht AK, Miranda-Laferte E, Langsenlehner T, Ritter O, Eder P. TRPC4α and TRPC4β Similarly Affect Neonatal Cardiomyocyte Survival during Chronic GPCR Stimulation. PLoS One 2016; 11:e0168446. [PMID: 27992507 PMCID: PMC5167390 DOI: 10.1371/journal.pone.0168446] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 12/01/2016] [Indexed: 11/19/2022] Open
Abstract
The Transient Receptor Potential Channel Subunit 4 (TRPC4) has been considered as a crucial Ca2+ component in cardiomyocytes promoting structural and functional remodeling in the course of pathological cardiac hypertrophy. TRPC4 assembles as homo or hetero-tetramer in the plasma membrane, allowing a non-selective Na+ and Ca2+ influx. Gαq protein-coupled receptor (GPCR) stimulation is known to increase TRPC4 channel activity and a TRPC4-mediated Ca2+ influx which has been regarded as ideal Ca2+ source for calcineurin and subsequent nuclear factor of activated T-cells (NFAT) activation. Functional properties of TRPC4 are also based on the expression of the TRPC4 splice variants TRPC4α and TRPC4β. Aim of the present study was to analyze cytosolic Ca2+ signals, signaling, hypertrophy and vitality of cardiomyocytes in dependence on the expression level of either TRPC4α or TRPC4β. The analysis of Ca2+ transients in neonatal rat cardiomyocytes (NRCs) showed that TRPC4α and TRPC4β affected Ca2+ cycling in beating cardiomyocytes with both splice variants inducing an elevation of the Ca2+ transient amplitude at baseline and TRPC4β increasing the Ca2+ peak during angiotensin II (Ang II) stimulation. NRCs infected with TRPC4β (Ad-C4β) also responded with a sustained Ca2+ influx when treated with Ang II under non-pacing conditions. Consistent with the Ca2+ data, NRCs infected with TRPC4α (Ad-C4α) showed an elevated calcineurin/NFAT activity and a baseline hypertrophic phenotype but did not further develop hypertrophy during chronic Ang II/phenylephrine stimulation. Down-regulation of endogenous TRPC4α reversed these effects, resulting in less hypertrophy of NRCs at baseline but a markedly increased hypertrophic enlargement after chronic agonist stimulation. Ad-C4β NRCs did not exhibit baseline calcineurin/NFAT activity or hypertrophy but responded with an increased calcineurin/NFAT activity after GPCR stimulation. However, this effect was not translated into an increased propensity towards hypertrophy but rather less hypertrophy during GPCR stimulation. Further analyses revealed that, although hypertrophy was preserved in Ad-C4α NRCs and even attenuated in Ad-C4β NRCs, cardiomyocytes had an increased apoptosis rate and thus were less viable after chronic GPCR stimulation. These findings suggest that TRPC4α and TRPC4β differentially affect Ca2+ signals, calcineurin/NFAT signaling and hypertrophy but similarly impair cardiomyocyte viability during GPCR stimulation.
Collapse
Affiliation(s)
- Nadine Kirschmer
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Sandra Bandleon
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Viktor von Ehrlich-Treuenstätt
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Sonja Hartmann
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, United States of America
| | - Alice Schaaf
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Anna-Karina Lamprecht
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | | | - Tanja Langsenlehner
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, Graz, Austria
| | - Oliver Ritter
- Department of Cardiology and Pulmology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany
| | - Petra Eder
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
14
|
Dynamic NHERF interaction with TRPC4/5 proteins is required for channel gating by diacylglycerol. Proc Natl Acad Sci U S A 2016; 114:E37-E46. [PMID: 27994151 DOI: 10.1073/pnas.1612263114] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The activation mechanism of the classical transient receptor potential channels TRPC4 and -5 via the Gq/11 protein-phospholipase C (PLC) signaling pathway has remained elusive so far. In contrast to all other TRPC channels, the PLC product diacylglycerol (DAG) is not sufficient for channel activation, whereas TRPC4/5 channel activity is potentiated by phosphatidylinositol 4,5-bisphosphate (PIP2) depletion. As a characteristic structural feature, TRPC4/5 channels contain a C-terminal PDZ-binding motif allowing for binding of the scaffolding proteins Na+/H+ exchanger regulatory factor (NHERF) 1 and 2. PKC inhibition or the exchange of threonine for alanine in the C-terminal PDZ-binding motif conferred DAG sensitivity to the channel. Altogether, we present a DAG-mediated activation mechanism for TRPC4/5 channels tightly regulated by NHERF1/2 interaction. PIP2 depletion evokes a C-terminal conformational change of TRPC5 proteins leading to dynamic dissociation of NHERF1/2 from the C terminus of TRPC5 as a prerequisite for DAG sensitivity. We show that NHERF proteins are direct regulators of ion channel activity and that DAG sensitivity is a distinctive hallmark of TRPC channels.
Collapse
|
15
|
Chen X, Li W, Riley AM, Soliman M, Chakraborty S, Stamatkin CW, Obukhov AG. Molecular Determinants of the Sensitivity to Gq/11-Phospholipase C-dependent Gating, Gd3+ Potentiation, and Ca2+ Permeability in the Transient Receptor Potential Canonical Type 5 (TRPC5) Channel. J Biol Chem 2016; 292:898-911. [PMID: 27920205 DOI: 10.1074/jbc.m116.755470] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/24/2016] [Indexed: 11/06/2022] Open
Abstract
Transient receptor potential canonical type 5 (TRPC5) is a Ca2+-permeable cation channel that is highly expressed in the brain and is implicated in motor coordination, innate fear behavior, and seizure genesis. The channel is activated by a signal downstream of the G-protein-coupled receptor (GPCR)-Gq/11-phospholipase C (PLC) pathway. In this study we aimed to identify the molecular mechanisms involved in regulating TRPC5 activity. We report that Arg-593, a residue located in the E4 loop near the TRPC5 extracellular Gd3+ binding site, is critical for conferring the sensitivity to GPCR-Gq/11-PLC-dependent gating on TRPC5. Indeed, guanosine 5'-O-(thiotriphosphate) and GPCR agonists only weakly activate the TRPC5R593A mutant, whereas the addition of Gd3+ rescues the mutant's sensitivity to GPCR-Gq/11-PLC-dependent gating. Computer modeling suggests that Arg-593 may cross-bridge the E3 and E4 loops, forming the "molecular fulcrum." While validating the model using site-directed mutagenesis, we found that the Tyr-542 residue is critical for establishing a functional Gd3+ binding site, the Tyr-541 residue participates in fine-tuning Gd3+-sensitivity, and that the Asn-584 residue determines Ca2+ permeability of the TRPC5 channel. This is the first report providing molecular insights into the molecular mechanisms regulating the sensitivity to GPCR-Gq/11-PLC-dependent gating of a receptor-operated channel.
Collapse
Affiliation(s)
- Xingjuan Chen
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Wennan Li
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Ashley M Riley
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Mario Soliman
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Saikat Chakraborty
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Christopher W Stamatkin
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Alexander G Obukhov
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
16
|
Toman O, Kabickova T, Vit O, Fiser R, Polakova KM, Zach J, Linhartova J, Vyoral D, Petrak J. Proteomic analysis of imatinib-resistant CML-T1 cells reveals calcium homeostasis as a potential therapeutic target. Oncol Rep 2016; 36:1258-68. [PMID: 27430982 PMCID: PMC4968618 DOI: 10.3892/or.2016.4945] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 03/26/2016] [Indexed: 11/16/2022] Open
Abstract
Chronic myeloid leukemia (CML) therapy has markedly improved patient prognosis after introduction of imatinib mesylate for clinical use. However, a subset of patients develops resistance to imatinib and other tyrosine kinase inhibitors (TKIs), mainly due to point mutations in the region encoding the kinase domain of the fused BCR-ABL oncogene. To identify potential therapeutic targets in imatinib-resistant CML cells, we derived imatinib-resistant CML-T1 human cell line clone (CML-T1/IR) by prolonged exposure to imatinib in growth media. Mutational analysis revealed that the Y235H mutation in BCR-ABL is probably the main cause of CML-T1/IR resistance to imatinib. To identify alternative therapeutic targets for selective elimination of imatinib-resistant cells, we compared the proteome profiles of CML-T1 and CML-T1/IR cells using 2-DE-MS. We identified eight differentially expressed proteins, with strongly upregulated Na+/H+ exchanger regulatory factor 1 (NHERF1) in the resistant cells, suggesting that this protein may influence cytosolic pH, Ca2+ concentration or signaling pathways such as Wnt in CML-T1/IR cells. We tested several compounds including drugs in clinical use that interfere with the aforementioned processes and tested their relative toxicity to CML-T1 and CML-T1/IR cells. Calcium channel blockers, calcium signaling antagonists and modulators of calcium homeostasis, namely thapsigargin, ionomycin, verapamil, carboxyamidotriazole and immunosuppressive drugs cyclosporine A and tacrolimus (FK-506) were selectively toxic to CML-T1/IR cells. The putative cellular targets of these compounds in CML-T1/IR cells are postulated in this study. We propose that Ca2+ homeostasis can be a potential therapeutic target in CML cells resistant to TKIs. We demonstrate that a proteomic approach may be used to characterize a TKI-resistant population of CML cells enabling future individualized treatment options for patients.
Collapse
Affiliation(s)
- O Toman
- Institute of Hematology and Blood Transfusion, CZ-12820 Prague 2, Czech Republic
| | - T Kabickova
- Institute of Hematology and Blood Transfusion, CZ-12820 Prague 2, Czech Republic
| | - O Vit
- BIOCEV, First Faculty of Medicine, Charles University in Prague, CZ-25250 Vestec, Czech Republic
| | - R Fiser
- Department of Genetics and Microbiology, Faculty of Natural Sciences, Charles University in Prague, CZ-12843 Prague, Czech Republic
| | - K Machova Polakova
- Institute of Hematology and Blood Transfusion, CZ-12820 Prague 2, Czech Republic
| | - J Zach
- Institute of Hematology and Blood Transfusion, CZ-12820 Prague 2, Czech Republic
| | - J Linhartova
- Institute of Hematology and Blood Transfusion, CZ-12820 Prague 2, Czech Republic
| | - D Vyoral
- Institute of Hematology and Blood Transfusion, CZ-12820 Prague 2, Czech Republic
| | - J Petrak
- Institute of Hematology and Blood Transfusion, CZ-12820 Prague 2, Czech Republic
| |
Collapse
|
17
|
Critical roles of Gi/o proteins and phospholipase C-δ1 in the activation of receptor-operated TRPC4 channels. Proc Natl Acad Sci U S A 2016; 113:1092-7. [PMID: 26755577 DOI: 10.1073/pnas.1522294113] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transient Receptor Potential Canonical (TRPC) proteins form nonselective cation channels commonly known to be activated downstream from receptors that signal through phospholipase C (PLC). Although TRPC3/C6/C7 can be directly activated by diacylglycerols produced by PLC breakdown of phosphatidylinositol 4,5-bisphosphate (PIP2), the mechanism by which the PLC pathway activates TRPC4/C5 remains unclear. We show here that TRPC4 activation requires coincident stimulation of Gi/o subgroup of G proteins and PLCδ, with a preference for PLCδ1 over PLCδ3, but not necessarily the PLCβ pathway commonly thought to be involved in receptor-operated TRPC activation. In HEK293 cells coexpressing TRPC4 and Gi/o-coupled µ opioid receptor, µ agonist elicited currents biphasically, with an initial slow phase preceding a rapidly developing phase. The currents were dependent on intracellular Ca(2+) and PIP2. Reducing PIP2 through phosphatases abolished the biphasic kinetics and increased the probability of channel activation by weak Gi/o stimulation. In both HEK293 cells heterologously expressing TRPC4 and renal carcinoma-derived A-498 cells endogenously expressing TRPC4, channel activation was inhibited by knocking down PLCδ1 levels and almost completely eliminated by a dominant-negative PLCδ1 mutant and a constitutively active RhoA mutant. Conversely, the slow phase of Gi/o-mediated TRPC4 activation was diminished by inhibiting RhoA or enhancing PLCδ function. Our data reveal an integrative mechanism of TRPC4 on detection of coincident Gi/o, Ca(2+), and PLC signaling, which is further modulated by the small GTPase RhoA. This mechanism is not shared with the closely related TRPC5, implicating unique roles of TRPC4 in signal integration in brain and other systems.
Collapse
|
18
|
Constantin B. Role of Scaffolding Proteins in the Regulation of TRPC-Dependent Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:379-403. [PMID: 27161237 DOI: 10.1007/978-3-319-26974-0_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Plasma membrane ion channels, and in particular TRPC channels need a specific membrane environment and association with scaffolding, signaling, and cytoskeleton proteins in order to play their important functional role. The molecular composition of TRPC channels is an important factor in determining channel activation mechanisms. TRPC proteins are incorporated in macromolecular complexes including several key Ca(2 +) signaling proteins as well as proteins involved in vesicle trafficking, cytoskeletal interactions, and scaffolding. Evidence has been provided for association of TRPC with calmodulin (CaM), IP3R, PMCA, Gq/11, RhoA, and a variety of scaffolding proteins. The interaction between TRPC channels with adaptor proteins, determines their mode of regulation as well as their cellular localization and function. Adaptor proteins do not display any enzymatic activity but act as scaffold for the building of signaling complexes. The scaffolding proteins are involved in the assembling of these Ca(2+) signaling complexes, the correct sub-cellular localization of protein partners, and the regulation of the TRPC channelosome. In particular, these proteins, via their multiple protein-protein interaction motifs, can interact with various ion channels involved in the transmembrane potential, and membrane excitability. Scaffolding proteins are key components for the functional organization of TRPC channelosomes that serves as a platform regulating slow Ca(2+) entry, spatially and temporally controlled [Ca(2+)]i signals and Ca(2+) -dependent cellular functions.
Collapse
Affiliation(s)
- Bruno Constantin
- Laboratory STIM, ERL-7368 CNRS-Université de Poitiers, 1, rue Georges Bonnet, Bat. B36, Pôle Biologie-Santé, 86000, Poitiers, France.
| |
Collapse
|
19
|
Shen B, Wong CO, Lau OC, Woo T, Bai S, Huang Y, Yao X. Plasma membrane mechanical stress activates TRPC5 channels. PLoS One 2015; 10:e0122227. [PMID: 25849346 PMCID: PMC4388645 DOI: 10.1371/journal.pone.0122227] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 02/19/2015] [Indexed: 01/14/2023] Open
Abstract
Mechanical forces exerted on cells impose stress on the plasma membrane. Cells sense this stress and elicit a mechanoelectric transduction cascade that initiates compensatory mechanisms. Mechanosensitive ion channels in the plasma membrane are responsible for transducing the mechanical signals to electrical signals. However, the mechanisms underlying channel activation in response to mechanical stress remain incompletely understood. Transient Receptor Potential (TRP) channels serve essential functions in several sensory modalities. These channels can also participate in mechanotransduction by either being autonomously sensitive to mechanical perturbation or by coupling to other mechanosensory components of the cell. Here, we investigated the response of a TRP family member, TRPC5, to mechanical stress. Hypoosmolarity triggers Ca2+ influx and cationic conductance through TRPC5. Importantly, for the first time we were able to record the stretch-activated TRPC5 current at single-channel level. The activation threshold for TRPC5 was found to be 240 mOsm for hypoosmotic stress and between −20 and −40 mmHg for pressure applied to membrane patch. In addition, we found that disruption of actin filaments suppresses TRPC5 response to hypoosmotic stress and patch pipette pressure, but does not prevent the activation of TRPC5 by stretch-independent mechanisms, indicating that actin cytoskeleton is an essential transduction component that confers mechanosensitivity to TRPC5. In summary, our findings establish that TRPC5 can be activated at the single-channel level when mechanical stress on the cell reaches a certain threshold.
Collapse
Affiliation(s)
- Bing Shen
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong, China
- Department of Physiology, Anhui Medical University, Hefei, China
| | - Ching-On Wong
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong, China
- * E-mail: (XY); (CW)
| | - On-Chai Lau
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Theodosia Woo
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Suwen Bai
- Department of Physiology, Anhui Medical University, Hefei, China
| | - Yu Huang
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong, China
- * E-mail: (XY); (CW)
| |
Collapse
|
20
|
Nilius B, Szallasi A. Transient Receptor Potential Channels as Drug Targets: From the Science of Basic Research to the Art of Medicine. Pharmacol Rev 2014; 66:676-814. [DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
21
|
Abstract
Human canonical transient receptor potential channel 5 (TRPC5) has been cloned from the Xq23 region on chromosome X as a suspect in nonsyndromic mental retardation. TRPC5 is a Ca(2+)-permeable cation channel predominantly expressed in the CNS, including the hippocampus, cerebellum, amygdala, sensory neurons, and retina. It also shows more restricted expression in the periphery, notably in the kidney and cardiovascular system. Homotetrameric TRPC5 channels are primarily activated by receptors coupled to Gq and phospholipase C and/or Gi proteins, but TRPC5 channels may also gate in a store-dependent manner, which requires other partner proteins such TRPC1, STIM1, and Orai1. There is an impressive array of other activators of TRPC5 channels, such as nitric oxide, lysophospholipids, sphingosine-1-phosphate, reduced thioredoxin, protons, lanthanides, and calcium, and many can cause its direct activation. Moreover, TRPC5 shows constitutive activity, and it is responsive to membrane stretch and cold. Thus, TRPC5 channels have significant potential for synergistic activation and may serve as an important focal point in Ca(2+) signalling and electrogenesis. Moreover, TRPC5 functions in partnership with about 60 proteins, including TRPC1, TRPC4, calmodulin, IP3 receptors, NHERF, NCS-1, junctate, stathmin 2, Ca(2+)-binding protein 1, caveolin, and SESTD1, while its desensitisation is mediated by both protein kinases A and C. TRPC5 has a distinct voltage dependence shared only with its closest relative, TRPC4. Its unique N-shaped activation curve underlined by intracellular Mg(2+) block seems to be perfectly "shaped" to trigger action potential discharge, but not to grossly interfere with the action potential shape. The range of biological functions of TRPC5 channels is also impressive, from neurotransmission to control of axon guidance and vascular smooth muscle cell migration and contractility. Recent studies of Trpc5 gene knockouts begin to uncover its roles in fear, anxiety, seizures, and cold sensing.
Collapse
Affiliation(s)
- Alexander V Zholos
- Department of Biophysics, Educational and Scientific Centre "Institute of Biology", Taras Shevchenko Kiev National University, Kiev, 03022, Ukraine,
| |
Collapse
|
22
|
Ong HL, de Souza LB, Cheng KT, Ambudkar IS. Physiological functions and regulation of TRPC channels. Handb Exp Pharmacol 2014; 223:1005-34. [PMID: 24961978 DOI: 10.1007/978-3-319-05161-1_12] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The TRP-canonical (TRPC) subfamily, which consists of seven members (TRPC1-TRPC7), are Ca(2+)-permeable cation channels that are activated in response to receptor-mediated PIP2 hydrolysis via store-dependent and store-independent mechanisms. These channels are involved in a variety of physiological functions in different cell types and tissues. Of these, TRPC6 has been linked to a channelopathy resulting in human disease. Two key players of the store-dependent regulatory pathway, STIM1 and Orai1, interact with some TRPC channels to gate and regulate channel activity. The Ca(2+) influx mediated by TRPC channels generates distinct intracellular Ca(2+) signals that regulate downstream signaling events and consequent cell functions. This requires localization of TRPC channels in specific plasma membrane microdomains and precise regulation of channel function which is coordinated by various scaffolding, trafficking, and regulatory proteins.
Collapse
Affiliation(s)
- Hwei Ling Ong
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | | |
Collapse
|
23
|
Abstract
TRP channels constitute a large superfamily of cation channel forming proteins, all related to the gene product of the transient receptor potential (trp) locus in Drosophila. In mammals, 28 different TRP channel genes have been identified, which exhibit a large variety of functional properties and play diverse cellular and physiological roles. In this article, we provide a brief and systematic summary of expression, function, and (patho)physiological role of the mammalian TRP channels.
Collapse
Affiliation(s)
- Maarten Gees
- Laboratory Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Campus Gasthuisberg, Leuven, Belgium
| | | | | | | |
Collapse
|
24
|
Kim H, Kim J, Jeon JP, Myeong J, Wie J, Hong C, Kim HJ, Jeon JH, So I. The roles of G proteins in the activation of TRPC4 and TRPC5 transient receptor potential channels. Channels (Austin) 2012; 6:333-43. [PMID: 22878724 PMCID: PMC3508772 DOI: 10.4161/chan.21198] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
TRPC4 and TRPC5 channels are important regulators of electrical excitability in both gastrointestinal myocytes and neurons. Much is known regarding the assembly and function of these channels including TRPC1 as a homotetramer or a heteromultimer and the roles that their interacting proteins play in controlling these events. Further, they are one of the best-studied targets of G protein-coupled receptors and growth factors in general and Gαq protein coupled receptor or epidermal growth factor in particular. However, our understanding of the roles of Gαi/o proteins on TRPC4/5 channels is still rudimentary. We discuss potential roles for Gαi/o proteins in channel activation in addition to their known role in cellular signaling.
Collapse
Affiliation(s)
- Hana Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Moccia F, Berra-Romani R, Tanzi F. Update on vascular endothelial Ca 2+ signalling: A tale of ion channels, pumps and transporters. World J Biol Chem 2012; 3:127-58. [PMID: 22905291 PMCID: PMC3421132 DOI: 10.4331/wjbc.v3.i7.127] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/04/2012] [Accepted: 07/11/2012] [Indexed: 02/05/2023] Open
Abstract
A monolayer of endothelial cells (ECs) lines the lumen of blood vessels and forms a multifunctional transducing organ that mediates a plethora of cardiovascular processes. The activation of ECs from as state of quiescence is, therefore, regarded among the early events leading to the onset and progression of potentially lethal diseases, such as hypertension, myocardial infarction, brain stroke, and tumor. Intracellular Ca2+ signals have long been know to play a central role in the complex network of signaling pathways regulating the endothelial functions. Notably, recent work has outlined how any change in the pattern of expression of endothelial channels, transporters and pumps involved in the modulation of intracellular Ca2+ levels may dramatically affect whole body homeostasis. Vascular ECs may react to both mechanical and chemical stimuli by generating a variety of intracellular Ca2+ signals, ranging from brief, localized Ca2+ pulses to prolonged Ca2+ oscillations engulfing the whole cytoplasm. The well-defined spatiotemporal profile of the subcellular Ca2+ signals elicited in ECs by specific extracellular inputs depends on the interaction between Ca2+ releasing channels, which are located both on the plasma membrane and in a number of intracellular organelles, and Ca2+ removing systems. The present article aims to summarize both the past and recent literature in the field to provide a clear-cut picture of our current knowledge on the molecular nature and the role played by the components of the Ca2+ machinery in vascular ECs under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Francesco Moccia
- Francesco Moccia, Franco Tanzi, Department of Biology and Biotechnologies "Lazzaro Spallanzani", Laboratory of Physiology, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | | | | |
Collapse
|
26
|
Kumar S, Chakraborty S, Barbosa C, Brustovetsky T, Brustovetsky N, Obukhov AG. Mechanisms controlling neurite outgrowth in a pheochromocytoma cell line: the role of TRPC channels. J Cell Physiol 2012; 227:1408-19. [PMID: 21618530 DOI: 10.1002/jcp.22855] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transient Receptor Potential Canonical (TRPC) channels are implicated in modulating neurite outgrowth. The expression pattern of TRPCs changes significantly during brain development, suggesting that fine-tuning TRPC expression may be important for orchestrating neuritogenesis. To study how alterations in the TRPC expression pattern affect neurite outgrowth, we used nerve growth factor (NGF)-differentiated rat pheochromocytoma 12 (PC12) cells, a model system for neuritogenesis. In PC12 cells, NGF markedly up-regulated TRPC1 and TRPC6 expression, but down-regulated TRPC5 expression while promoting neurite outgrowth. Overexpression of TRPC1 augmented, whereas TRPC5 overexpression decelerated NGF-induced neurite outgrowth. Conversely, shRNA-mediated knockdown of TRPC1 decreased, whereas shRNA-mediated knockdown of TRPC5 increased NGF-induced neurite extension. Endogenous TRPC1 attenuated the anti-neuritogenic effect of overexpressed TRPC5 in part by forming the heteromeric TRPC1-TRPC5 channels. Previous reports suggested that TRPC6 may facilitate neurite outgrowth. However, we found that TRPC6 overexpression slowed down neuritogenesis, whereas dominant negative TRPC6 (DN-TRPC6) facilitated neurite outgrowth in NGF-differentiated PC12 cells. Consistent with these findings, hyperforin, a neurite outgrowth promoting factor, decreased TRPC6 expression in NGF-differentiated PC12 cells. Using pharmacological and molecular biological approaches, we determined that NGF up-regulated TRPC1 and TRPC6 expression via a p75(NTR)-IKK(2)-dependent pathway that did not involve TrkA receptor signaling in PC12 cells. Similarly, NGF up-regulated TRPC1 and TRPC6 via an IKK(2) dependent pathway in primary cultured hippocampal neurons. Thus, our data suggest that a balance of TRPC1, TRPC5, and TRPC6 expression determines neurite extension rate in neural cells, with TRPC6 emerging as an NGF-dependent "molecular damper" maintaining a submaximal velocity of neurite extension.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Cellular and Integrative Physiology, IUPUI-Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | |
Collapse
|
27
|
Jiang LH, Gamper N, Beech DJ. Properties and therapeutic potential of transient receptor potential channels with putative roles in adversity: focus on TRPC5, TRPM2 and TRPA1. Curr Drug Targets 2011; 12:724-36. [PMID: 21291387 PMCID: PMC3267159 DOI: 10.2174/138945011795378568] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 06/28/2010] [Indexed: 01/13/2023]
Abstract
Mammals contain 28 genes encoding Transient Receptor Potential (TRP) proteins. The proteins assemble into cationic channels, often with calcium permeability. Important roles in physiology and disease have emerged and so there is interest in whether the channels might be suitable therapeutic drug targets. Here we review selected members of three subfamilies of mammalian TRP channel (TRPC5, TRPM2 and TRPA1) that show relevance to sensing of adversity by cells and biological systems. Summarized are the cellular and tissue distributions, general properties, endogenous modulators, protein partners, cellular and tissue functions, therapeutic potential, and pharmacology. TRPC5 is stimulated by receptor agonists and other factors that include lipids and metal ions; it heteromultimerises with other TRPC proteins and is involved in cell movement and anxiety control. TRPM2 is activated by hydrogen peroxide; it is implicated in stress-related inflammatory, vascular and neurodegenerative conditions. TRPA1 is stimulated by a wide range of irritants including mustard oil and nicotine but also, controversially, noxious cold and mechanical pressure; it is implicated in pain and inflammatory responses, including in the airways. The channels have in common that they show polymodal stimulation, have activities that are enhanced by redox factors, are permeable to calcium, and are facilitated by elevations of intracellular calcium. Developing inhibitors of the channels could lead to new agents for a variety of conditions: for example, suppressing unwanted tissue remodeling, inflammation, pain and anxiety, and addressing problems relating to asthma and stroke.
Collapse
Affiliation(s)
- L H Jiang
- Institute of Membrane & Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | | | | |
Collapse
|
28
|
Takahashi N, Mori Y. TRP Channels as Sensors and Signal Integrators of Redox Status Changes. Front Pharmacol 2011; 2:58. [PMID: 22016736 PMCID: PMC3192318 DOI: 10.3389/fphar.2011.00058] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 09/20/2011] [Indexed: 12/21/2022] Open
Abstract
Proteins are capable of sensing the redox status of cells. Cysteine residues, which react with oxidants, reductants, and electrophiles, have been increasingly recognized as the mediators of this redox sensitivity. Cation channels encoded by the transient receptor potential (trp) gene superfamily are characterized by a wide variety of activation triggers that act from outside and inside the cell. Recent studies have revealed that a class of TRP channels is sensitive to changes in redox status and is notably susceptible to modifications of cysteine residues, such as oxidation, electrophilic reaction, and S-nitrosylation of sulfhydryls. In this review, we focus on TRP channels, which directly sense redox status, and discuss the biological significance of cysteine modifications and the consequences of this chemical reaction for physiological responses.
Collapse
Affiliation(s)
- Nobuaki Takahashi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University Kyoto, Japan
| | | |
Collapse
|
29
|
Chakraborty S, Berwick ZC, Bartlett PJ, Kumar S, Thomas AP, Sturek M, Tune JD, Obukhov AG. Bromoenol lactone inhibits voltage-gated Ca2+ and transient receptor potential canonical channels. J Pharmacol Exp Ther 2011; 339:329-40. [PMID: 21795434 DOI: 10.1124/jpet.111.183673] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Circulating hormones stimulate the phospholipase Cβ (PLC)/Ca(2+) influx pathway to regulate numerous cell functions, including vascular tone. It was proposed previously that Ca(2+)-independent phospholipase A(2) (iPLA(2))-dependent store-operated Ca(2+) influx channels mediate hormone-induced contractions in isolated arteries, because bromoenol lactone (BEL), a potent irreversible inhibitor of iPLA(2), inhibited such contractions. However, the effects of BEL on other channels implicated in mediating hormone-induced vessel contractions, specifically voltage-gated Ca(2+) (Ca(V)1.2) and transient receptor potential canonical (TRPC) channels, have not been defined clearly. Using isometric tension measurements, we found that thapsigargin-induced contractions were ∼34% of those evoked by phenylephrine or KCl. BEL completely inhibited not only thapsigargin- but also phenylephrine- and KCl-induced ring contractions, suggesting that Ca(V)1.2 and receptor-operated TRPC channels also may be sensitive to BEL. Therefore, we investigated the effects of BEL on heterologously expressed Ca(V)1.2 and TRPC channels in human embryonic kidney cells, a model system that allows probing of individual protein function without interference from other signaling elements of native cells. We found that low micromolar concentrations of BEL inhibited Ca(V)1.2, TRPC5, TRPC6, and heteromeric TRPC1-TRPC5 channels in an iPLA(2)-independent manner. BEL also attenuated PLC activity, suggesting that the compound may inhibit TRPC channel activity in part by interfering with an initial PLC-dependent step required for TRPC channel activation. Conversely, BEL did not affect endogenous voltage-gated K(+) channels in human embryonic kidney cells. Our findings support the hypothesis that iPLA(2)-dependent store-operated Ca(2+) influx channels and iPLA(2)-independent hormone-operated TRPC channels can serve as smooth muscle depolarization triggers to activate Ca(V)1.2 channels and to regulate vascular tone.
Collapse
Affiliation(s)
- Saikat Chakraborty
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indiana University-Purdue University Indianapolis, 635 Barnhill Dr., MS360A, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Groschner K. Polymodal TRPC signaling: Emerging role in phenotype switching and tissue remodeling. Commun Integr Biol 2011; 3:393-5. [PMID: 21057623 DOI: 10.4161/cib.3.5.12131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 04/21/2010] [Indexed: 12/17/2022] Open
Abstract
TRPC proteins have been implicated in a large array of Ca(2+) signaling processes and are considered as pore-forming subunits of unique polymodal channel sensors. The mechanisms of TRPC activation are so far incompletely understood but appear to involve a concert of signals that are generated typically downstream of receptor-mediated activation of phospholipase C. Specifically for the TRPC1/4/5 subfamily the activating scenario is ill-defined and appears enigmatic due to the observation of multiple modes of activation. TRPC4 was initially described as a store-operated cation channel and was repeatedly proposed as a pivotal element of the store-operated signaling pathways of various tissues. However, classical reconstitution of TRPC4 complexes in expression systems as well as recent knock-down strategies provided evidence against store-dependent regulation of this channel and raised considerable doubt in its proposed prominent role agonist-induced Ca(2+) signaling. Recent analysis of the function of TRPC4 in vascular endothelial cells of divergent phenotype revealed a novel aspect of TRPC signaling, extending the current concept of TRPC regulation by a phenotype-dependent switch between Ca(2+) transport and a potential intracellular scaffold function of the TRPC protein.
Collapse
Affiliation(s)
- Klaus Groschner
- Institute of Pharmaceutical Sciences-Pharmacology and Toxicology; University of Graz; Graz, Austria
| |
Collapse
|
31
|
AL-Shawaf E, Tumova S, Naylor J, Majeed Y, Li J, Beech DJ. GVI phospholipase A2 role in the stimulatory effect of sphingosine-1-phosphate on TRPC5 cationic channels. Cell Calcium 2011; 50:343-50. [PMID: 21742378 PMCID: PMC3195672 DOI: 10.1016/j.ceca.2011.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 06/08/2011] [Accepted: 06/09/2011] [Indexed: 11/16/2022]
Abstract
The Transient Receptor Potential Canonical 5 (TRPC5) protein forms calcium-permeable cationic channels that are stimulated by G protein-coupled receptor agonists. The signaling pathways of such agonist effects are poorly understood. Here we investigated the potential for involvement of lysophosphatidylcholine (LPC) and arachidonic acid generated by group 6 (GVI) phospholipase A2 (PLA2) enzymes, focusing on stimulation of TRPC5 by sphingosine-1-phosphate (S1P) which acts via a pertussis toxin-sensitive (Gi/o protein) pathway without Ca2+-release. Experiments were on HEK 293 cells containing conditional expression of human TRPC5. Channel activity was recorded using an intracellular calcium indicator or whole-cell patch-clamp and PLA2 activity was detected using 3H-arachidonic acid. S1P stimulated PLA2 and TRPC5 activities. Both effects were suppressed by the GVI PLA2 inhibitor bromoenol lactone. Knock-down of GVI PLA2 by RNA interference suppressed channel activity evoked by S1P whereas activity evoked by the direct channel stimulator LPC was unaffected. Arachidonic acid did not stimulate the channels. Prior exposure of channels to LPC but not arachidonic acid suppressed channel activity evoked by S1P but not gadolinium, a putative direct stimulator of the channels. The data suggest roles of LPC and GVI PLA2 in S1P-evoked TRPC5 activity.
Collapse
Affiliation(s)
- Eman AL-Shawaf
- Multidisciplinary Cardiovascular Research Centre and the Institute of Membrane & Systems Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | | | | | | | | | | |
Collapse
|
32
|
Sung TS, Jeon JP, Kim BJ, Hong C, Kim SY, Kim J, Jeon JH, Kim HJ, Suh CK, Kim SJ, So I. Molecular determinants of PKA-dependent inhibition of TRPC5 channel. Am J Physiol Cell Physiol 2011; 301:C823-32. [PMID: 21734191 DOI: 10.1152/ajpcell.00351.2010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Canonical transient receptor potential (TRPC) channels are Ca(2+)-permeable, nonselective cation channels that are widely expressed in numerous cell types. Here, we demonstrate a new mechanism of TPRC isofom 5 (TRPC5) regulation, via cAMP signaling via Gα(s). Monovalent cation currents in human embryonic kidney-293 cells transfected with TRPC5 were induced by G protein activation with intracellular perfusion of GTPγS or by muscarinic stimulation. This current could be inhibited by a membrane-permeable analog of cAMP, 8-bromo-cAMP, by isoproterenol, by a constitutively active form of Gα(s) [Gα(s) (Q227L)], and by forskolin. These inhibitory effects were blocked by the protein kinase A (PKA) inhibitors, KT-5720 and H-89, as well as by two point mutations at consensus PKA phosphorylation sites on TRPC5 (S794A and S796A). Surface expression of several mutated versions of TRPC5, quantified using surface biotinylation, were not affected by Gα(s) (Q227L), suggesting that trafficking of this channel does not underlie the regulation we report. This mechanism of inhibition was also found to be important for the closely related channel, TRPC4, in particular for TRPC4α, although TRPC4β was also affected. However, this form of regulation was not found to be involved in TRPC6 and transient receptor potential vanilloid 6 function. In murine intestinal smooth muscle cells, muscarinic stimulation-induced cation currents were mediated by TRPC4 (>80%) and TRPC6. In murine intestinal smooth muscle cells, 8-bromo-cAMP, adrenaline, and isoproterenol decreased nonselective cation currents activated by muscarinic stimulation or GTPγS. Together, these results suggest that TRPC5 is directly phosphorylated by G(s)/cAMP/PKA at positions S794 and S796. This mechanism may be physiologically important in visceral tissues, where muscarinic receptor and β(2)-adrenergic receptor are involved in the relaxation and contraction of smooth muscles.
Collapse
Affiliation(s)
- Tae Sik Sung
- Department of Physiology, Seoul National University College of Medicine, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mori Y, Kajimoto T, Nakao A, Takahashi N, Kiyonaka S. Receptor Signaling Integration by TRP Channelsomes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:373-89. [DOI: 10.1007/978-94-007-0265-3_21] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Reboreda A, Jiménez-Díaz L, Navarro-López JD. TRP channels and neural persistent activity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:595-613. [PMID: 21290318 DOI: 10.1007/978-94-007-0265-3_32] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
One of the integrative properties of the nervous system is its capability to, by transient motor commands or brief sensory stimuli, evoke persistent neuronal changes, mainly as a sustained, tonic action potential firing. This neural activity, named persistent activity, is found in a good number of brain regions and is thought to be a neural substrate for short-term storage and accumulation of sensory or motor information [1]. Examples of this persistent neural activity have been reported in prefrontal [2] and entorhinal [3] cortices, as part of the neural mechanisms involved in short-term working memory [4]. Interestingly, the general organization of the motor systems assumes the presence of bursts of short-lasting motor commands encoding movement characteristics such as velocity, duration, and amplitude, followed by a maintained tonic firing encoding the position at which the moving appendage should be maintained [5, 6]. Generation of qualitatively similar sustained discharges have also been found in spinal and supraspinal regions in relation to pain processing [7, 8]. Thus, persistent neural activity seems to be necessary for both behavioral (positions of fixation) and cognitive (working memory) processes. Persistent firing mechanisms have been proposed to involve the participation of a non-specific cationic current (CAN current) mainly mediated by activation of TRPC channels. Because the function and generation of persistent activity is still poorly understood, here we aimed to review and discuss the putative role of TRP-like channels on its generation and/or maintenance.
Collapse
Affiliation(s)
- Antonio Reboreda
- Section of Physiology, Department of Functional Biology and Health Sciences, School of Biology, University of Vigo, Campus Lagoas-Marcosende 36310 Vigo (Pontevedra), Spain.
| | | | | |
Collapse
|
35
|
Gees M, Colsoul B, Nilius B. The role of transient receptor potential cation channels in Ca2+ signaling. Cold Spring Harb Perspect Biol 2010; 2:a003962. [PMID: 20861159 DOI: 10.1101/cshperspect.a003962] [Citation(s) in RCA: 310] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The 28 mammalian members of the super-family of transient receptor potential (TRP) channels are cation channels, mostly permeable to both monovalent and divalent cations, and can be subdivided into six main subfamilies: the TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin), TRPML (mucolipin), and the TRPA (ankyrin) groups. TRP channels are widely expressed in a large number of different tissues and cell types, and their biological roles appear to be equally diverse. In general, considered as polymodal cell sensors, they play a much more diverse role than anticipated. Functionally, TRP channels, when activated, cause cell depolarization, which may trigger a plethora of voltage-dependent ion channels. Upon stimulation, Ca2+ permeable TRP channels generate changes in the intracellular Ca2+ concentration, [Ca2+]i, by Ca2+ entry via the plasma membrane. However, more and more evidence is arising that TRP channels are also located in intracellular organelles and serve as intracellular Ca2+ release channels. This review focuses on three major tasks of TRP channels: (1) the function of TRP channels as Ca2+ entry channels; (2) the electrogenic actions of TRPs; and (3) TRPs as Ca2+ release channels in intracellular organelles.
Collapse
Affiliation(s)
- Maarten Gees
- KU Leuven, Department of Molecular Cell Biology, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, bus 802, Leuven, Belgium
| | | | | |
Collapse
|
36
|
Dadon D, Minke B. Cellular functions of transient receptor potential channels. Int J Biochem Cell Biol 2010; 42:1430-45. [PMID: 20399884 DOI: 10.1016/j.biocel.2010.04.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 04/08/2010] [Accepted: 04/08/2010] [Indexed: 11/26/2022]
Abstract
Transient Receptor Potential channels are polymodal cellular sensors involved in a wide variety of cellular processes, mainly by increasing cellular Ca(2+). In this review we focus on the roles of these channels in: (i) cell death (ii) proliferation and differentiation and (iii) transmitter release. Cell death: Ca(2+) influx participates in apoptotic and necrotic cell death. The Ca(2+) permeability and high sensitivity of part of these channels to oxidative/metabolic stress make them important participants in cell death. Several examples are given. Transient Receptor Potential Melastatin 2 is activated by H(2)O(2), inducing cell death through an increase in cellular Ca(2+) and activation of Poly ADP-Ribose Polymerase. Exposure of cultured cortical neurons to oxygen-glucose deprivation, in vitro, causes cell death via cation influx, mediated by Transient Receptor Potential Melastatin 7. Metabolic stress constitutively activates the Ca(2+) permeable Transient Receptor Potential channels of Drosophila photoreceptor in the dark, potentially leading to retinal degeneration. Similar sensitivity to metabolic stress characterizes several mammalian Transient Receptor Potential Canonical channels. Proliferation and differentiation: The rise in cytosolic Ca(2+) induces cell growth, differentiation and proliferation via activation of several transcription factors. Activating a variety of store operated and Transient Receptor Potential channels cause a rise in cytosolic Ca(2+), making these channels components involved in proliferation and differentiation. Transmitter release: Transient Receptor Potential Melastatin 7 channels reside in synaptic vesicles and regulate neurotransmitter release by a mechanism that is not entirely clear. All the above features of Transient Receptor Potential channels make them crucial components in important, sometimes conflicting, cellular processes that still need to be explored.
Collapse
Affiliation(s)
- Daniela Dadon
- Department of Medical Neurobiology, The Institute of Medical Research Israel-Canada and the Kühne Minerva Center, for Studies of Visual Transduction, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
| | | |
Collapse
|
37
|
Al-Shawaf E, Naylor J, Taylor H, Riches K, Milligan CJ, O'Regan D, Porter KE, Li J, Beech DJ. Short-term stimulation of calcium-permeable transient receptor potential canonical 5-containing channels by oxidized phospholipids. Arterioscler Thromb Vasc Biol 2010; 30:1453-9. [PMID: 20378846 DOI: 10.1161/atvbaha.110.205666] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine whether calcium-permeable channels are targets for the oxidized phospholipids: 1-palmitoyl-2-glutaroyl-phosphatidylcholine (PGPC) and 1-palmitoyl-2-oxovaleroyl-phosphatidylcholine (POVPC). METHODS AND RESULTS Oxidized phospholipids are key factors in inflammation and associated diseases, including atherosclerosis; however, the initial reception mechanisms for cellular responses to the factors are poorly understood. Low micromolar concentrations of PGPC and POVPC evoked increases in intracellular calcium in human embryonic kidney 293 cells that overexpressed human transient receptor potential canonical 5 (TRPC5) but not human TRP melastatin (TRPM) 2 or 3. The results of electrophysiological experiments confirmed stimulation of TRPC5. To investigate relevance to endogenous channels, we studied proliferating vascular smooth muscle cells from patients undergoing coronary artery bypass surgery. PGPC and POVPC elicited calcium entry that was inhibited by anti-TRPC5 or anti-TRPC1 antibodies or dominant-negative mutant TRPC5. Calcium release did not occur. The effect was functionally relevant because it enhanced cell migration. The actions of PGPC and POVPC depended on G(i/o) proteins but not on previously identified G protein-coupled receptors for oxidized phospholipids. CONCLUSIONS Stimulation of calcium-permeable TRPC5-containing channels may be an early event in cellular responses to oxidized phospholipids that couples to cell migration and requires an unidentified G protein-coupled receptor.
Collapse
Affiliation(s)
- Eman Al-Shawaf
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, England
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sabourin J, Lamiche C, Vandebrouck A, Magaud C, Rivet J, Cognard C, Bourmeyster N, Constantin B. Regulation of TRPC1 and TRPC4 cation channels requires an alpha1-syntrophin-dependent complex in skeletal mouse myotubes. J Biol Chem 2009; 284:36248-36261. [PMID: 19812031 DOI: 10.1074/jbc.m109.012872] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The dystrophin-associated protein complex (DAPC) is essential for skeletal muscle, and the lack of dystrophin in Duchenne muscular dystrophy results in a reduction of DAPC components such as syntrophins and in fiber necrosis. By anchoring various molecules, the syntrophins may confer a role in cell signaling to the DAPC. Calcium disorders and abnormally elevated cation influx in dystrophic muscle cells have suggested that the DAPC regulates some sarcolemmal cationic channels. We demonstrated previously that mini-dystrophin and alpha1-syntrophin restore normal cation entry in dystrophin-deficient myotubes and that sarcolemmal TRPC1 channels associate with dystrophin and the bound PDZ domain of alpha1-syntrophin. This study shows that small interfering RNA (siRNA) silencing of alpha1-syntrophin dysregulated cation influx in myotubes. Moreover, deletion of the PDZ-containing domain prevented restoration of normal cation entry by alpha1-syntrophin transfection in dystrophin-deficient myotubes. TRPC1 and TRPC4 channels are expressed at the sarcolemma of muscle cells; forced expression or siRNA silencing showed that cation influx regulated by alpha1-syntrophin is supported by TRPC1 and TRPC4. A molecular association was found between TRPC1 and TRPC4 channels and the alpha1-syntrophin-dystrophin complex. TRPC1 and TRPC4 channels may form sarcolemmal channels anchored to the DAPC, and alpha1-syntrophin is necessary to maintain the normal regulation of TRPC-supported cation entry in skeletal muscle. Cation channels with DAPC form a signaling complex that modulates cation entry and may be crucial for normal calcium homeostasis in skeletal muscles.
Collapse
Affiliation(s)
- Jessica Sabourin
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Coralie Lamiche
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Aurelie Vandebrouck
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Christophe Magaud
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Jerome Rivet
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Christian Cognard
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Nicolas Bourmeyster
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Bruno Constantin
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France.
| |
Collapse
|
39
|
Blair NT, Kaczmarek JS, Clapham DE. Intracellular calcium strongly potentiates agonist-activated TRPC5 channels. J Gen Physiol 2009; 133:525-46. [PMID: 19398778 PMCID: PMC2712973 DOI: 10.1085/jgp.200810153] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 04/13/2009] [Indexed: 11/20/2022] Open
Abstract
TRPC5 is a calcium (Ca(2+))-permeable nonselective cation channel expressed in several brain regions, including the hippocampus, cerebellum, and amygdala. Although TRPC5 is activated by receptors coupled to phospholipase C, the precise signaling pathway and modulatory signals remain poorly defined. We find that during continuous agonist activation, heterologously expressed TRPC5 currents are potentiated in a voltage-dependent manner ( approximately 5-fold at positive potentials and approximately 25-fold at negative potentials). The reversal potential, doubly rectifying current-voltage relation, and permeability to large cations such as N-methyl-d-glucamine remain unchanged during this potentiation. The TRPC5 current potentiation depends on extracellular Ca(2+): replacement by Ba(2+) or Mg(2+) abolishes it, whereas the addition of 10 mM Ca(2+) accelerates it. The site of action for Ca(2+) is intracellular, as simultaneous fura-2 imaging and patch clamp recordings indicate that potentiation is triggered at approximately 1 microM [Ca(2+)]. This potentiation is prevented when intracellular Ca(2+) is tightly buffered, but it is promoted when recording with internal solutions containing elevated [Ca(2+)]. In cell-attached and excised inside-out single-channel recordings, increases in internal [Ca(2+)] led to an approximately 10-20-fold increase in channel open probability, whereas single-channel conductance was unchanged. Ca(2+)-dependent potentiation should result in TRPC5 channel activation preferentially during periods of repetitive firing or coincident neurotransmitter receptor activation.
Collapse
Affiliation(s)
- Nathaniel T Blair
- Howard Hughes Medical Institute, Department of Cardiology and Manton Center for Orphan Disease, Children's Hospital Boston, and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
40
|
Gomis A, Soriano S, Belmonte C, Viana F. Hypoosmotic- and pressure-induced membrane stretch activate TRPC5 channels. J Physiol 2008; 586:5633-49. [PMID: 18832422 DOI: 10.1113/jphysiol.2008.161257] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transient receptor potential (TRP) channels mediate a wide array of sensory functions. We investigated the role of TRPC5, a poorly characterized channel widely expressed in the central and peripheral nervous system, as a potential osmosensory protein. Here we show that hypoosmotic stimulation activates TRPC5 channels resulting in a large calcium influx. The response to osmotically induced membrane stretch is blocked by GsMTx-4, an inhibitor of stretch activated ion channels. Direct hypoosmotic activation of TRPC5 is independent of phospholipase C function. However, the osmotic response is inhibited in a cell line in which PIP(2) levels are reduced by regulated overexpression of a lipid phosphatase. The response was restored by increasing intracellular PIP(2) levels through the patch pipette. The mechano-sensitivity of the channel was probed in the whole-cell configuration by application of steps of positive pressure through the patch pipette. Pressure-induced membrane stretch also activated TRPC5 channels, suggesting its role as a transducer of osmo-mechanical stimuli. We also demonstrated the expression of TRPC5 in sensory neurones which together with the osmo-mechanical characteristics of TRPC5 channels suggest its putative role in mechanosensory transduction events.
Collapse
Affiliation(s)
- Ana Gomis
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández. Av. Ramón y Cajal s/n. 03550 Sant Joan d'Alacant, Alicante, Spain.
| | | | | | | |
Collapse
|
41
|
Kim BJ, Kim MT, Jeon JH, Kim SJ, So I. Involvement of phosphatidylinositol 4,5-bisphosphate in the desensitization of canonical transient receptor potential 5. Biol Pharm Bull 2008; 31:1733-1738. [PMID: 18758068 DOI: 10.1248/bpb.31.1733] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The classic transient receptor potential channel (TRPC) is a candidate for Ca(2+)-permeable cation channel in mammalian cells. TRPC5 is desensitized rapidly after activation by G protein-coupled receptor. Here we investigate the mechanisms of desensitization of TRPC5 using patch-clamp recording. TRPC5 was initially activated by muscarinic stimulation using 50 microM carbachol (CCh) and decayed rapidly in the presence of CCh (desensitization). Intracellularly-applied phosphatidylinositol 4,5-bisphosphate (PIP(2)) slowed the rate of desensitization. In contrast, several other phosphoinositides, including PI(3,4)P(2), PI(3,5)P(2), PI(3,4,5)P(3) and PI(4)P, had no effect on the desensitization of the TRPC5 current. This indicates that PIP(2) attenuates the desensitization of the TRPC5 current in a highly selective manner. Neither wortmannin, an inhibitor of phosphatidylinositol 4-kinase, or poly-L-lysine (PLL), a scavenger of PIP(2), had any effect on desensitization of the TRPC5 current. PIP(2) breakdown appears to be a required step in the desensitization of TRPC5 current, but PIP(2) depletion alone was insufficient for channel desensitization. TRPC5 was inhibited by cytochalasin D treatment. In mouse ileal myocytes, the desensitization of CCh-activated inward current (I(CCh)) also slowed in the presence of PIP(2) in recording pipettes. These results indicate that PIP(2) is involved in the desensitization of TRPC5 currents.
Collapse
Affiliation(s)
- Byung Joo Kim
- Center for Bio-Artificial Muscle and Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
42
|
Obukhov AG, Nowycky MC. TRPC5 channels undergo changes in gating properties during the activation-deactivation cycle. J Cell Physiol 2008; 216:162-71. [PMID: 18247362 DOI: 10.1002/jcp.21388] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
TRPC5 are non-specific cation channels activated through phospholipase C-dependent pathways, although the precise gating mechanism is unknown. TRPC5 current-voltage relationships (I-Vs) change systematically during the activation-deactivation cycle, shifting between outwardly rectifying and doubly rectifying shapes. Since several TRP family members exhibit voltage-dependent properties, we investigated whether the various I-V relationships were due to changes in gating. Using patch-clamp recordings of rat TRPC5 transfected HEK293 cells, we found that TRPC5 currents had distinct biophysical characteristics correlated with individual I-V shapes, a phenomenon we call 'phases.' At rest, channels were closed at most potentials, although strong depolarizations (>+80 mV) stimulated small outward currents (Phase 0). For 10-15 sec after activation, voltage steps evoked small inward and large outward currents with time- and voltage-dependent kinetics (Phase 1, outwardly-rectifying I-Vs). At maximal inward amplitude, currents were voltage-independent at all potentials (Phase 2, doubly-rectifying I-Vs owing to Mg2+ block). During desensitization (Phase 3), currents reverted to a Phase 1-like voltage-dependence. La3+ ions potentiated inward TRPC5 currents by promoting a reversible transition from Phase 3 to Phase 2. Single channel recordings revealed asymmetric conductance properties with values of approximately 40 pS at negative potentials and approximately 130 pS at >+60 mV. Mutation of D633, a cytoplasmic residue that mediates Mg2+ block, decreased channel activity at negative potentials during Phase 2. We conclude that TRPC5 gating properties can switch reversibly between voltage-dependent and voltage-independent states. The modulation of phase transitions by external agents such as La3+ and EBP50, a scaffolding protein, may constitute a novel mechanism for regulation of channel activity.
Collapse
Affiliation(s)
- Alexander G Obukhov
- Department Pharmacology & Physiology, UMDNJ, New Jersey Medical School, Newark, New Jersey 07103, USA
| | | |
Collapse
|
43
|
Pedersen SF, Owsianik G, Nilius B. TRP channels: an overview. Cell Calcium 2008; 38:233-52. [PMID: 16098585 DOI: 10.1016/j.ceca.2005.06.028] [Citation(s) in RCA: 544] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Accepted: 06/28/2005] [Indexed: 12/12/2022]
Abstract
The TRP ("transient receptor potential") family of ion channels now comprises more than 30 cation channels, most of which are permeable for Ca2+, and some also for Mg2+. On the basis of sequence homology, the TRP family can be divided in seven main subfamilies: the TRPC ('Canonical') family, the TRPV ('Vanilloid') family, the TRPM ('Melastatin') family, the TRPP ('Polycystin') family, the TRPML ('Mucolipin') family, the TRPA ('Ankyrin') family, and the TRPN ('NOMPC') family. The cloning and characterization of members of this cation channel family has exploded during recent years, leading to a plethora of data on the roles of TRPs in a variety of tissues and species, including mammals, insects, and yeast. The present review summarizes the most pertinent recent evidence regarding the structural and functional properties of TRP channels, focusing on the regulation and physiology of mammalian TRPs.
Collapse
Affiliation(s)
- Stine Falsig Pedersen
- Department of Biochemistry, Institute for Molecular Biology and Physiology, University of Copenhagen, Denmark
| | | | | |
Collapse
|
44
|
Otsuguro KI, Tang J, Tang Y, Xiao R, Freichel M, Tsvilovskyy V, Ito S, Flockerzi V, Zhu MX, Zholos AV. Isoform-specific inhibition of TRPC4 channel by phosphatidylinositol 4,5-bisphosphate. J Biol Chem 2008; 283:10026-36. [PMID: 18230622 DOI: 10.1074/jbc.m707306200] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Full-length transient receptor potential (TRP) cation channel TRPC4alpha and shorter TRPC4beta lacking 84 amino acids in the cytosolic C terminus are expressed in smooth muscle and endothelial cells where they regulate membrane potential and Ca(2+) influx. In common with other "classical" TRPCs, TRPC4 is activated by G(q)/phospholipase C-coupled receptors, but the underlying mechanism remains elusive. Little is also known about any isoform-specific channel regulation. Here we show that TRPC4alpha but not TRPC4beta was strongly inhibited by intracellularly applied phosphatidylinositol 4,5-bisphosphate (PIP(2)). In contrast, several other phosphoinositides (PI), including PI(3,4)P(2), PI(3,5)P(2), and PI(3,4,5)P(3), had no effect or even potentiated TRPC4alpha indicating that PIP(2) inhibits TRPC4alpha in a highly selective manner. We show that PIP(2) binds to the C terminus of TRPC4alpha but not that of TRPC4beta in vitro. Its inhibitory action was dependent on the association of TRPC4alpha with actin cytoskeleton as it was prevented by cytochalasin D treatment or by the deletion of the C-terminal PDZ-binding motif (Thr-Thr-Arg-Leu) that links TRPC4 to F-actin through the sodium-hydrogen exchanger regulatory factor and ezrin. PIP(2) breakdown appears to be a required step in TRPC4alpha channel activation as PIP(2) depletion alone was insufficient for channel opening, which additionally required Ca(2+) and pertussis toxin-sensitive G(i/o) proteins. Thus, TRPC4 channels integrate a variety of G-protein-dependent stimuli, including a PIP(2)/cytoskeleton dependence reminiscent of the TRPC4-like muscarinic agonist-activated cation channels in ileal myocytes.
Collapse
Affiliation(s)
- Ken-ichi Otsuguro
- Cardiovascular Biomedical Research Centre, School of Medicine and Dentistry, Queen's University Belfast, Belfast, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Odell AF, Van Helden DF, Scott JL. The spectrin cytoskeleton influences the surface expression and activation of human transient receptor potential channel 4 channels. J Biol Chem 2007; 283:4395-407. [PMID: 18048348 DOI: 10.1074/jbc.m709729200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Despite over a decade of research, only recently have the mechanisms governing transient receptor potential channel (TRPC) channel function begun to emerge, with an essential role for accessory proteins in this process. We previously identified a tyrosine phosphorylation event as critical in the plasma membrane translocation and activation of hTRPC4 channels following epidermal growth factor (EGF) receptor activation. To further characterize the signaling events underlying this process, a yeast-two hybrid screen was performed on the C terminus of hTRPC4. The intracellular C-terminal region from proline 686 to leucine 977 was used to screen a human brain cDNA library. Two members of the spectrin family, alphaII- and betaV-spectrin, were identified as binding partners. The interaction of hTRPC4 with alphaII-spectrin and betaV-spectrin was confirmed by glutathione S-transferase pulldown and co-immunoprecipitation experiments. Deletion analysis identified amino acids 730-758 of hTRPC4 as critical for the interaction with this region located within a coiled-coil domain, juxtaposing the Ca(2+)/calmodulin- and IP(3)R-binding region (CIRB domain). This region is deleted in the proposed deltahTRPC4 splice variant form, which failed to undergo both EGF-induced membrane insertion and activation, providing a genetic mechanism for regulating channel activity. We also demonstrate that the exocytotic insertion and activation of hTRPC4 following EGF application is accompanied by dissociation from alphaII-spectrin. Furthermore, depletion of alphaII-spectrin by small interference RNA reduces the basal surface expression of alphahTRPC4 and prevents the enhanced membrane insertion in response to EGF application. Importantly, depletion of alphaII-spectrin did not affect the expression of the delta variant. Taken together, these results demonstrate that a direct interaction between hTRPC4 and the spectrin cytoskeleton is involved in the regulation of hTRPC4 surface expression and activation.
Collapse
Affiliation(s)
- Adam F Odell
- School of Biomedical Sciences, Faculty of Health, University of Newcastle, Level 5, MSB, University Drive, New South Wales 2308, Australia.
| | | | | |
Collapse
|
46
|
Wu G, Lu ZH, Obukhov AG, Nowycky MC, Ledeen RW. Induction of calcium influx through TRPC5 channels by cross-linking of GM1 ganglioside associated with alpha5beta1 integrin initiates neurite outgrowth. J Neurosci 2007; 27:7447-58. [PMID: 17626205 PMCID: PMC6672619 DOI: 10.1523/jneurosci.4266-06.2007] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Previous studies demonstrated that cross-linking of GM1 ganglioside with multivalent ligands, such as B subunit of cholera toxin (CtxB), induced Ca2+ influx through an unidentified, voltage-independent channel in several cell types. Application of CtxB to undifferentiated NG108-15 cells resulted in outgrowth of axon-like neurites in a Ca2+ influx-dependent manner. In this study, we demonstrate that CtxB-induced Ca2+ influx is mediated by TRPC5 channels, naturally expressed in these cells and primary neurons. Both Ca2+ influx and neurite induction were blocked by TRPC5 small interfering RNA (siRNA). Pretreatment of NG108-15 cells with neuraminidase increased cell-surface GM1 and greatly enhanced the signal. GM1 was not directly associated with TRPC5 but rather with alpha5beta1 integrin, which opened the channel through a signaling sequence after cross-linking of the GM1/integrin complex. This cascade included autophosphorylation of focal adhesion kinase and subsequent activation of phospholipase Cgamma (PLCgamma) and phosphoinositide-3 kinase [PI(3)K]. Pharmacological blockers that inhibited tyrosine kinase, PLC, and PI(3)K suppressed both CtxB-induced Ca2+ influx and neurite outgrowth. These were also suppressed by SK&F96365, a nonspecific transient receptor potential channel blocker. Confocal immunocytochemistry revealed that GM1 cross-linking induced colocalization of GM1 with these signaling elements in sprouting regions of plasma membrane. In primary cerebellar granular neurons (CGNs), TRPC5 was detected at 2 d in vitro (2 DIV), a stage corresponding to CtxB-stimulated Ca2+ influx. Neurite outgrowth in CGNs, determined at 3 DIV, was accelerated by CtxB and suppressed by TRPC5 siRNA and the above blockers. The crucial role of GM1 was indicated with CGNs from ganglio-series null mice, in which growth of axons was significantly retarded.
Collapse
Affiliation(s)
- Gusheng Wu
- Department of Neurology and Neurosciences, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103, USA.
| | | | | | | | | |
Collapse
|
47
|
Abstract
NHE3 is the brush-border (BB) Na+/H+exchanger of small intestine, colon, and renal proximal tubule which is involved in large amounts of neutral Na+absorption. NHE3 is a highly regulated transporter, being both stimulated and inhibited by signaling that mimics the postprandial state. It also undergoes downregulation in diarrheal diseases as well as changes in renal disorders. For this regulation, NHE3 exists in large, multiprotein complexes in which it associates with at least nine other proteins. This review deals with short-term regulation of NHE3 and the identity and function of its recognized interacting partners and the multiprotein complexes in which NHE3 functions.
Collapse
Affiliation(s)
- Mark Donowitz
- Department of Medicine, GI Division, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
48
|
Ambudkar IS, Ong HL. Organization and function of TRPC channelosomes. Pflugers Arch 2007; 455:187-200. [PMID: 17486362 DOI: 10.1007/s00424-007-0252-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2007] [Accepted: 03/10/2007] [Indexed: 12/20/2022]
Abstract
TRPC proteins constitute a family of conserved Ca2+-permeable cation channels which are activated in response to agonist-stimulated PIP2 hydrolysis. These channels were initially proposed to be components of the store-operated calcium entry channel (SOC). Subsequent studies have provided substantial evidence that some TRPCs contribute to SOC activity. TRPC proteins have also been shown to form agonist-stimulated calcium entry channels that are not store-operated but are likely regulated by PIP2 or diacylglycerol. Further, and consistent with the presently available data, selective homomeric or heteromeric interactions between TRPC monomers generate distinct agonist-stimulated cation permeable channels. We suggest that interaction between TRPC monomers, as well as the association of these channels with accessory proteins, determines their mode of regulation as well as their cellular localization and function. Currently identified accessory proteins include key Ca2+ signaling proteins as well as proteins involved in vesicle trafficking, cytoskeletal interactions, and scaffolding. Studies reported until now demonstrate that TRPC proteins are segregated into specific Ca2+ signaling complexes which can generate spatially and temporally controlled [Ca2+]i signals. Thus, the functional organization of TRPC channelosomes dictates not only their regulation by extracellular stimuli but also serves as a platform to coordinate specific downstream cellular functions that are regulated as a consequence of Ca2+ entry. This review will focus on the accessory proteins of TRPC channels and discuss the functional implications of TRPC channelosomes and their assembly in microdomains.
Collapse
Affiliation(s)
- Indu S Ambudkar
- Secretory Physiology Section, Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
49
|
Cayouette S, Boulay G. Intracellular trafficking of TRP channels. Cell Calcium 2007; 42:225-32. [PMID: 17368756 DOI: 10.1016/j.ceca.2007.01.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 01/30/2007] [Accepted: 01/31/2007] [Indexed: 11/18/2022]
Abstract
Thirteen years ago, it was suggested that exocytotic insertion of store-operated channels into the plasma membrane lead to increased Ca(2+) entry in non-excitable cells upon G protein-coupled or tyrosine kinase receptor stimulation. Since the discovery of the TRP channel superfamily and their involvement in receptor-induced Ca(2+) entry, many studies have shown that different members of the TRP superfamily translocate into the plasma membrane upon stimulation. While the exact molecular mechanism by which TRP channels insert into the plasma membrane is unknown, TRP-binding proteins have been shown to directly regulate this trafficking. This review summarizes recent advances related to the mechanism of TRP channel trafficking, focusing on the role of TRP-binding proteins.
Collapse
Affiliation(s)
- Sylvie Cayouette
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | |
Collapse
|
50
|
Abstract
TRP channels, in particular the TRPC and TRPV subfamilies, have emerged as important constituents of the receptor-activated Ca2+ influx mechanism triggered by hormones, growth factors, and neurotransmitters through activation ofphospholipase C (PLC). Several TRPC channels are also activated by passive depletion of endoplasmic reticulum (ER) Ca2+. Although in several studies the native TRP channels faithfully reproduce the respective recombinant channels, more often the properties of Ca2+ entry and/or the store-operated current are strikingly different from that of the TRP channels expressed in the same cells. The present review aims to discuss this disparity in the context of interaction of TRPC channels with auxiliary proteins that may alter the permeation and regulation of TRPC channels.
Collapse
Affiliation(s)
- K Kiselyov
- Department of Biological Sciences University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | | | |
Collapse
|