1
|
Shin S, Gombedza FC, Awuah Boadi E, Yiu AJ, Roy SK, Bandyopadhyay BC. Reduction of TRPC1/TRPC3 mediated Ca 2+-signaling protects oxidative stress-induced COPD. Cell Signal 2023; 107:110681. [PMID: 37062436 PMCID: PMC10542863 DOI: 10.1016/j.cellsig.2023.110681] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/14/2023] [Accepted: 04/13/2023] [Indexed: 04/18/2023]
Abstract
Oxidative stress is a predisposing factor in Chronic Obstructive Pulmonary Disease (COPD). Specifically, pulmonary epithelial (PE) cells reduce antioxidant capacity during COPD because of the continuous production of reactive oxygen species (ROS). However, the molecular pathogenesis that governs such ROS activity is unclear. Here we show that the dysregulation of intracellular calcium concentration ([Ca2+]i) in PE cells from COPD patients, compared to the healthy PE cells, is associated with the robust functional expressions of Transient Receptor Potential Canonical (TRPC)1 and TRPC3 channels, and Ca2+ entry (SOCE) components, Stromal Interaction Molecule 1 (STIM1) and ORAI1 channels. Additionally, the elevated expression levels of fibrotic, inflammatory, oxidative, and apoptotic markers in cells from COPD patients suggest detrimental pathway activation, thereby reducing the ability of lung remodeling. To further delineate the mechanism, we used human lung epithelial cell line, A549, since the behavior of SOCE and the expression patterns of TRPC1/C3, STIM1, and ORAI1 were much like PE cells. Notably, the knockdown of TRPC1/C3 in A549 cells substantially reduced the SOCE-induced [Ca2+]i rise, and reversed the ROS-mediated oxidative, fibrotic, inflammatory, and apoptotic responses, thus confirming the role of TRPC1/C3 in SOCE driven COPD-like condition. Higher TRPC1/C3, STIM1, and ORAI1 expressions, along with a greater Ca2+ entry, via SOCE in ROS-induced A549 cells, led to the rise in oxidative, fibrotic, inflammatory, and apoptotic gene expression, specifically through the extracellular signal-regulated kinase (ERK) pathway. Abatement of TRPC1 and/or TRPC3 reduced the mobilization of [Ca2+]i and reversed apoptotic gene expression and ERK activation, signifying the involvement of TRPC1/C3. Together these data suggest that TRPC1/C3 and SOCE facilitate the COPD condition through ROS-mediated cell death, thus implicating their likely roles as potential therapeutic targets for COPD. SUMMARY: Alterations in Ca2+ signaling modalities in normal pulmonary epithelial cells exhibit COPD through oxidative stress and cellular injury, compromising repair, which was alleviated through inhibition of store-operated calcium entry. SUBJECT AREA: Calcium, ROS, Cellular signaling, lung disease.
Collapse
Affiliation(s)
- Samuel Shin
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Farai C Gombedza
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Eugenia Awuah Boadi
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Allen J Yiu
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Sanjit K Roy
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America
| | - Bidhan C Bandyopadhyay
- From Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, United States of America.
| |
Collapse
|
2
|
Choi BE, Shin S, Evans S, Singh BB, Bandyopadhyay BC. Ablation of TRPC3 disrupts Ca 2+ signaling in salivary ductal cells and promotes sialolithiasis. Sci Rep 2023; 13:5772. [PMID: 37031239 PMCID: PMC10082769 DOI: 10.1038/s41598-023-32602-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/30/2023] [Indexed: 04/10/2023] Open
Abstract
Clinical studies and structural analyses of salivary stones strongly suggest a linkage between higher saliva calcium (Ca2+) and salivary stone formation, sialolithiasis; however, the process and the mechanism leading to Ca2+ overload during sialolithiasis is not well understood. Here, we show that TRPC3 null (-/-) mice presented with a reduction in Ca2+ entry and current in ductal cells with higher saliva [Ca2+] suggesting diminished transepithelial Ca2+ flux across the salivary ductal cells, leaving more Ca2+ in ductal fluid. Significantly, we found that TRPC3 was expressed in mice and human salivary ductal cells, while intraductal stones were detected in both mice (TRPC3-/-) and patient (sialolithiasis) salivary glands. To identify the mechanism, we found that TRPC3 was crucial in preventing the expression of calcification genes (BMP2/6, Runx2) in ductal cells which may be due to higher extracellular Ca2+ in SMG tissues. Similarly, inflammatory (IL6, NLRP3), fibrotic (FN1, TGFβ1) and apoptotic (Bax1/Bcl2) markers were also elevated, suggesting that the loss of TRPC3 induces genetic changes that leads to salivary gland cell death and induction of inflammatory response. Overall, ablation of TRPC3-/- leads to higher saliva [Ca2+], along with elevated detrimental gene expressions, altogether contributing to salivary gland stone formation.
Collapse
Affiliation(s)
- Bok-Eum Choi
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
| | - Samuel Shin
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC, 20064, USA
| | - Sade Evans
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
| | - Brij B Singh
- Department of Periodontics, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA.
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC, 20064, USA.
| |
Collapse
|
3
|
Staruschenko A, Ma R, Palygin O, Dryer SE. Ion channels and channelopathies in glomeruli. Physiol Rev 2023; 103:787-854. [PMID: 36007181 PMCID: PMC9662803 DOI: 10.1152/physrev.00013.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
An essential step in renal function entails the formation of an ultrafiltrate that is delivered to the renal tubules for subsequent processing. This process, known as glomerular filtration, is controlled by intrinsic regulatory systems and by paracrine, neuronal, and endocrine signals that converge onto glomerular cells. In addition, the characteristics of glomerular fluid flow, such as the glomerular filtration rate and the glomerular filtration fraction, play an important role in determining blood flow to the rest of the kidney. Consequently, disease processes that initially affect glomeruli are the most likely to lead to end-stage kidney failure. The cells that comprise the glomerular filter, especially podocytes and mesangial cells, express many different types of ion channels that regulate intrinsic aspects of cell function and cellular responses to the local environment, such as changes in glomerular capillary pressure. Dysregulation of glomerular ion channels, such as changes in TRPC6, can lead to devastating glomerular diseases, and a number of channels, including TRPC6, TRPC5, and various ionotropic receptors, are promising targets for drug development. This review discusses glomerular structure and glomerular disease processes. It also describes the types of plasma membrane ion channels that have been identified in glomerular cells, the physiological and pathophysiological contexts in which they operate, and the pathways by which they are regulated and dysregulated. The contributions of these channels to glomerular disease processes, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, as well as the development of drugs that target these channels are also discussed.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
- James A. Haley Veterans Hospital, Tampa, Florida
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, Texas
| |
Collapse
|
4
|
Sun Y, Zboril EK, De La Chapa JJ, Chai X, Da Conceicao VN, Valdez MC, McHardy SF, Gonzales CB, Singh BB. Inhibition of Ca 2+ entry by capsazepine analog CIDD-99 prevents oral squamous carcinoma cell proliferation. Front Physiol 2022; 13:969000. [PMID: 36187775 PMCID: PMC9521718 DOI: 10.3389/fphys.2022.969000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Oral cancer patients have a poor prognosis, with approximately 66% of patients surviving 5-years after diagnosis. Treatments for oral cancer are limited and have many adverse side effects; thus, further studies are needed to develop drugs that are more efficacious. To achieve this objective, we developed CIDD-99, which produces cytotoxic effects in multiple oral squamous cell carcinoma (OSCC) cell lines. While we demonstrated that CIDD-99 induces ER stress and apoptosis in OSCC, the mechanism was unclear. Investigation of the Bcl-family of proteins showed that OSCC cells treated with CIDD-99 undergo downregulation of Bcl-XL and Bcl-2 anti-apoptotic proteins and upregulation of Bax (pro-apoptotic). Importantly, OSCC cells treated with CIDD-99 displayed decreased calcium signaling in a dose and time-dependent manner, suggesting that blockage of calcium signaling is the key mechanism that induces cell death in OSCC. Indeed, CIDD-99 anti-proliferative effects were reversed by the addition of exogenous calcium. Moreover, electrophysiological properties further established that calcium entry was via the non-selective TRPC1 channel and prolonged CIDD-99 incubation inhibited STIM1 expression. CIDD-99 inhibition of calcium signaling also led to ER stress and inhibited mitochondrial complexes II and V in vitro. Taken together, these findings suggest that inhibition of TRPC mediates induction of ER stress and mitochondrial dysfunction as a part of the cellular response to CIDD-99 in OSCC.
Collapse
Affiliation(s)
- Yuyang Sun
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Emily K. Zboril
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jorge J. De La Chapa
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Xiufang Chai
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | | | - Matthew C. Valdez
- Department of Chemistry and the Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Stanton F. McHardy
- Department of Chemistry and the Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Cara B. Gonzales
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Brij B. Singh
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
5
|
Distribution and Assembly of TRP Ion Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1349:111-138. [PMID: 35138613 DOI: 10.1007/978-981-16-4254-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the last several decades, a large family of ion channels have been identified and studied intensively as cellular sensors for diverse physical and/or chemical stimuli. Named transient receptor potential (TRP) channels, they play critical roles in various aspects of cellular physiology. A large number of human hereditary diseases are found to be linked to TRP channel mutations, and their dysregulations lead to acute or chronical health problems. As TRP channels are named and categorized mostly based on sequence homology rather than functional similarities, they exhibit substantial functional diversity. Rapid advances in TRP channel study have been made in recent years and reported in a vast body of literature; a summary of the latest advancements becomes necessary. This chapter offers an overview of current understandings of TRP channel distribution and subunit assembly.
Collapse
|
6
|
Zergane M, Kuebler WM, Michalick L. Heteromeric TRP Channels in Lung Inflammation. Cells 2021; 10:cells10071654. [PMID: 34359824 PMCID: PMC8307017 DOI: 10.3390/cells10071654] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/09/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Activation of Transient Receptor Potential (TRP) channels can disrupt endothelial barrier function, as their mediated Ca2+ influx activates the CaM (calmodulin)/MLCK (myosin light chain kinase)-signaling pathway, and thereby rearranges the cytoskeleton, increases endothelial permeability and thus can facilitate activation of inflammatory cells and formation of pulmonary edema. Interestingly, TRP channel subunits can build heterotetramers, whereas heteromeric TRPC1/4, TRPC3/6 and TRPV1/4 are expressed in the lung endothelium and could be targeted as a protective strategy to reduce endothelial permeability in pulmonary inflammation. An update on TRP heteromers and their role in lung inflammation will be provided with this review.
Collapse
Affiliation(s)
- Meryam Zergane
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.Z.); (L.M.)
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.Z.); (L.M.)
- German Centre for Cardiovascular Research (DZHK), 10785 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
- The Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Department of Surgery and Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence:
| | - Laura Michalick
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.Z.); (L.M.)
- German Centre for Cardiovascular Research (DZHK), 10785 Berlin, Germany
| |
Collapse
|
7
|
Czeredys M. Dysregulation of Neuronal Calcium Signaling via Store-Operated Channels in Huntington's Disease. Front Cell Dev Biol 2020; 8:611735. [PMID: 33425919 PMCID: PMC7785827 DOI: 10.3389/fcell.2020.611735] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder that is characterized by motor, cognitive, and psychiatric problems. It is caused by a polyglutamine expansion in the huntingtin protein that leads to striatal degeneration via the transcriptional dysregulation of several genes, including genes that are involved in the calcium (Ca2+) signalosome. Recent research has shown that one of the major Ca2+ signaling pathways, store-operated Ca2+ entry (SOCE), is significantly elevated in HD. SOCE refers to Ca2+ flow into cells in response to the depletion of endoplasmic reticulum Ca2+ stores. The dysregulation of Ca2+ homeostasis is postulated to be a cause of HD progression because the SOCE pathway is indirectly and abnormally activated by mutant huntingtin (HTT) in γ-aminobutyric acid (GABA)ergic medium spiny neurons (MSNs) from the striatum in HD models before the first symptoms of the disease appear. The present review summarizes recent studies that revealed a relationship between HD pathology and elevations of SOCE in different models of HD, including YAC128 mice (a transgenic model of HD), cellular HD models, and induced pluripotent stem cell (iPSC)-based GABAergic medium spiny neurons (MSNs) that are obtained from adult HD patient fibroblasts. SOCE in MSNs was shown to be mediated by currents through at least two different channel groups, Ca2+ release-activated Ca2+ current (ICRAC) and store-operated Ca2+ current (ISOC), which are composed of stromal interaction molecule (STIM) proteins and Orai or transient receptor potential channel (TRPC) channels. Their role under physiological and pathological conditions in HD are discussed. The role of Huntingtin-associated protein 1 isoform A in elevations of SOCE in HD MSNs and potential compounds that may stabilize elevations of SOCE in HD are also summarized. Evidence is presented that shows that the dysregulation of molecular components of SOCE or pathways upstream of SOCE in HD MSN neurons is a hallmark of HD, and these changes could lead to HD pathology, making them potential therapeutic targets.
Collapse
Affiliation(s)
- Magdalena Czeredys
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Abstract
Transient receptor potential (TRP) channels comprise a diverse family of ion channels, the majority of which are calcium permeable and show sophisticated regulatory patterns in response to various environmental cues. Early studies led to the recognition of TRP channels as environmental and chemical sensors. Later studies revealed that TRP channels mediated the regulation of intracellular calcium. Mutations in TRP channel genes result in abnormal regulation of TRP channel function or expression, and interfere with normal spatial and temporal patterns of intracellular local Ca2+ distribution. The resulting dysregulation of multiple downstream effectors, depending on Ca2+ homeostasis, is associated with hallmarks of cancer pathophysiology, including enhanced proliferation, survival and invasion of cancer cells. These findings indicate that TRP channels affect multiple events that control cellular fate and play a key role in cancer progression. This review discusses the accumulating evidence supporting the role of TRP channels in tumorigenesis, with emphasis on prostate cancer. [BMB Reports 2020; 53(3): 125-132].
Collapse
Affiliation(s)
- Dongki Yang
- Departments of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Jaehong Kim
- Departments of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea
| |
Collapse
|
9
|
Coronas V, Terrié E, Déliot N, Arnault P, Constantin B. Calcium Channels in Adult Brain Neural Stem Cells and in Glioblastoma Stem Cells. Front Cell Neurosci 2020; 14:600018. [PMID: 33281564 PMCID: PMC7691577 DOI: 10.3389/fncel.2020.600018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
The brain of adult mammals, including humans, contains neural stem cells (NSCs) located within specific niches of which the ventricular-subventricular zone (V-SVZ) is the largest one. Under physiological conditions, NSCs proliferate, self-renew and produce new neurons and glial cells. Several recent studies established that oncogenic mutations in adult NSCs of the V-SVZ are responsible for the emergence of malignant primary brain tumors called glioblastoma. These aggressive tumors contain a small subpopulation of cells, the glioblastoma stem cells (GSCs), that are endowed with proliferative and self-renewal abilities like NSCs from which they may arise. GSCs are thus considered as the cells that initiate and sustain tumor growth and, because of their resistance to current treatments, provoke tumor relapse. A growing body of studies supports that Ca2+ signaling controls a variety of processes in NSCs and GSCs. Ca2+ is a ubiquitous second messenger whose fluctuations of its intracellular concentrations are handled by channels, pumps, exchangers, and Ca2+ binding proteins. The concerted action of the Ca2+ toolkit components encodes specific Ca2+ signals with defined spatio-temporal characteristics that determine the cellular responses. In this review, after a general overview of the adult brain NSCs and GSCs, we focus on the multiple roles of the Ca2+ toolkit in NSCs and discuss how GSCs hijack these mechanisms to promote tumor growth. Extensive knowledge of the role of the Ca2+ toolkit in the management of essential functions in healthy and pathological stem cells of the adult brain should help to identify promising targets for clinical applications.
Collapse
Affiliation(s)
- Valérie Coronas
- Laboratoire STIM, Université de Poitiers-CNRS ERL 7003, Poitiers, France
| | - Elodie Terrié
- Laboratoire STIM, Université de Poitiers-CNRS ERL 7003, Poitiers, France
| | - Nadine Déliot
- Laboratoire STIM, Université de Poitiers-CNRS ERL 7003, Poitiers, France
| | - Patricia Arnault
- Laboratoire STIM, Université de Poitiers-CNRS ERL 7003, Poitiers, France
| | - Bruno Constantin
- Laboratoire STIM, Université de Poitiers-CNRS ERL 7003, Poitiers, France
| |
Collapse
|
10
|
Canonical Transient Receptor Potential (TRPC) Channels in Nociception and Pathological Pain. Neural Plast 2020; 2020:3764193. [PMID: 32273889 PMCID: PMC7115173 DOI: 10.1155/2020/3764193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/26/2020] [Accepted: 03/07/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic pathological pain is one of the most intractable clinical problems faced by clinicians and can be devastating for patients. Despite much progress we have made in understanding chronic pain in the last decades, its underlying mechanisms remain elusive. It is assumed that abnormal increase of calcium levels in the cells is a key determinant in the transition from acute to chronic pain. Exploring molecular players mediating Ca2+ entry into cells and molecular mechanisms underlying activity-dependent changes in Ca2+ signaling in the somatosensory pain pathway is therefore helpful towards understanding the development of chronic, pathological pain. Canonical transient receptor potential (TRPC) channels form a subfamily of nonselective cation channels, which permit the permeability of Ca2+ and Na+ into the cells. Initiation of Ca2+ entry pathways by these channels triggers the development of many physiological and pathological functions. In this review, we will focus on the functional implication of TRPC channels in nociception with the elucidation of their role in the detection of external stimuli and nociceptive hypersensitivity.
Collapse
|
11
|
Asghar MY, Törnquist K. Transient Receptor Potential Canonical (TRPC) Channels as Modulators of Migration and Invasion. Int J Mol Sci 2020; 21:E1739. [PMID: 32138386 PMCID: PMC7084769 DOI: 10.3390/ijms21051739] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Calcium (Ca2+) is perhaps the most versatile signaling molecule in cells. Ca2+ regulates a large number of key events in cells, ranging from gene transcription, motility, and contraction, to energy production and channel gating. To accomplish all these different functions, a multitude of channels, pumps, and transporters are necessary. A group of channels participating in these processes is the transient receptor potential (TRP) family of cation channels. These channels are divided into 29 subfamilies, and are differentially expressed in man, rodents, worms, and flies. One of these subfamilies is the transient receptor potential canonical (TRPC) family of channels. This ion channel family comprises of seven isoforms, labeled TRPC1-7. In man, six functional forms are expressed (TRPC1, TRPC3-7), whereas TRPC2 is a pseudogene; thus, not functionally expressed. In this review, we will describe the importance of the TRPC channels and their interacting molecular partners in the etiology of cancer, particularly in regard to regulating migration and invasion.
Collapse
Affiliation(s)
- Muhammad Yasir Asghar
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland;
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6A, 20520 Turku, Finland
| | - Kid Törnquist
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland;
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6A, 20520 Turku, Finland
| |
Collapse
|
12
|
Lopez JR, Uryash A, Faury G, Estève E, Adams JA. Contribution of TRPC Channels to Intracellular Ca 2 + Dyshomeostasis in Smooth Muscle From mdx Mice. Front Physiol 2020; 11:126. [PMID: 32153426 PMCID: PMC7044154 DOI: 10.3389/fphys.2020.00126] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/04/2020] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an irreversible muscle disease characterized by a progressive loss of muscle function, decreased ambulation, and ultimately death as a result of cardiac or respiratory failure. DMD is caused by the lack of dystrophin, a protein that is important for membrane stability and signaling in excitable cells. Although vascular smooth muscle cells (VSMCs) dysfunction occurs in many pathological conditions, little is known about vascular smooth muscle function in DMD. We have previously shown that striated muscle cells, as well as neurons isolated from dystrophic (mdx) mice have higher intracellular Ca2+ ([Ca2+]i) and Na+ ([Na+]i) concentrations and decreased cell viability in comparison with wild type (Wt). Experiments were carried out in isolated VSMCs from mdx (a murine model of DMD) and congenic C57BL/10SnJ Wt mice. We found elevated [Ca2+]i and [Na+]i in VSMCs from mdx mice compared to Wt. Exposure to 1-oleoyl-2-acetyl-sn-glycerol (OAG), a TRPC3 and TRPC6 channel activator, induced a greater elevation of [Ca2+]i and [Na+]i in mdx than Wt VSMCs. The OAG induced increases in [Ca2+]i could be abolished by either removal of extracellular Ca2+ or by SAR7334, a blocker of TRPC3 and TRPC 6 channels in both genotypes. Mdx and Wt VSMCs were susceptible to muscle cell stretch-induced elevations of [Ca2+]i and [Na+]i which was completely inhibited by GsMTx-4, a mechanosensitive ion channel inhibitor. Western blots showed a significant upregulation of TRPC1 -3, -6 proteins in mdx VSMCs compare to age-matched Wt. The lack of dystrophin in mdx VSMCs produced a profound alteration of [Ca2+]i and [Na+]i homeostasis that appears to be mediated by TRPC channels. Moreover, we have been able to demonstrate pharmacologically that the enhanced stretch-induced elevation of intracellular [Ca2+] and concomitant cell damage in mdx VSMCs also appears to be mediated through TRPC1, -3 and -6 channel activation.
Collapse
Affiliation(s)
- Jose R Lopez
- Department of Research, Mount Sinai Medical Center, Miami, FL, United States
| | - Arkady Uryash
- Department of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| | - Gilles Faury
- HP2, CHU Grenoble Alpes, Inserm, University Grenoble Alpes, Grenoble, France
| | - Eric Estève
- HP2, CHU Grenoble Alpes, Inserm, University Grenoble Alpes, Grenoble, France
| | - Jose A Adams
- Department of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| |
Collapse
|
13
|
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
|
14
|
Vanoye CG, Sakakura M, Follis RM, Trevisan AJ, Narayan M, Li J, Sanders CR, Carter BD. Peripheral myelin protein 22 modulates store-operated calcium channel activity, providing insights into Charcot-Marie-Tooth disease etiology. J Biol Chem 2019; 294:12054-12065. [PMID: 31213528 PMCID: PMC6690708 DOI: 10.1074/jbc.ra118.006248] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 06/14/2019] [Indexed: 12/24/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is a peripheral neuropathy associated with gene duplication and point mutations in the peripheral myelin protein 22 (PMP22) gene. However, the role of PMP22 in Schwann cell physiology and the mechanisms by which PMP22 mutations cause CMT are not well-understood. On the basis of homology between PMP22 and proteins associated with modulation of ion channels, we hypothesized that PMP22 alters ion channel activity. Using whole-cell electrophysiology, we show here that heterologous PMP22 expression increases the amplitude of currents similar to those ascribed to store-operated calcium (SOC) channels, particularly those involving transient receptor canonical channel 1 (TrpC1). These channels help replenish Ca2+ in the endoplasmic reticulum (ER) following stimulus-induced depletion. Currents with similar properties were recorded in WT but not pmp22-/- mouse Schwann cells. Heterologous expression of the CMT-associated PMP22_L16P variant, which fails to reach the plasma membrane and localizes to the ER, led to larger currents than WT PMP22. Similarly, Schwann cells isolated from Trembler J (TrJ; PMP22_L16P) mice had larger currents than WT littermates. Calcium imaging in live nerves and cultured Schwann cells revealed elevated intracellular Ca2+ in TrJ mice compared with WT. Moreover, we found that PMP22 co-immunoprecipitated with stromal interaction molecule 1 (STIM1), the Ca2+ sensor SOC channel subunit in the ER. These results suggest that in the ER, PMP22 interacts with STIM1 and increases Ca2+ influx through SOC channels. Excess or mutant PMP22 in the ER may elevate intracellular Ca2+ levels, which could contribute to CMT pathology.
Collapse
Affiliation(s)
- Carlos G Vanoye
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232; Center for Human Genetics, Vanderbilt University, Nashville, Tennessee 37232.
| | - Masayoshi Sakakura
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 7232; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232
| | - Rose M Follis
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 7232; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | | | - Malathi Narayan
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 7232; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | - Jun Li
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232; Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232
| | - Charles R Sanders
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 7232; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | - Bruce D Carter
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 7232; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232.
| |
Collapse
|
15
|
Ibeh CL, Yiu AJ, Kanaras YL, Paal E, Birnbaumer L, Jose PA, Bandyopadhyay BC. Evidence for a regulated Ca 2+ entry in proximal tubular cells and its implication in calcium stone formation. J Cell Sci 2019; 132:jcs.225268. [PMID: 30910829 DOI: 10.1242/jcs.225268] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 03/14/2019] [Indexed: 12/14/2022] Open
Abstract
Calcium phosphate (CaP) crystals, which begin to form in the early segments of the loop of Henle (LOH), are known to act as precursors for calcium stone formation. The proximal tubule (PT), which is just upstream of the LOH and is a major site for Ca2+ reabsorption, could be a regulator of such CaP crystal formation. However, PT Ca2+ reabsorption is mostly described as being paracellular. Here, we show the existence of a regulated transcellular Ca2+ entry pathway in luminal membrane PT cells induced by Ca2+-sensing receptor (CSR, also known as CASR)-mediated activation of transient receptor potential canonical 3 (TRPC3) channels. In support of this idea, we found that both CSR and TRPC3 are physically and functionally coupled at the luminal membrane of PT cells. More importantly, TRPC3-deficient mice presented with a deficiency in PT Ca2+ entry/transport, elevated urinary [Ca2+], microcalcifications in LOH and urine microcrystals formations. Taken together, these data suggest that a signaling complex comprising CSR and TRPC3 exists in the PT and can mediate transcellular Ca2+ transport, which could be critical in maintaining the PT luminal [Ca2+] to mitigate formation of the CaP crystals in LOH and subsequent formation of calcium stones.
Collapse
Affiliation(s)
- Cliff-Lawrence Ibeh
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA
| | - Allen J Yiu
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA.,Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University, Washington DC, DC 20037, USA
| | - Yianni L Kanaras
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA
| | - Edina Paal
- Pathology and Laboratory Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA
| | - Lutz Birnbaumer
- Division of Intramural Research, NIEHS, Research Triangle Park, Durham, NC 27709, USA.,Institute for Biomedical Research (BIOMED), Catholic University of Argentina, C1107AFF Buenos Aires, Argentina
| | - Pedro A Jose
- Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University, Washington DC, DC 20037, USA.,Department of Pharmacology and Physiology, The George Washington University, Washington DC, DC 20037, USA
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington DC, DC 20422, USA .,Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University, Washington DC, DC 20037, USA.,Department of Pharmacology and Physiology, The George Washington University, Washington DC, DC 20037, USA
| |
Collapse
|
16
|
TRPC-mediated Ca 2+ signaling and control of cellular functions. Semin Cell Dev Biol 2019; 94:28-39. [PMID: 30738858 DOI: 10.1016/j.semcdb.2019.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/30/2019] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
Abstract
Canonical members of the TRP superfamily of ion channels have long been recognized as key elements of Ca2+ handling in a plethora of cell types. The emerging role of TRPC channels in human physiopathology has generated considerable interest in their pharmacological targeting, which requires detailed understanding of their molecular function. Although consent has been reached that receptor-phospholipase C (PLC) pathways and generation of lipid mediators constitute the prominent upstream signaling process that governs channel activity, multimodal sensing features of TRPC complexes have been demonstrated repeatedly. Downstream signaling by TRPC channels is similarly complex and involves the generation of local and global cellular Ca2+ rises, which are well-defined in space and time to govern specific cellular functions. These TRPC-mediated Ca2+ signals rely in part on Ca2+ permeation through the channels, but are essentially complemented by secondary mechanisms such as Ca2+ mobilization from storage sites and Na+/Ca2+ exchange, which involve coordinated interaction with signaling partners. Consequently, the control of cell functions by TRPC molecules is critically determined by dynamic assembly and subcellular targeting of the TRPC complexes. The very recent availability of high-resolution structure information on TRPC channel complexes has paved the way towards a comprehensive understanding of signal transduction by TRPC channels. Here, we summarize current concepts of cation permeation in TRPC complexes, TRPC-mediated shaping of cellular Ca2+ signals and the associated control of specific cell functions.
Collapse
|
17
|
Picardo MCD, Sugimura YK, Dorst KE, Kallurkar PS, Akins VT, Ma X, Teruyama R, Guinamard R, Kam K, Saha MS, Del Negro CA. Trpm4 ion channels in pre-Bötzinger complex interneurons are essential for breathing motor pattern but not rhythm. PLoS Biol 2019; 17:e2006094. [PMID: 30789900 PMCID: PMC6400419 DOI: 10.1371/journal.pbio.2006094] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 03/05/2019] [Accepted: 02/05/2019] [Indexed: 12/21/2022] Open
Abstract
Inspiratory breathing movements depend on pre-Bötzinger complex (preBötC) interneurons that express calcium (Ca2+)-activated nonselective cationic current (ICAN) to generate robust neural bursts. Hypothesized to be rhythmogenic, reducing ICAN is predicted to slow down or stop breathing; its contributions to motor pattern would be reflected in the magnitude of movements (output). We tested the role(s) of ICAN using reverse genetic techniques to diminish its putative ion channels Trpm4 or Trpc3 in preBötC neurons in vivo. Adult mice transduced with Trpm4-targeted short hairpin RNA (shRNA) progressively decreased the tidal volume of breaths yet surprisingly increased breathing frequency, often followed by gasping and fatal respiratory failure. Mice transduced with Trpc3-targeted shRNA survived with no changes in breathing. Patch-clamp and field recordings from the preBötC in mouse slices also showed an increase in the frequency and a decrease in the magnitude of preBötC neural bursts in the presence of Trpm4 antagonist 9-phenanthrol, whereas the Trpc3 antagonist pyrazole-3 (pyr-3) showed inconsistent effects on magnitude and no effect on frequency. These data suggest that Trpm4 mediates ICAN, whose influence on frequency contradicts a direct role in rhythm generation. We conclude that Trpm4-mediated ICAN is indispensable for motor output but not the rhythmogenic core mechanism of the breathing central pattern generator.
Collapse
Affiliation(s)
- Maria Cristina D. Picardo
- Department of Applied Science, Integrated Science Center, William & Mary, Williamsburg, Virginia, United States of America
| | - Yae K. Sugimura
- Department of Applied Science, Integrated Science Center, William & Mary, Williamsburg, Virginia, United States of America
| | - Kaitlyn E. Dorst
- Department of Applied Science, Integrated Science Center, William & Mary, Williamsburg, Virginia, United States of America
| | - Prajkta S. Kallurkar
- Department of Applied Science, Integrated Science Center, William & Mary, Williamsburg, Virginia, United States of America
| | - Victoria T. Akins
- Department of Applied Science, Integrated Science Center, William & Mary, Williamsburg, Virginia, United States of America
| | - Xingru Ma
- Department of Applied Science, Integrated Science Center, William & Mary, Williamsburg, Virginia, United States of America
| | - Ryoichi Teruyama
- Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Romain Guinamard
- Signalisation, Electrophysiologie et Imagerie des Lésions d’Ischémie-Reperfusion Myocardique, Normandie Université, UNICAEN, Caen, France
| | - Kaiwen Kam
- Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University, Chicago, Illinois, United States of America
| | - Margaret S. Saha
- Department of Biology, Integrated Science Center, William & Mary, Williamsburg, Virginia, United States of America
| | - Christopher A. Del Negro
- Department of Applied Science, Integrated Science Center, William & Mary, Williamsburg, Virginia, United States of America
| |
Collapse
|
18
|
Secondo A, Bagetta G, Amantea D. On the Role of Store-Operated Calcium Entry in Acute and Chronic Neurodegenerative Diseases. Front Mol Neurosci 2018; 11:87. [PMID: 29623030 PMCID: PMC5874322 DOI: 10.3389/fnmol.2018.00087] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/06/2018] [Indexed: 12/22/2022] Open
Abstract
In both excitable and non-excitable cells, calcium (Ca2+) signals are maintained by a highly integrated process involving store-operated Ca2+ entry (SOCE), namely the opening of plasma membrane (PM) Ca2+ channels following the release of Ca2+ from intracellular stores. Upon depletion of Ca2+ store, the stromal interaction molecule (STIM) senses Ca2+ level reduction and migrates from endoplasmic reticulum (ER)-like sites to the PM where it activates the channel proteins Orai and/or the transient receptor potential channels (TRPC) prompting Ca2+ refilling. Accumulating evidence suggests that SOCE dysregulation may trigger perturbation of intracellular Ca2+ signaling in neurons, glia or hematopoietic cells, thus participating to the pathogenesis of diverse neurodegenerative diseases. Under acute conditions, such as ischemic stroke, neuronal SOCE can either re-establish Ca2+ homeostasis or mediate Ca2+ overload, thus providing a non-excitotoxic mechanism of ischemic neuronal death. The dualistic role of SOCE in brain ischemia is further underscored by the evidence that it also participates to endothelial restoration and to the stabilization of intravascular thrombi. In Parkinson's disease (PD) models, loss of SOCE triggers ER stress and dysfunction/degeneration of dopaminergic neurons. Disruption of neuronal SOCE also underlies Alzheimer's disease (AD) pathogenesis, since both in genetic mouse models and in human sporadic AD brain samples, reduced SOCE contributes to synaptic loss and cognitive decline. Unlike the AD setting, in the striatum from Huntington's disease (HD) transgenic mice, an increased STIM2 expression causes elevated synaptic SOCE that was suggested to underlie synaptic loss in medium spiny neurons. Thus, pharmacological inhibition of SOCE is beneficial to synapse maintenance in HD models, whereas the same approach may be anticipated to be detrimental to cortical and hippocampal pyramidal neurons. On the other hand, up-regulation of SOCE may be beneficial during AD. These intriguing findings highlight the importance of further mechanistic studies to dissect the molecular pathways, and their corresponding targets, involved in synaptic dysfunction and neuronal loss during aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Napoli, Italy
| | - Giacinto Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, Section of Preclinical and Translational Pharmacology, University of Calabria, Cosenza, Italy
| | - Diana Amantea
- Department of Pharmacy, Health and Nutritional Sciences, Section of Preclinical and Translational Pharmacology, University of Calabria, Cosenza, Italy
| |
Collapse
|
19
|
Li W, Ding Y, Smedley C, Wang Y, Chaudhari S, Birnbaumer L, Ma R. Increased glomerular filtration rate and impaired contractile function of mesangial cells in TRPC6 knockout mice. Sci Rep 2017. [PMID: 28646178 PMCID: PMC5482875 DOI: 10.1038/s41598-017-04067-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The present study was conducted to determine if TRPC6 regulates glomerular filtration rate (GFR) and the contractile function of glomerular mesangial cells (MCs). GFR was assessed in conscious TRPC6 wild type and knockout mice, and in anesthetized rats with and without in vivo knockdown of TRPC6 in kidneys. We found that GFR was significantly greater, and serum creatinine level was significantly lower in TRPC6 deficient mice. Consistently, local knockdown of TRPC6 in kidney using TRPC6 specific shRNA construct significantly attenuated Ang II-induced GFR decline in rats. Furthermore, Ang II-stimulated contraction and Ca2+ entry were significantly suppressed in primary MCs isolated from TRPC6 deficient mice, and the Ca2+ response could be rescued by re-introducing TRPC6. Moreover, inhibition of reverse mode of Na+-Ca2+ exchange by KB-R7943 significantly reduced Ca2+ entry response in TRPC6-expressing, but not in TRPC6-knocked down MCs. Ca2+ entry response was also significantly attenuated in Na+ free solution. Single knockdown of TRPC6 and TRPC1 resulted in a comparable suppression on Ca2+ entry with double knockdown of both. These results suggest that TRPC6 may regulate GFR by modulating MC contractile function through multiple Ca2+ signaling pathways.
Collapse
Affiliation(s)
- Weizu Li
- Department of Pharmacology, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Yanfeng Ding
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA
| | - Crystal Smedley
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA
| | - Yanxia Wang
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA
| | - Sarika Chaudhari
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA
| | - Lutz Birnbaumer
- Transmembrane Signaling Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, 27709, USA
| | - Rong Ma
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA.
| |
Collapse
|
20
|
Belkacemi T, Niermann A, Hofmann L, Wissenbach U, Birnbaumer L, Leidinger P, Backes C, Meese E, Keller A, Bai X, Scheller A, Kirchhoff F, Philipp SE, Weissgerber P, Flockerzi V, Beck A. TRPC1- and TRPC3-dependent Ca 2+ signaling in mouse cortical astrocytes affects injury-evoked astrogliosis in vivo. Glia 2017. [PMID: 28636132 DOI: 10.1002/glia.23180] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Following brain injury astrocytes change into a reactive state, proliferate and grow into the site of lesion, a process called astrogliosis, initiated and regulated by changes in cytoplasmic Ca2+ . Transient receptor potential canonical (TRPC) channels may contribute to Ca2+ influx but their presence and possible function in astrocytes is not known. By RT-PCR and RNA sequencing we identified transcripts of Trpc1, Trpc2, Trpc3, and Trpc4 in FACS-sorted glutamate aspartate transporter (GLAST)-positive cultured mouse cortical astrocytes and subcloned full-length Trpc1 and Trpc3 cDNAs from these cells. Ca2+ entry in cortical astrocytes depended on TRPC3 and was increased in the absence of Trpc1. After co-expression of Trpc1 and Trpc3 in HEK-293 cells both proteins co-immunoprecipitate and form functional heteromeric channels, with TRPC1 reducing TRPC3 activity. In vitro, lack of Trpc3 reduced astrocyte proliferation and migration whereas the TRPC3 gain-of-function moonwalker mutation and Trpc1 deficiency increased astrocyte migration. In vivo, astrogliosis and cortex edema following stab wound injury were reduced in Trpc3-/- but increased in Trpc1-/- mice. In summary, our results show a decisive contribution of TRPC3 to astrocyte Ca2+ signaling, which is even augmented in the absence of Trpc1, in particular following brain injury. Targeted therapies to reduce TRPC3 channel activity in astrocytes might therefore be beneficial in traumatic brain injury.
Collapse
Affiliation(s)
- Thabet Belkacemi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Homburg, 66421, Germany
| | - Alexander Niermann
- Experimentelle und Klinische Pharmakologie und Toxikologie, Homburg, 66421, Germany
| | - Laura Hofmann
- Experimentelle und Klinische Pharmakologie und Toxikologie, Homburg, 66421, Germany
| | - Ulrich Wissenbach
- Experimentelle und Klinische Pharmakologie und Toxikologie, Homburg, 66421, Germany
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, 27709, USA.,Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, C1107AFF, Argentina
| | | | - Christina Backes
- Klinische Bioinformatik, Universität des Saarlandes, Saarbrücken, 66123, Germany
| | - Eckart Meese
- Institut für Humangenetik, Homburg, 66421, Germany
| | - Andreas Keller
- Klinische Bioinformatik, Universität des Saarlandes, Saarbrücken, 66123, Germany
| | - Xianshu Bai
- Molekulare Physiologie, Universität des Saarlandes, Homburg, 66421, Germany
| | - Anja Scheller
- Molekulare Physiologie, Universität des Saarlandes, Homburg, 66421, Germany
| | - Frank Kirchhoff
- Molekulare Physiologie, Universität des Saarlandes, Homburg, 66421, Germany
| | - Stephan E Philipp
- Experimentelle und Klinische Pharmakologie und Toxikologie, Homburg, 66421, Germany
| | - Petra Weissgerber
- Experimentelle und Klinische Pharmakologie und Toxikologie, Homburg, 66421, Germany
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Homburg, 66421, Germany
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie, Homburg, 66421, Germany.,Zentrum für Human- und Molekularbiologie, Homburg, 66421, Germany
| |
Collapse
|
21
|
Transient receptor potential canonical type 3 channels: Interactions, role and relevance - A vascular focus. Pharmacol Ther 2017; 174:79-96. [DOI: 10.1016/j.pharmthera.2017.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
22
|
Ambudkar IS, de Souza LB, Ong HL. TRPC1, Orai1, and STIM1 in SOCE: Friends in tight spaces. Cell Calcium 2017; 63:33-39. [PMID: 28089266 PMCID: PMC5466534 DOI: 10.1016/j.ceca.2016.12.009] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 12/29/2016] [Accepted: 12/29/2016] [Indexed: 01/07/2023]
Abstract
Store-operated calcium entry (SOCE) is a ubiquitous Ca2+ entry pathway that is activated in response to depletion of ER-Ca2+ stores and critically controls the regulation of physiological functions in miscellaneous cell types. The transient receptor potential canonical 1 (TRPC1) is the first member of the TRPC channel subfamily to be identified as a molecular component of SOCE. While TRPC1 has been shown to contribute to SOCE and regulate various functions in many cells, none of the reported TRPC1-mediated currents resembled ICRAC, the highly Ca2+-selective store-dependent current first identified in lymphocytes and mast cells. Almost a decade after the cloning of TRPC1 two proteins were identified as the primary components of the CRAC channel. The first, STIM1, is an ER-Ca2+ sensor protein involved in activating SOCE. The second, Orai1 is the pore-forming component of the CRAC channel. Co-expression of STIM1 and Orai1 generated robust ICRAC. Importantly, STIM1 was shown to also activate TRPC1 via its C-terminal polybasic domain, which is distinct from its Orai1-activating domain, SOAR. In addition, TRPC1 function critically depends on Orai1-mediated Ca2+ entry which triggers recruitment of TRPC1 into the plasma membrane where it is then activated by STIM1. TRPC1 and Orai1 form discrete STIM1-gated channels that generate distinct Ca2+ signals and regulate specific cellular functions. Surface expression of TRPC1 can be modulated by trafficking of the channel to and from the plasma membrane, resulting in changes to the phenotype of TRPC1-mediated current and [Ca2+]i signals. Thus, TRPC1 is activated downstream of Orai1 and modifies the initial [Ca2+]i signal generated by Orai1 following store depletion. This review will summarize the important findings that underlie the current concepts for activation and regulation of TRPC1, as well as its impact on cell function.
Collapse
Affiliation(s)
- Indu S Ambudkar
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Lorena Brito de Souza
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - 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
|
Yiu AJ, Ibeh CL, Roy SK, Bandyopadhyay BC. Melamine induces Ca 2+-sensing receptor activation and elicits apoptosis in proximal tubular cells. Am J Physiol Cell Physiol 2017; 313:C27-C41. [PMID: 28381520 DOI: 10.1152/ajpcell.00225.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 01/06/2023]
Abstract
Melamine causes renal tubular cell injury through inflammation, fibrosis, and apoptosis. Although melamine affects the rise in intracellular Ca2+ concentration ([Ca2+]i), reactive oxygen species (ROS) production, and proapoptotic pathway activation, the mechanism of upstream Ca2+ signaling is unknown. Because melamine has some structural similarities with l-amino acids, which endogenously activate Ca2+-sensing receptors (CSR), we examined the effect of melamine on CSR-induced Ca2+ signaling and apoptotic cell death. We show here that melamine activates CSR, causing a sustained Ca2+ entry in the renal epithelial cell line, LLC-PK1. Moreover, such CSR stimulation resulted in a rise in [Ca2+]i, leading to enhanced ROS production. Furthermore, melamine-induced elevated [Ca2+]i and ROS production caused a dose-dependent increase in apoptotic (by DAPI staining, DNA laddering, and annexin V assay) and necrotic (propidium iodide staining) cell death. Upon examining the downstream mechanism, we found that transforming growth factor β1 (TGF-β1), which increases extracellular matrix genes and proapoptotic signaling, was also upregulated at lower doses of melamine, which could be due to an early event inducing apoptosis. Additionally, cells exposed to melamine displayed a rise in pERK activation and lactate dehydrogenase release resulting in cytotoxicity. These results offer a novel insight into the molecular mechanisms by which melamine exerts its effect on CSR, causing a sustained elevation of [Ca2+]i, leading to ROS generation, fibronectin production, proapoptotic pathway activation, and renal cell damage. Together, these results thus suggest that melamine-induced apoptosis and/or necrosis may subsequently result in acute kidney injury and promote kidney stone formation.
Collapse
Affiliation(s)
- Allen J Yiu
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, Washington, District of Columbia.,Department of Pharmacology and Physiology, School of Medicine, George Washington University, Washington, District of Columbia; and
| | - Cliff-Lawrence Ibeh
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, Washington, District of Columbia
| | - Sanjit K Roy
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, Washington, District of Columbia
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, Washington, District of Columbia; .,Department of Pharmacology and Physiology, School of Medicine, George Washington University, Washington, District of Columbia; and.,Department of Pharmacology and Physiology, Georgetown University, Washington, District of Columbia
| |
Collapse
|
24
|
Subedi KP, Ong HL, Ambudkar IS. Assembly of ER-PM Junctions: A Critical Determinant in the Regulation of SOCE and TRPC1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 981:253-276. [PMID: 29594865 DOI: 10.1007/978-3-319-55858-5_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Store-operated calcium entry (SOCE), a unique plasma membrane Ca2+ entry mechanism, is activated when ER-[Ca2+] is decreased. SOCE is mediated via the primary channel, Orai1, as well as others such as TRPC1. STIM1 and STIM2 are ER-Ca2+ sensor proteins that regulate Orai1 and TRPC1. SOCE requires assembly of STIM proteins with the plasma membrane channels which occurs within distinct regions in the cell that have been termed as endoplasmic reticulum (ER)-plasma membrane (PM) junctions. The PM and ER are in close proximity to each other within this region, which allows STIM1 in the ER to interact with and activate either Orai1 or TRPC1 in the plasma membrane. Activation and regulation of SOCE involves dynamic assembly of various components that are involved in mediating Ca2+ entry as well as those that determine the formation and stabilization of the junctions. These components include proteins in the cytosol, ER and PM, as well as lipids in the PM. Recent studies have also suggested that SOCE and its components are compartmentalized within ER-PM junctions and that this process might require remodeling of the plasma membrane lipids and reorganization of structural and scaffolding proteins. Such compartmentalization leads to the generation of spatially- and temporally-controlled Ca2+signals that are critical for regulating many downstream cellular functions.
Collapse
Affiliation(s)
- Krishna P Subedi
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, NIDCR, NIH, Bethesda, MD, USA
| | - Hwei Ling Ong
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, NIDCR, NIH, Bethesda, MD, USA
| | - Indu S Ambudkar
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, NIDCR, NIH, Bethesda, MD, USA.
| |
Collapse
|
25
|
STIM-TRP Pathways and Microdomain Organization: Ca 2+ Influx Channels: The Orai-STIM1-TRPC Complexes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:139-157. [PMID: 28900913 DOI: 10.1007/978-3-319-57732-6_8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ca2+ influx by plasma membrane Ca2+ channels is the crucial component of the receptor-evoked Ca2+ signal. The two main Ca2+ influx channels of non-excitable cells are the Orai and TRPC families of Ca2+ channels. These channels are activated in response to cell stimulation and Ca2+ release from the endoplasmic reticulum (ER). The protein that conveys the Ca2+ content of the ER to the plasma membrane is the ER Ca2+ sensor STIM1. STIM1 activates the Orai channels and is obligatory for channel opening. TRPC channels can function in two modes, as STIM1-dependent and STIM1-independent. When activated by STIM1, both channel types function at the ER/PM (plasma membrane) junctions. This chapter describes the properties and regulation of the channels by STIM1, with emphasis how and when TRPC channels function as STIM1-dependent and STIM1-independent modes and their unique Ca2+-dependent physiological functions that are not shared with the Orai channels.
Collapse
|
26
|
STIM-TRP Pathways and Microdomain Organization: Contribution of TRPC1 in Store-Operated Ca 2+ Entry: Impact on Ca 2+ Signaling and Cell Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:159-188. [PMID: 28900914 DOI: 10.1007/978-3-319-57732-6_9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Store-operated calcium entry (SOCE) is a ubiquitous Ca2+ entry pathway that is activated in response to depletion of ER-Ca2+ stores and critically controls the regulation of physiological functions in a wide variety of cell types. The transient receptor potential canonical (TRPC) channels (TRPCs 1-7), which are activated by stimuli leading to PIP2 hydrolysis, were first identified as molecular components of SOCE channels. While TRPC1 was associated with SOCE and regulation of function in several cell types, none of the TRPC members displayed I CRAC, the store-operated current identified in lymphocytes and mast cells. Intensive search finally led to the identification of Orai1 and STIM1 as the primary components of the CRAC channel. Orai1 was established as the pore-forming channel protein and STIM1 as the ER-Ca2+ sensor protein involved in activation of Orai1. STIM1 also activates TRPC1 via a distinct domain in its C-terminus. However, TRPC1 function depends on Orai1-mediated Ca2+ entry, which triggers recruitment of TRPC1 into the plasma membrane where it is activated by STIM1. TRPC1 and Orai1 form distinct store-operated Ca2+ channels that regulate specific cellular functions. It is now clearly established that regulation of TRPC1 trafficking can change plasma membrane levels of the channel, the phenotype of the store-operated Ca2+ current, as well as pattern of SOCE-mediated [Ca2+]i signals. Thus, TRPC1 is activated downstream of Orai1 and modifies the initial [Ca2+]i signal generated by Orai1. This review will highlight current concepts of the activation and regulation of TRPC1 channels and its impact on cell function.
Collapse
|
27
|
Abstract
Neurotrophins, including nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), bind to their high-affinity receptors to promote neuronal survival during brain development. One of the key downstream pathways is the phospholipase C (PLC) pathway, which not only plays a central role in calcium release from internal store but also in activation of TRPC channels coupled with neurotrophin receptors. TRPC channels are required for the neurotrophin-mediated neuronal protective effects. In addition, activation of TRPC channels is able to protect neurons in the absence of neurotrophin. In some circumstances, TRPC channels coupled with metabotropic glutamate receptor may mediate the excitotoxicity by calcium overload. One of the key questions in the field is the channel gating mechanisms; understanding of which would help design compounds to modulate the channel properties. The development and identification of TRPC channel agonists or blockers are promising and may unveil new therapeutic drugs for the treatment of neurodegenerative diseases and epilepsy.
Collapse
|
28
|
Ma R, Du J, Sours S, Ding M. Store-Operated Ca2+ Channel in Renal Microcirculation and Glomeruli. Exp Biol Med (Maywood) 2016; 231:145-53. [PMID: 16446490 DOI: 10.1177/153537020623100204] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Store-operated Ca2+ channel (SOC) is defined as a channel that opens in response to depletion of the internal Ca2+ stores. During the last decade, many investigators have made a great effort to identify and characterize SOC, and to evaluate its physiologic function and pathophysiologic relevance in a variety of cell lines, primary cultures, and native tissues. To date, accumulating evidence has demonstrated that SOC is an essential Ca2+ entry mechanism in vascular smooth-muscle cells of renal microvasculature and glomerular mesangial cells, both of which tightly control glomerular hemodynamics and filtration. Store-operated Ca2+, combined with other types of Ca2+ entry channels, constitutes a profile of Ca2+ changes in response to physiologic vasoconstrictors and, thereby, regulates renal microcirculation and mesangial function. In addition, SOC is associated with altered Ca2+ signaling occurring in diseased kidneys, such as diabetic nephropathy. Although the gating mechanism and molecular identity of SOC are still enigmatic and may be cell-type and tissue specific, data from several independent groups suggest that protein kinase C plays an important role in SOC activation and that certain isoforms of canonical transient receptor potential (TRPC) proteins are candidates of SOC in renal mlcrovessels and mesangial cells.
Collapse
Affiliation(s)
- Rong Ma
- Department of Integrative Physiology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | | | | | | |
Collapse
|
29
|
Lepannetier S, Zanou N, Yerna X, Emeriau N, Dufour I, Masquelier J, Muccioli G, Tajeddine N, Gailly P. Sphingosine-1-phosphate-activated TRPC1 channel controls chemotaxis of glioblastoma cells. Cell Calcium 2016; 60:373-383. [PMID: 27638096 DOI: 10.1016/j.ceca.2016.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/25/2016] [Accepted: 09/08/2016] [Indexed: 01/21/2023]
Abstract
TRP channels are involved in the control of a broad range of cellular functions such as cell proliferation and motility. We investigated the gating mechanism of TRPC1 channel and its role in U251 glioblastoma cells migration in response to chemotaxis by platelet-derived growth factor (PDGF). PDGF induced an influx of Ca2+ that was partially inhibited after pretreatment of the cells with SKI-II, a specific inhibitor of sphingosine kinase producing sphingosine-1-P (S1P). S1P by itself also induced an entry of Ca2+. Interestingly, PDGF- and S1P-induced entries of Ca2+ were lost in siRNA-TRPC1 treated cells. PDGF-induced chemotaxis of U251 cells was dramatically inhibited in cells treated with SKI-II. This effect was almost completely rescued by addition of synthetic S1P. Chemotaxis was also completely lost in siRNA-TRPC1 treated cells and interestingly, the rescue of migration of cells treated with SKI-II by S1P was dependent on the expression of TRPC1. Immunocytochemistry revealed that, in response to PDGF, TRPC1 translocated from inside of the cell to the front of migration (lamellipodes). This effect seemed PI3K dependent as it was inhibited by cell pre-treatment with LY294002, a PI3-kinase inhibitor. Our results thus identify S1P as a potential activator of TRPC1, a channel involved in cell orientation during chemotaxis by PDGF.
Collapse
Affiliation(s)
- Sophie Lepannetier
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Nadège Zanou
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Xavier Yerna
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Noémie Emeriau
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Inès Dufour
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Julien Masquelier
- Université catholique de Louvain, Louvain Drug Research Institute, av. Mounier 72, box B1.72.01, 1200 Brussels, Belgium
| | - Giulio Muccioli
- Université catholique de Louvain, Louvain Drug Research Institute, av. Mounier 72, box B1.72.01, 1200 Brussels, Belgium
| | - Nicolas Tajeddine
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium
| | - Philippe Gailly
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, 1200 Brussels, Belgium.
| |
Collapse
|
30
|
Ku CY, Babich L, Word RA, Zhong M, Ulloa A, Monga M, Sanborn BM. Expression of Transient Receptor Channel Proteins in Human Fundal Myometrium in Pregnancy. ACTA ACUST UNITED AC 2016; 13:217-25. [PMID: 16527499 DOI: 10.1016/j.jsgi.2005.12.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Cation channels comprised of transient receptor potential (TrpC) proteins may play a role in signal-regulated calcium entry and calcium homeostasis in myometrium. The objective of this study was to determine the relative abundance of specific TrpC mRNAs expressed in human myometrium and determine if TrpC mRNA and protein concentrations differ in fundal myometrium before and after the onset of labor. METHODS A quantitative real-time polymerase chain reaction (Q-RT-PCR) procedure was developed for determining the concentration of TrpC mRNA expression in immortalized and primary human myometrial cells and myometrial fundus tissues from patients before and after the onset of labor. The corresponding TrpC proteins were detected by Western blot analysis and immunohistochemistry. RESULTS hTrpC1, 3, 4, 5, 6, and 7 mRNAs were expressed in two lines of immortalized human myometrial cells and in primary human myocytes. In all of these cells, hTrpC1 and hTrpC4 mRNAs were the most abundant, followed by hTrpC6. A similar distribution was observed in fundal myometrium samples from patients before and after the onset of labor. hTrpC4 mRNA was significantly lower after the onset of labor; there were no significant changes in the concentrations of other TrpC mRNAs. Immunohistochemistry identified hTrpC1, 3, 4, and 6 proteins in myometrial smooth muscle cells. Western blot analysis of myometrial membranes demonstrated no statistically significant changes in hTrpC1, 3, 4, and 6 proteins between samples collected before and after the onset of labor. CONCLUSIONS We have demonstrated that hTrpC1 and hTrpC4 are the most abundant TrpC mRNAs in human myometrium, with TrpC6 being the next most abundant. There was no increase in TrpC mRNA or protein in fundal myometrium with the onset of labor. Nonetheless, these isoforms may play significant roles in signal regulated calcium entry in human myometrium.
Collapse
Affiliation(s)
- Chun-Ying Ku
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Shin DM, Son A, Park S, Kim MS, Ahuja M, Muallem S. The TRPCs, Orais and STIMs in ER/PM Junctions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:47-66. [PMID: 27161224 DOI: 10.1007/978-3-319-26974-0_3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The Ca(2+) second messenger is initiated at ER/PM junctions and propagates into the cell interior to convey the receptor information. The signal is maintained by Ca(2+) influx across the plasma membrane through the Orai and TRPC channels. These Ca(2+) influx channels form complexes at ER/PM junctions with the ER Ca(2+) sensor STIM1, which activates the channels. The function of STIM1 is modulated by other STIM isoforms like STIM1L, STIM2 and STIM2.1/STIM2β and by SARAF, which mediates the Ca(2+)-dependent inhibition of Orai channels. The ER/PM junctions are formed at membrane contact sites by tethering proteins that generate several types of ER/PM junctions, such as PI(4,5)P2-poor and PI(4,5)P2-rich domains. This chapter discusses several properties of the TRPC channels, the Orai channels and the STIMs, their key interacting proteins and how interaction of the STIMs with the channels gates their activity. The chapter closes by highlighting open questions and potential future directions in this field.
Collapse
Affiliation(s)
- Dong Min Shin
- Department of Oral Biology, BK 21 PLUS Project, Yonsei University College of Dentistry, Seoul, 120-752, South Korea.
| | - Aran Son
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, 20892, USA
| | - Seonghee Park
- Department of Physiology, School of Medicine, EwhaWomans University, 911-1 Mok-6-dong, Yang Chun-gu, Seoul, 158-710, South Korea
| | - Min Seuk Kim
- Department of Oral Physiology, School of Dentistry, Wonkwang University, Iksan City, Jeonbuk, South Korea
| | - Malini Ahuja
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, 20892, USA
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
32
|
Ong HL, de Souza LB, Ambudkar IS. Role of TRPC Channels in Store-Operated Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:87-109. [DOI: 10.1007/978-3-319-26974-0_5] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
33
|
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
|
34
|
NADPH oxidases—do they play a role in TRPC regulation under hypoxia? Pflugers Arch 2015; 468:23-41. [DOI: 10.1007/s00424-015-1731-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 12/25/2022]
|
35
|
Fu J, Gao Z, Shen B, Zhu MX. Canonical transient receptor potential 4 and its small molecule modulators. SCIENCE CHINA-LIFE SCIENCES 2014; 58:39-47. [PMID: 25480324 DOI: 10.1007/s11427-014-4772-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 09/06/2014] [Indexed: 02/04/2023]
Abstract
Canonical transient receptor potential 4 (TRPC4) forms non-selective cation channels that contribute to phospholipase C-dependent Ca(2+) entry into cells following stimulation of G protein coupled receptors and receptor tyrosine kinases. Moreover, the channels are regulated by pertussis toxin-sensitive Gi/o proteins, lipids, and various other signaling mechanisms. TRPC4-containing channels participate in the regulation of a variety of physiological functions, including excitability of both gastrointestinal smooth muscles and brain neurons. This review is to present recent advances in the understanding of physiology and development of small molecular modulators of TRPC4 channels.
Collapse
Affiliation(s)
- Jie Fu
- Department of Physiology, Anhui Medical University, Hefei, 230032, China
| | | | | | | |
Collapse
|
36
|
de Souza LB, Ambudkar IS. Trafficking mechanisms and regulation of TRPC channels. Cell Calcium 2014; 56:43-50. [PMID: 25012489 DOI: 10.1016/j.ceca.2014.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 02/06/2023]
Abstract
TRPC channels are Ca(2+)-permeable cation channels which are regulated downstream from receptor-coupled PIP2 hydrolysis. These channels contribute to a wide variety of cellular functions. Loss or gain of channel function has been associated with dysfunction and aberrant physiology. TRPC channel functions are influenced by their physical and functional interactions with numerous proteins that determine their regulation, scaffolding, trafficking, as well as their effects on the downstream cellular processes. Such interactions also compartmentalize the Ca(2+) signals arising from TRPC channels. A large number of studies demonstrate that trafficking is a critical mode by which plasma membrane localization and surface expression of TRPC channels are regulated. This review will provide an overview of intracellular trafficking pathways as well as discuss the current state of knowledge regarding the mechanisms and components involved in trafficking of the seven members of the TRPC family (TRPC1-TRPC7).
Collapse
Affiliation(s)
- Lorena Brito de Souza
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States.
| | - Indu S Ambudkar
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
37
|
Physiological Function and Characterization of TRPCs in Neurons. Cells 2014; 3:455-75. [PMID: 24852263 PMCID: PMC4092863 DOI: 10.3390/cells3020455] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 04/22/2014] [Accepted: 05/13/2014] [Indexed: 12/14/2022] Open
Abstract
Ca2+ entry is essential for regulating vital physiological functions in all neuronal cells. Although neurons are engaged in multiple modes of Ca2+ entry that regulates variety of neuronal functions, we will only discuss a subset of specialized Ca2+-permeable non-selective Transient Receptor Potential Canonical (TRPC) channels and summarize their physiological and pathological role in these excitable cells. Depletion of endoplasmic reticulum (ER) Ca2+ stores, due to G-protein coupled receptor activation, has been shown to activate TRPC channels in both excitable and non-excitable cells. While all seven members of TRPC channels are predominately expressed in neuronal cells, the ion channel properties, mode of activation, and their physiological responses are quite distinct. Moreover, many of these TRPC channels have also been suggested to be associated with neuronal development, proliferation and differentiation. In addition, TRPCs also regulate neurosecretion, long-term potentiation and synaptic plasticity. Similarly, perturbations in Ca2+ entry via the TRPC channels have been also suggested in a spectrum of neuropathological conditions. Hence, understanding the precise involvement of TRPCs in neuronal function and in neurodegenerative conditions would presumably unveil avenues for plausible therapeutic interventions for these devastating neuronal diseases.
Collapse
|
38
|
Woo JS, Lee KJ, Huang M, Cho CH, Lee EH. Heteromeric TRPC3 with TRPC1 formed via its ankyrin repeats regulates the resting cytosolic Ca2+ levels in skeletal muscle. Biochem Biophys Res Commun 2014; 446:454-9. [PMID: 24613381 DOI: 10.1016/j.bbrc.2014.02.127] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 02/26/2014] [Indexed: 01/05/2023]
Abstract
The main tasks of skeletal muscle are muscle contraction and relaxation, which are mediated by changes in cytosolic Ca(2+) levels. Canonical-type transient receptor potential 3 (TRPC3) contains an ankyrin repeat (AR) region at the N-terminus (38-188 amino acids) and forms extracellular Ca(2+)-entry channels by homo or heteromerization with other TRP subtypes in various cells including skeletal myotubes. However, previous research has not determined which region(s) of TRPC3 is responsible for the heteromerization, whether the AR region participates in the heteromerizations, or what is the role of heteromeric TRPC3s in skeletal muscle. In the present study, the heteromerization of TRPC3 with TRPC1 was first examined by GST pull-down assays of TRPC3 portions with TRPC1. The portion containing the AR region of TRPC3 was bound to the TRPC1, but the binding was inhibited by the very end sub-region of the TRPC3 (1-37 amino acids). In-silico studies have suggested that the very end sub-region possibly induces a structural change in the AR region. Second, the very end sub-region of TRPC3 was expressed in mouse primary skeletal myotubes, resulting in a dominant-negative inhibition of heteromeric TRPC3/1 formation. In addition, the skeletal myotubes expressing the very end sub-region showed a decrease in resting cytosolic Ca(2+) levels. These results suggest that the AR region of TRPC3 could mediate the heteromeric TRPC3/1 formation, and the heteromeric TRPC3/1 could participate in regulating the resting cytosolic Ca(2+) levels in skeletal muscle.
Collapse
Affiliation(s)
- Jin Seok Woo
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Keon Jin Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Mei Huang
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Chung-Hyun Cho
- Department of Pharmacology and Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Eun Hui Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea.
| |
Collapse
|
39
|
Lee KP, Choi S, Hong JH, Ahuja M, Graham S, Ma R, So I, Shin DM, Muallem S, Yuan JP. Molecular determinants mediating gating of Transient Receptor Potential Canonical (TRPC) channels by stromal interaction molecule 1 (STIM1). J Biol Chem 2014; 289:6372-6382. [PMID: 24464579 DOI: 10.1074/jbc.m113.546556] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transient receptor potential canonical (TRPC) channels mediate a critical part of the receptor-evoked Ca(2+) influx. TRPCs are gated open by the endoplasmic reticulum Ca(2+) sensor STIM1. Here we asked which stromal interaction molecule 1 (STIM1) and TRPC domains mediate the interaction between them and how this interaction is used to open the channels. We report that the STIM1 Orai1-activating region domain of STIM1 interacts with the TRPC channel coiled coil domains (CCDs) and that this interaction is essential for opening the channels by STIM1. Thus, disruption of the N-terminal (NT) CCDs by triple mutations eliminated TRPC surface localization and reduced binding of STIM1 to TRPC1 and TRPC5 while increasing binding to TRPC3 and TRPC6. Single mutations in TRPC1 NT or C-terminal (CT) CCDs reduced interaction and activation of TRPC1 by STIM1. Remarkably, single mutations in the TRPC3 NT CCD enhanced interaction and regulation by STIM1. Disruption in the TRPC3 CT CCD eliminated regulation by STIM1 and the enhanced interaction caused by NT CCD mutations. The NT CCD mutations converted TRPC3 from a TRPC1-dependent to a TRPC1-independent, STIM1-regulated channel. TRPC1 reduced the FRET between BFP-TRPC3 and TRPC3-YFP and between CFP-TRPC3-YFP upon stimulation. Accordingly, knockdown of TRPC1 made TRPC3 STIM1-independent. STIM1 dependence of TRPC3 was reconstituted by the TRPC1 CT CCD alone. Knockout of Trpc1 and Trpc3 similarly inhibited Ca(2+) influx, and inhibition of Trpc3 had no further effect on Ca(2+) influx in Trpc1(-/-) cells. Cell stimulation enhanced the formation of Trpc1-Stim1-Trpc3 complexes. These findings support a model in which the TRPC3 NT and CT CCDs interact to shield the CT CCD from interaction with STIM1. The TRPC1 CT CCD dissociates this interaction to allow the STIM1 Orai1-activating region within STIM1 access to the TRPC3 CT CCD and regulation of TRPC3 by STIM1. These studies provide evidence that the TRPC channel CCDs participate in channel gating.
Collapse
Affiliation(s)
- Kyu Pil Lee
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Seok Choi
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892; Department of Physiology, College of Medicine, Chosun University, Gwangju 501-759, Republic of Korea
| | - Jeong Hee Hong
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Malini Ahuja
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Sarabeth Graham
- Department of Integrative Physiology, Cardiovascular Research Institute, University of North Texas Health Sciences Center, Fort Worth, Texas 76107
| | - Rong Ma
- Department of Integrative Physiology, Cardiovascular Research Institute, University of North Texas Health Sciences Center, Fort Worth, Texas 76107
| | - Insuk So
- Department of Physiology and Biophysics, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Dong Min Shin
- Department of Oral Biology, BK21 PLUS Project, Yonsei University, Seoul 120-752, Republic of Korea
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892.
| | - Joseph P Yuan
- Department of Integrative Physiology, Cardiovascular Research Institute, University of North Texas Health Sciences Center, Fort Worth, Texas 76107
| |
Collapse
|
40
|
Abstract
The TRPC1 ion channel was the first mammalian TRP channel to be cloned. In humans, it is encoded by the TRPC1 gene located in chromosome 3. The protein is predicted to consist of six transmembrane segments with the N- and C-termini located in the cytoplasm. The extracellular loop connecting transmembrane segments 5 and 6 participates in the formation of the ionic pore region. Inside the cell, TRPC1 is present in the endoplasmic reticulum, plasma membrane, intracellular vesicles, and primary cilium, an antenna-like sensory organelle functioning as a signaling platform. In human and rodent tissues, it shows an almost ubiquitous expression. TRPC1 interacts with a diverse group of proteins including ion channel subunits, receptors, and cytosolic proteins to mediate its effect on Ca(2+) signaling. It primarily functions as a cation nonselective channel within pathways controlling Ca(2+) entry in response to cell surface receptor activation. Through these pathways, it affects basic cell functions, such as proliferation and survival, differentiation, secretion, and cell migration, as well as cell type-specific functions such as chemotropic turning of neuronal growth cones and myoblast fusion. The biological role of TRPC1 has been studied in genetically engineered mice where the Trpc1 gene has been experimentally ablated. Although these mice live to adulthood, they show defects in several organs and tissues, such as the cardiovascular, central nervous, skeletal and muscular, and immune systems. Genetic and functional studies have implicated TRPC1 in diabetic nephropathy, Parkinson's disease, Huntington's disease, Duchenne muscular dystrophy, cancer, seizures, and Darier-White skin disease.
Collapse
Affiliation(s)
- Vasyl Nesin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | | |
Collapse
|
41
|
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
|
42
|
Choi S, Maleth J, Jha A, Lee KP, Kim MS, So I, Ahuja M, Muallem S. The TRPCs-STIM1-Orai interaction. Handb Exp Pharmacol 2014; 223:1035-54. [PMID: 24961979 DOI: 10.1007/978-3-319-05161-1_13] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ca(2+) signaling entails receptor-stimulated Ca(2+) release from the ER stores that serves as a signal to activate Ca(2+) influx channels present at the plasma membrane, the store-operated Ca(2+) channels (SOCs). The two known SOCs are the Orai and TRPC channels. The SOC-dependent Ca(2+) influx mediates and sustains virtually all Ca(2+)-dependent regulatory functions. The signal that transmits the Ca(2+) content of the ER stores to the plasma membrane is the ER resident, Ca(2+)-binding protein STIM1. STIM1 is a multidomain protein that clusters and dimerizes in response to Ca(2+) store depletion leading to activation of Orai and TRPC channels. Activation of the Orais by STIM1 is obligatory for their function as SOCs, while TRPC channels can function as both STIM1-dependent and STIM1-independent channels. Here we discuss the different mechanisms by which STIM1 activates the Orai and TRPC channels, the emerging specific and non-overlapping physiological functions of Ca(2+) influx mediated by the two channel types, and argue that the TRPC channels should be the preferred therapeutic target to control the toxic effect of excess Ca(2+) influx.
Collapse
Affiliation(s)
- Seok Choi
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
TRPC3 represents one of the first identified mammalian relative of the Drosophila trp gene product. Despite extensive biochemical and biophysical characterization as well as ambitious attempts to uncover its physiological role in native cell systems, the channel protein still represents a rather enigmatic member of the TRP superfamily. TRPC3 is significantly expressed in the brain and heart and appears of (patho)physiological importance in both non-excitable and excitable cells, being potentially involved in a wide spectrum of Ca(2+) signaling mechanisms. TRPC3 cation channels display unique gating and regulatory properties that allow for recognition and integration of multiple input stimuli including lipid mediators, cellular Ca(2+) gradients, as well as redox signals. Physiological/pathophysiological functions of this highly versatile cation channel protein are as yet incompletely understood. Its ability to associate in a dynamic manner with a variety of partner proteins enables TRPC3 to serve coordination of multiple downstream signaling pathways and control of divergent cellular functions. Here, we summarize current knowledge on ion channel features as well as possible signaling functions of TRPC3 and discuss the potential biological relevance of this signaling molecule.
Collapse
Affiliation(s)
- Michaela Lichtenegger
- Institute of Pharmaceutical Sciences - Pharmacology and Toxicology, University of Graz, A-8010, Graz, Austria
| | | |
Collapse
|
44
|
Makino A, Firth AL, Yuan JXJ. Endothelial and smooth muscle cell ion channels in pulmonary vasoconstriction and vascular remodeling. Compr Physiol 2013; 1:1555-602. [PMID: 23733654 DOI: 10.1002/cphy.c100023] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The pulmonary circulation is a low resistance and low pressure system. Sustained pulmonary vasoconstriction and excessive vascular remodeling often occur under pathophysiological conditions such as in patients with pulmonary hypertension. Pulmonary vasoconstriction is a consequence of smooth muscle contraction. Many factors released from the endothelium contribute to regulating pulmonary vascular tone, while the extracellular matrix in the adventitia is the major determinant of vascular wall compliance. Pulmonary vascular remodeling is characterized by adventitial and medial hypertrophy due to fibroblast and smooth muscle cell proliferation, neointimal proliferation, intimal, and plexiform lesions that obliterate the lumen, muscularization of precapillary arterioles, and in situ thrombosis. A rise in cytosolic free Ca(2+) concentration ([Ca(2+)]cyt) in pulmonary artery smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction, while increased release of mitogenic factors, upregulation (or downregulation) of ion channels and transporters, and abnormalities in intracellular signaling cascades are key to the remodeling of the pulmonary vasculature. Changes in the expression, function, and regulation of ion channels in PASMC and pulmonary arterial endothelial cells play an important role in the regulation of vascular tone and development of vascular remodeling. This article will focus on describing the ion channels and transporters that are involved in the regulation of pulmonary vascular function and structure and illustrating the potential pathogenic role of ion channels and transporters in the development of pulmonary vascular disease.
Collapse
Affiliation(s)
- Ayako Makino
- Department of Medicine, The University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
45
|
Abstract
Transient receptor potential (TRP) channels are cellular sensors for a wide spectrum of physical and chemical stimuli. They are involved in the formation of sight, hearing, touch, smell, taste, temperature, and pain sensation. TRP channels also play fundamental roles in cell signaling and allow the host cell to respond to benign or harmful environmental changes. As TRP channel activation is controlled by very diverse processes and, in many cases, exhibits complex polymodal properties, understanding how each TRP channel responds to its unique forms of activation energy is both crucial and challenging. The past two decades witnessed significant advances in understanding the molecular mechanisms that underlie TRP channels activation. This review focuses on our current understanding of the molecular determinants for TRP channel activation.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, USA.
| |
Collapse
|
46
|
Katz B, Oberacker T, Richter D, Tzadok H, Peters M, Minke B, Huber A. Drosophila TRP and TRPL are assembled as homomultimeric channels in vivo. J Cell Sci 2013; 126:3121-33. [PMID: 23687378 DOI: 10.1242/jcs.123505] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Family members of the cationic transient receptor potential (TRP) channels serve as sensors and transducers of environmental stimuli. The ability of different TRP channel isoforms of specific subfamilies to form heteromultimers and the structural requirements for channel assembly are still unresolved. Although heteromultimerization of different mammalian TRP channels within single subfamilies has been described, even within a subfamily (such as TRPC) not all members co-assemble with each other. In Drosophila photoreceptors two TRPC channels, TRP and TRP-like protein (TRPL) are expressed together in photoreceptors where they generate the light-induced current. The formation of functional TRP-TRPL heteromultimers in cell culture and in vitro has been reported. However, functional in vivo assays have shown that each channel functions independently of the other. Therefore, the issue of whether TRP and TRPL form heteromultimers in vivo is still unclear. In the present study we investigated the ability of TRP and TRPL to form heteromultimers, and the structural requirements for channel assembly, by studying assembly of GFP-tagged TRP and TRPL channels and chimeric TRP and TRPL channels, in vivo. Interaction studies of tagged and native channels as well as native and chimeric TRP-TRPL channels using co-immunoprecipitation, immunocytochemistry and electrophysiology, critically tested the ability of TRP and TRPL to interact. We found that TRP and TRPL assemble exclusively as homomultimeric channels in their native environment. The above analyses revealed that the transmembrane regions of TRP and TRPL do not determine assemble specificity of these channels. However, the C-terminal regions of both TRP and TRPL predominantly specify the assembly of homomeric TRP and TRPL channels.
Collapse
Affiliation(s)
- Ben Katz
- Department of Medical Neurobiology, Faculty of Medicine and the Edmond and Lily Safra Center for Brain Sciences, Hebrew University, Jerusalem 91120, Israel
| | | | | | | | | | | | | |
Collapse
|
47
|
Piron M, Villereal ML. Chronic exposure to stress hormones alters the subtype of store-operated channels expressed in H19-7 hippocampal neuronal cells. J Cell Physiol 2013; 228:1332-43. [DOI: 10.1002/jcp.24289] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 11/12/2012] [Indexed: 11/07/2022]
|
48
|
Contribution and regulation of TRPC channels in store-operated Ca2+ entry. CURRENT TOPICS IN MEMBRANES 2013; 71:149-79. [PMID: 23890115 DOI: 10.1016/b978-0-12-407870-3.00007-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Store-operated calcium entry (SOCE) is activated in response to depletion of the endoplasmic reticulum-Ca(2+) stores following stimulation of plasma membrane receptors that couple to PIP2 hydrolysis and IP3 generation. Search for the molecular components of SOCE channels led to the identification of mammalian transient receptor potential canonical (TRPC) family of calcium-permeable channels (TRPC1-TRPC7), which are all activated in response to stimuli that result in PIP2 hydrolysis. While several TRPCs, including TRPC1, TRPC3, and TRPC4, have been implicated in SOCE, the data are most consistent for TRPC1. Extensive studies in cell lines and knockout mouse models have established the contribution of TRPC1 to SOCE. Furthermore, there is a critical functional interaction between TRPC1 and the key components of SOCE, STIM1, and Orai1, which determines the activation of TRPC1. Orai1-mediated Ca(2+) entry is required for recruitment of TRPC1 and its insertion into surface membranes while STIM1 gates the channel. Notably, TRPC1 and Orai1 generate distinct patterns of Ca(2+) signals in cells that are decoded for the regulation of specific cellular functions. Thus, SOCE appears to be a complex process that depends on temporal and spatial coordination of several distinct steps mediated by proteins in different cellular compartments. Emerging data suggest that, in many cell types, the net Ca(2+) entry measured in response to store depletion is the result of the coordinated regulation of different calcium-permeable ion channels. Orai1 and STIM1 are central players in this process, and by mediating recruitment or activation of other Ca(2+) channels, Orai1-CRAC function can elicit rapid changes in global and local [Ca(2+)]i signals in cells. It is most likely that the type of channels and the [Ca(2+)]i signature that are generated by this process reflect the physiological function of the cell that is regulated by Ca(2+).
Collapse
|
49
|
Kochukov MY, Balasubramanian A, Noel RC, Marrelli SP. Role of TRPC1 and TRPC3 channels in contraction and relaxation of mouse thoracic aorta. J Vasc Res 2012; 50:11-20. [PMID: 23095462 DOI: 10.1159/000342461] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 07/04/2012] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND/AIMS Canonical transient receptor potential (TRPC) channels modulate membrane potential and intracellular Ca(2+). We examined the role of TRPC1 and TRPC3 channels in vasocontraction and relaxation in mouse aorta. METHODS Vasocontraction and relaxation of aorta from wild-type (WT), TRPC1 KO and TRPC3 knockout (KO) mice were measured for phenylephrine (Phe) and carbachol (CCh). Intracellular Ca(2+) was measured in primary aorta endothelial cells (EC) and whole cell K(+) current in freshly isolated smooth muscle cells (SMC). RESULTS AND CONCLUSION TRPC1 KO aorta showed increased vasocontraction to Phe compared to WT and TRPC3 KO aorta due to diminished role of BK(Ca) channels. BK(Ca) mRNA (aorta) and whole cell current (SMC) were reduced versus WT. Contraction in WT aorta was increased to TRPC1 KO level by BK(Ca) channel inhibition. Relaxation to CCh was reduced in TRPC1 KO and TRPC3 KO aortas with concomitant reduction in EC Ca(2+) response. Pyr3 (TRPC3 blocker) reduced the Ca(2+) response to CCh in EC from WT, but not TRPC3 KO mice. In summary, TRPC1 attenuates receptor-mediated contraction through activation and/or expression of SMC BK(Ca) channels while TRPC3 does not contribute to receptor-mediated constriction. Both TRPC1 and TRPC3 participate in EC Ca(2+) influx and vasorelaxation of aorta.
Collapse
Affiliation(s)
- M Y Kochukov
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
50
|
TRPC1 protein forms only one type of native store-operated channels in HEK293 cells. Biochimie 2012; 95:347-53. [PMID: 23079337 DOI: 10.1016/j.biochi.2012.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/05/2012] [Indexed: 11/23/2022]
Abstract
TRPC1 is a major component of store-operated calcium entry in many cell types. In our previous studies, three types of endogenous store-operated calcium channels have been described in HEK293 cells, but it remained unknown which of these channels are composed of TRPC1 proteins. Here, this issue has been addressed by performing single-channel analysis in HEK293 cells transfected with anti-TRPC1 siRNA (siTPRC1) or a TPRC1-encoding plasmid. The results show that thapsigargin-or agonist-induced calcium influx is significantly attenuated in siTRPC1-transfected HEK293 cells. TRPC1 knockdown by siRNA results in the disappearance of store-operated I(max) channels, while the properties of I(min) and I(NS) channels are unaffected. In HEK293 cells with overexpressed TRPC1 protein, the unitary current-voltage relationship of exogenous TRPC1 channels is almost linear, with a slope conductance of about 17 pS. The extrapolated reversal potential of expressed TRPC1 channels is +30 mV. Therefore, the main electrophysiological and regulatory properties of expressed TRPC1 and native I(max) channels are identical. Moreover, TRPC1 overexpression in HEK293 cells results in an increased number of store-operated I(max) channels. All these data allow us to conclude that TRPC1 protein forms native store-operated I(max) channels but is not an essential subunit for other store-operated channel types in HEK293 cells.
Collapse
|