1
|
Tessier N, Ducrozet M, Dia M, Badawi S, Chouabe C, Crola Da Silva C, Ovize M, Bidaux G, Van Coppenolle F, Ducreux S. TRPV1 Channels Are New Players in the Reticulum-Mitochondria Ca 2+ Coupling in a Rat Cardiomyoblast Cell Line. Cells 2023; 12:2322. [PMID: 37759544 PMCID: PMC10529771 DOI: 10.3390/cells12182322] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
The Ca2+ release in microdomains formed by intercompartmental contacts, such as mitochondria-associated endoplasmic reticulum membranes (MAMs), encodes a signal that contributes to Ca2+ homeostasis and cell fate control. However, the composition and function of MAMs remain to be fully defined. Here, we focused on the transient receptor potential vanilloid 1 (TRPV1), a Ca2+-permeable ion channel and a polymodal nociceptor. We found TRPV1 channels in the reticular membrane, including some at MAMs, in a rat cardiomyoblast cell line (SV40-transformed H9c2) by Western blotting, immunostaining, cell fractionation, and proximity ligation assay. We used chemical and genetic probes to perform Ca2+ imaging in four cellular compartments: the endoplasmic reticulum (ER), cytoplasm, mitochondrial matrix, and mitochondrial surface. Our results showed that the ER Ca2+ released through TRPV1 channels is detected at the mitochondrial outer membrane and transferred to the mitochondria. Finally, we observed that prolonged TRPV1 modulation for 30 min alters the intracellular Ca2+ equilibrium and influences the MAM structure or the hypoxia/reoxygenation-induced cell death. Thus, our study provides the first evidence that TRPV1 channels contribute to MAM Ca2+ exchanges.
Collapse
Affiliation(s)
- Nolwenn Tessier
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| | - Mallory Ducrozet
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| | - Maya Dia
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| | - Sally Badawi
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| | - Christophe Chouabe
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| | - Claire Crola Da Silva
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| | - Michel Ovize
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Hôpital Louis Pradel, Services d’Explorations Fonctionnelles Cardiovasculaires et CIC de Lyon, 69394 Lyon, France
| | - Gabriel Bidaux
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| | - Fabien Van Coppenolle
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| | - Sylvie Ducreux
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (N.T.); (M.D.); (M.D.); (S.B.); (C.C.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
| |
Collapse
|
2
|
Öz A, Çinar R, Naziroğlu M. TRPV1 stimulation increased oxidative neurotoxicity and apoptosis in the glia cell membrane but not in the perinuclear area: An evidence of TRPV1 subtype. Metab Brain Dis 2022; 37:2291-2304. [PMID: 35776390 DOI: 10.1007/s11011-022-01037-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/13/2022] [Indexed: 12/01/2022]
Abstract
Glia are essential neurons of the immune system in the central nervous system. The effective mission of glia depends on their activation, release of cytokines, and oxidative cleaning of debris material from neuronal cells. Accumulating evidence indicates that microglia activation-induced oxidative stress via the activation Ca2+ permeable TRPV1 channel has an essential role in the pathophysiology of neurodegenerative diseases. However, there is scarce information on the cytosolic localization of TRPV1 and the induction of oxidative cytotoxicity in the glia. Hence, we investigated the interactions between cytosolic TRPV1 expression levels and oxidative neurotoxicity in the BV2, C8-D1A, N9 glia, and DBTRG glioblastoma cells. We observed TRPV1 expression in the perinuclear area but not in the cell membrane in the BV2, C8-D1A, and N9 cells. Hence, we observed no activation of TRPV1 on the increase of mitochondrial free reactive oxygen species (mROS) and apoptosis in the cells after the capsaicin stimulation. However, we observed TRPV1 channel expression in the positive control (DBTRG) cell membranes. Hence, the Ca2+ influx, TRPV1 current density, apoptosis, and mROS levels were increased in the DBTRG cells after the capsaicin stimulation, although their levels were diminished by the treatment of the TRPV1 blocker (capsazepine). In conclusion, the presence of TRPV1 in the cell membrane of DBTRG cells induced excessive generation of mROS and apoptosis actions, although the presence of TRPV1 in the perinuclear area did not cause the actions. It seems that there is a subtype of TRPV1 in the perinuclear area, and it is not activated by the capsaicin.
Collapse
Affiliation(s)
- Ahmi Öz
- Department of Biophysics, School of Medicine, Suleyman Demirel University, TR-32260, Isparta, Turkey
| | - Ramazan Çinar
- Department of Neuroscience, Institute of Health Sciences, Suleyman Demirel University, TR-32260, Isparta, Turkey
| | - Mustafa Naziroğlu
- Department of Biophysics, School of Medicine, Suleyman Demirel University, TR-32260, Isparta, Turkey.
- Department of Neuroscience, Institute of Health Sciences, Suleyman Demirel University, TR-32260, Isparta, Turkey.
- Neuroscience Research Center (NOROBAM), Suleyman Demirel University, TR-32260, Isparta, Turkey.
- Drug Discovery Unit, BSN Health, Analyses, Innovation, Consultancy, Organization, Agriculture and Industry Ltd, TR-32260, Isparta, Turkey.
| |
Collapse
|
3
|
Xiao W, Chen Y. TRPV1 in male reproductive system: focus on sperm function. Mol Cell Biochem 2022; 477:2567-2579. [PMID: 35595954 DOI: 10.1007/s11010-022-04469-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 05/04/2022] [Indexed: 12/21/2022]
Abstract
The transient receptor potential vanilloid 1 (TRPV1) is a receptor used to perceive external noxious stimuli and participates in the regulation of various pathophysiological mechanisms in vivo by integrating multiple signals. The explosive growth in knowledge of TRPV1 stemmed from research on neuronal pain and heat sensation over the last decades and is being expanded tremendously in peripheral tissue research. The discovery that TRPV1 is functionally active in male animal and human reproductive tissues have attracted increasing attention in recent years. Indeed, many studies have indicated that TRPV1 is an endocannabinoid receptor that mediates Anandamide's regulation of sperm function. Other characteristics of the TRPV1 channel itself, such as calcium penetration and temperature sensitivity, have also been investigated, especially the possibility that TRPV1 could act as a mediator for sperm thermotaxis. In addition, some reproductive diseases appear to be related to the protective effects of TRPV1 on oxidative stress and heat stress. A better understanding of TRPV1 in these areas should provide strategies for tackling male infertility. This paper is the first to review the expression and mechanism of TRPV1 in the male reproductive system from molecular and cellular perspectives. A focus is given on sperm function, including calcium homeostasis, crosstalk with endocannabinoid system, participation in cholesterol-related sperm maturation, and thermotaxis, hoping to capture the current situation of this rapidly developing field.
Collapse
Affiliation(s)
- Wanglong Xiao
- Institute of Life Science and School of Life Science, Nanchang University, No. 999 Xuefu Avenue, Honggutan District, Nanchang, 330031, Jiangxi, People's Republic of China
| | - Ying Chen
- Institute of Life Science and School of Life Science, Nanchang University, No. 999 Xuefu Avenue, Honggutan District, Nanchang, 330031, Jiangxi, People's Republic of China.
- Key Laboratory of Reproductive Physiology and Pathology in Jiangxi Province, Nanchang, Jiangxi, People's Republic of China.
| |
Collapse
|
4
|
Dissecting the Mechanism of Action of Spiperone-A Candidate for Drug Repurposing for Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14030776. [PMID: 35159043 PMCID: PMC8834219 DOI: 10.3390/cancers14030776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Despite advances in primary and adjuvant treatments, approximately 50% of colorectal cancer (CRC) patients still die from recurrence and metastatic disease. Thus, alternative and more effective therapeutic approaches are expected to be developed. Drug repurposing is increasing interest in cancer therapy, as it represents a cheaper and faster alternative strategy to de novo drug synthesis. Psychiatric medications are promising as a new generation of antitumor drugs. Here, we demonstrate that spiperone—a licensed drug for the treatment of schizophrenia—induces apoptosis in CRC cells. Our data reveal that spiperone’s cytotoxicity in CRC cells is mediated by phospholipase C activation, intracellular calcium homeostasis dysregulation, and irreversible endoplasmic reticulum stress induction, resulting in lipid metabolism alteration and Golgi apparatus damage. By identifying new targetable pathways in CRC cells, our findings represent a promising starting point for the design of novel therapeutic strategies for CRC. Abstract Approximately 50% of colorectal cancer (CRC) patients still die from recurrence and metastatic disease, highlighting the need for novel therapeutic strategies. Drug repurposing is attracting increasing attention because, compared to traditional de novo drug discovery processes, it may reduce drug development periods and costs. Epidemiological and preclinical evidence support the antitumor activity of antipsychotic drugs. Herein, we dissect the mechanism of action of the typical antipsychotic spiperone in CRC. Spiperone can reduce the clonogenic potential of stem-like CRC cells (CRC-SCs) and induce cell cycle arrest and apoptosis, in both differentiated and CRC-SCs, at clinically relevant concentrations whose toxicity is negligible for non-neoplastic cells. Analysis of intracellular Ca2+ kinetics upon spiperone treatment revealed a massive phospholipase C (PLC)-dependent endoplasmic reticulum (ER) Ca2+ release, resulting in ER Ca2+ homeostasis disruption. RNA sequencing revealed unfolded protein response (UPR) activation, ER stress, and induction of apoptosis, along with IRE1-dependent decay of mRNA (RIDD) activation. Lipidomic analysis showed a significant alteration of lipid profile and, in particular, of sphingolipids. Damage to the Golgi apparatus was also observed. Our data suggest that spiperone can represent an effective drug in the treatment of CRC, and that ER stress induction, along with lipid metabolism alteration, represents effective druggable pathways in CRC.
Collapse
|
5
|
Dhandapani P, Dondapati SK, Zemella A, Bräuer D, Wüstenhagen DA, Mergler S, Kubick S. Targeted esterase-induced dye (TED) loading supports direct calcium imaging in eukaryotic cell-free systems. RSC Adv 2021; 11:16285-16296. [PMID: 35479141 PMCID: PMC9030739 DOI: 10.1039/d0ra08397f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/25/2021] [Indexed: 11/21/2022] Open
Abstract
Calcium imaging is an important functional tool for analysing ion channels, transporters and pumps for drug screening in living cells. Depicted eukaryotic cell-free systems utilize microsomes, derived from the endoplasmic reticulum to incorporate the synthesized membrane proteins-like ion channels. Carboxylesterase is required to cleave the acetoxymethyl ester moiety of the chemical calcium indicators in order to ensure its immobility across the endoplasmic reticulum membrane. Absence or an inadequate amount of carboxylesterase in the endoplasmic reticulum of different eukaryotic cells poses a hindrance to perform calcium imaging in microsomes. In this work, we try to overcome this drawback and adapt the cell-based calcium imaging principle to a cell-free protein synthesis platform. Carboxylesterase synthesized in a Spodoptera frugiperda Sf21 lysate translation system is established as a viable calcium imaging tool in microsomes. Cell-free synthesized carboxylesterase inside microsomes is validated with esterase and dye loading assays. Native proteins from the endoplasmic reticulum, such as ryanodine channels and calcium ATPase, are analysed. Cell-free synthesized transient receptor potential channels are used as model proteins to demonstrate the realization of this concept. Carboxylesterase, the key enzyme to handle ester-based dyes, is synthesized in microsomes using eukaryotic cell-free protein synthesis platform and established as a viable calcium imaging tool to analyze native and cell-free synthesized ion channels.![]()
Collapse
Affiliation(s)
- Priyavathi Dhandapani
- Fraunhofer Institute of Cell Therapy and Immunology, Branch of Bioanalytics and Bioprocesses (IZI-BB) Am Muehlenberg 13 Potsdam-Golm Germany
| | - Srujan Kumar Dondapati
- Fraunhofer Institute of Cell Therapy and Immunology, Branch of Bioanalytics and Bioprocesses (IZI-BB) Am Muehlenberg 13 Potsdam-Golm Germany
| | - Anne Zemella
- Fraunhofer Institute of Cell Therapy and Immunology, Branch of Bioanalytics and Bioprocesses (IZI-BB) Am Muehlenberg 13 Potsdam-Golm Germany
| | - Dennis Bräuer
- Fraunhofer Institute of Cell Therapy and Immunology, Branch of Bioanalytics and Bioprocesses (IZI-BB) Am Muehlenberg 13 Potsdam-Golm Germany
| | - Doreen Anja Wüstenhagen
- Fraunhofer Institute of Cell Therapy and Immunology, Branch of Bioanalytics and Bioprocesses (IZI-BB) Am Muehlenberg 13 Potsdam-Golm Germany
| | - Stefan Mergler
- Department of Ophthalmology, Charité - Universitätsmedizin Berlin Campus Virchow-Hospital Berlin Germany
| | - Stefan Kubick
- Fraunhofer Institute of Cell Therapy and Immunology, Branch of Bioanalytics and Bioprocesses (IZI-BB) Am Muehlenberg 13 Potsdam-Golm Germany .,Faculty of Health Sciences, Joint Faculty of Brandenburg University of Technology, Cottbus - Senftenberg, Theodor Fontane Medical School of Brandenburg, University of Potsdam Germany
| |
Collapse
|
6
|
Lemos FO, Bultynck G, Parys JB. A comprehensive overview of the complex world of the endo- and sarcoplasmic reticulum Ca 2+-leak channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119020. [PMID: 33798602 DOI: 10.1016/j.bbamcr.2021.119020] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 12/11/2022]
Abstract
Inside cells, the endoplasmic reticulum (ER) forms the largest Ca2+ store. Ca2+ is actively pumped by the SERCA pumps in the ER, where intraluminal Ca2+-binding proteins enable the accumulation of large amount of Ca2+. IP3 receptors and the ryanodine receptors mediate the release of Ca2+ in a controlled way, thereby evoking complex spatio-temporal signals in the cell. The steady state Ca2+ concentration in the ER of about 500 μM results from the balance between SERCA-mediated Ca2+ uptake and the passive leakage of Ca2+. The passive Ca2+ leak from the ER is often ignored, but can play an important physiological role, depending on the cellular context. Moreover, excessive Ca2+ leakage significantly lowers the amount of Ca2+ stored in the ER compared to normal conditions, thereby limiting the possibility to evoke Ca2+ signals and/or causing ER stress, leading to pathological consequences. The so-called Ca2+-leak channels responsible for Ca2+ leakage from the ER are however still not well understood, despite over 20 different proteins have been proposed to contribute to it. This review has the aim to critically evaluate the available evidence about the various channels potentially involved and to draw conclusions about their relative importance.
Collapse
Affiliation(s)
- Fernanda O Lemos
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
7
|
Yazici AT, Gianti E, Kasimova MA, Lee BH, Carnevale V, Rohacs T. Dual regulation of TRPV1 channels by phosphatidylinositol via functionally distinct binding sites. J Biol Chem 2021; 296:100573. [PMID: 33766560 PMCID: PMC8095115 DOI: 10.1016/j.jbc.2021.100573] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 11/11/2022] Open
Abstract
Regulation of the heat- and capsaicin-activated transient receptor potential vanilloid 1 (TRPV1) channel by phosphoinositides is complex and controversial. In the most recent TRPV1 cryo-EM structure, endogenous phosphatidylinositol (PtdIns) was detected in the vanilloid binding site, and phosphoinositides were proposed to act as competitive vanilloid antagonists. This model is difficult to reconcile with phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] being a well-established positive regulator of TRPV1. Here we show that in the presence of PtdIns(4,5)P2 in excised patches, PtdIns, but not PtdIns(4)P, partially inhibited TRPV1 activity at low, but not at high capsaicin concentrations. This is consistent with PtdIns acting as a competitive vanilloid antagonist. However, in the absence of PtdIns(4,5)P2, PtdIns partially stimulated TRPV1 activity. We computationally identified residues, which are in contact with PtdIns, but not with capsaicin in the vanilloid binding site. The I703A mutant of TRPV1 showed increased sensitivity to capsaicin, as expected when removing the effect of an endogenous competitive antagonist. I703A was not inhibited by PtdIns in the presence of PtdIns(4,5)P2, but it was still activated by PtdIns in the absence of PtdIns(4,5)P2 indicating that inhibition, but not activation by PtdIns proceeds via the vanilloid binding site. In molecular dynamics simulations, PtdIns was more stable than PtdIns(4,5)P2 in this inhibitory site, whereas PtdIns(4,5)P2 was more stable than PtdIns in a previously identified, nonoverlapping, putative activating binding site. Our data indicate that phosphoinositides regulate channel activity via functionally distinct binding sites, which may explain some of the complexities of the effects of these lipids on TRPV1.
Collapse
Affiliation(s)
- Aysenur Torun Yazici
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, New Jersey, USA
| | - Eleonora Gianti
- Institute for Computational Molecular Science, Temple University, Philadelphia, Pennsylvania, USA
| | - Marina A Kasimova
- Institute for Computational Molecular Science, Temple University, Philadelphia, Pennsylvania, USA
| | - Bo-Hyun Lee
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, New Jersey, USA
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, Temple University, Philadelphia, Pennsylvania, USA.
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, New Jersey, USA.
| |
Collapse
|
8
|
Juárez-Contreras R, Méndez-Reséndiz KA, Rosenbaum T, González-Ramírez R, Morales-Lázaro SL. TRPV1 Channel: A Noxious Signal Transducer That Affects Mitochondrial Function. Int J Mol Sci 2020; 21:ijms21238882. [PMID: 33255148 PMCID: PMC7734572 DOI: 10.3390/ijms21238882] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/24/2020] [Accepted: 10/31/2020] [Indexed: 12/19/2022] Open
Abstract
The Transient Receptor Vanilloid 1 (TRPV1) or capsaicin receptor is a nonselective cation channel, which is abundantly expressed in nociceptors. This channel is an important transducer of several noxious stimuli, having a pivotal role in pain development. Several TRPV1 studies have focused on understanding its structure and function, as well as on the identification of compounds that regulate its activity. The intracellular roles of these channels have also been explored, highlighting TRPV1′s actions in the homeostasis of Ca2+ in organelles such as the mitochondria. These studies have evidenced how the activation of TRPV1 affects mitochondrial functions and how this organelle can regulate TRPV1-mediated nociception. The close relationship between this channel and mitochondria has been determined in neuronal and non-neuronal cells, demonstrating that TRPV1 activation strongly impacts on cell physiology. This review focuses on describing experimental evidence showing that TRPV1 influences mitochondrial function.
Collapse
Affiliation(s)
- Rebeca Juárez-Contreras
- Department of Cognitive Neuroscience, Neurosciences Division, Institute of Cellular Physiology, National Autonomous University of Mexico, UNAM, Mexico City 04510, Mexico; (R.J.-C.); (K.A.M.-R.); (T.R.)
| | - Karina Angélica Méndez-Reséndiz
- Department of Cognitive Neuroscience, Neurosciences Division, Institute of Cellular Physiology, National Autonomous University of Mexico, UNAM, Mexico City 04510, Mexico; (R.J.-C.); (K.A.M.-R.); (T.R.)
| | - Tamara Rosenbaum
- Department of Cognitive Neuroscience, Neurosciences Division, Institute of Cellular Physiology, National Autonomous University of Mexico, UNAM, Mexico City 04510, Mexico; (R.J.-C.); (K.A.M.-R.); (T.R.)
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, “Dr. Manuel Gea González” General Hospital, Mexico City 14080, Mexico;
| | - Sara Luz Morales-Lázaro
- Department of Cognitive Neuroscience, Neurosciences Division, Institute of Cellular Physiology, National Autonomous University of Mexico, UNAM, Mexico City 04510, Mexico; (R.J.-C.); (K.A.M.-R.); (T.R.)
- Correspondence:
| |
Collapse
|
9
|
Role of the TRPV Channels in the Endoplasmic Reticulum Calcium Homeostasis. Cells 2020; 9:cells9020317. [PMID: 32013022 PMCID: PMC7072170 DOI: 10.3390/cells9020317] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/08/2020] [Accepted: 01/21/2020] [Indexed: 01/01/2023] Open
Abstract
It has been widely established that transient receptor potential vanilloid (TRPV) channels play a crucial role in calcium homeostasis in mammalian cells. Modulation of TRPV channels activity can modify their physiological function leading to some diseases and disorders like neurodegeneration, pain, cancer, skin disorders, etc. It should be noted that, despite TRPV channels importance, our knowledge of the TRPV channels functions in cells is mostly limited to their plasma membrane location. However, some TRPV channels were shown to be expressed in the endoplasmic reticulum where their modulation by activators and/or inhibitors was demonstrated to be crucial for intracellular signaling. In this review, we have intended to summarize the poorly studied roles and functions of these channels in the endoplasmic reticulum.
Collapse
|
10
|
Roles for the Endoplasmic Reticulum in Regulation of Neuronal Calcium Homeostasis. Cells 2019; 8:cells8101232. [PMID: 31658749 PMCID: PMC6829861 DOI: 10.3390/cells8101232] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
By influencing Ca2+ homeostasis in spatially and architecturally distinct neuronal compartments, the endoplasmic reticulum (ER) illustrates the notion that form and function are intimately related. The contribution of ER to neuronal Ca2+ homeostasis is attributed to the organelle being the largest reservoir of intracellular Ca2+ and having a high density of Ca2+ channels and transporters. As such, ER Ca2+ has incontrovertible roles in the regulation of axodendritic growth and morphology, synaptic vesicle release, and neural activity dependent gene expression, synaptic plasticity, and mitochondrial bioenergetics. Not surprisingly, many neurological diseases arise from ER Ca2+ dyshomeostasis, either directly due to alterations in ER resident proteins, or indirectly via processes that are coupled to the regulators of ER Ca2+ dynamics. In this review, we describe the mechanisms involved in the establishment of ER Ca2+ homeostasis in neurons. We elaborate upon how changes in the spatiotemporal dynamics of Ca2+ exchange between the ER and other organelles sculpt neuronal function and provide examples that demonstrate the involvement of ER Ca2+ dyshomeostasis in a range of neurological and neurodegenerative diseases.
Collapse
|
11
|
Hsu CC, Chien KH, Yarmishyn AA, Buddhakosai W, Wu WJ, Lin TC, Chiou SH, Chen JT, Peng CH, Hwang DK, Chen SJ, Chang YL. Modulation of osmotic stress-induced TRPV1 expression rescues human iPSC-derived retinal ganglion cells through PKA. Stem Cell Res Ther 2019; 10:284. [PMID: 31547874 PMCID: PMC6755708 DOI: 10.1186/s13287-019-1363-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/25/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
Background Transient receptor potential vanilloid 1 (TRPV1), recognized as a hyperosmolarity sensor, is a crucial ion channel involved in the pathogenesis of neural and glial signaling. Recently, TRPV1 was determined to play a role in retinal physiology and visual transmission. In this study, we sought to clarify the role of TRPV1 and the downstream pathway in the osmotic stress-related retina ganglion cell (RGC) damage. Methods First, we modified the RGC differentiation protocol to obtain a homogeneous RGC population from human induced pluripotent stem cells (hiPSCs). Subsequently, we induced high osmotic pressure in the hiPSC-derived RGCs by administering NaCl solution and observed the behavior of the TRPV1 channel and its downstream cascade. Results We obtained a purified RGC population from the heterogeneous retina cell population using our modified method. Our findings revealed that TRPV1 was activated after 24 h of NaCl treatment. Upregulation of TRPV1 was noted with autophagy and apoptosis induction. Downstream protein expression analysis indicated increased phosphorylation of CREB and downregulated brain-derived neurotrophic factor (BDNF). However, hyperosmolarity-mediated defective morphological change and apoptosis of RGCs, CREB phosphorylation, and BDNF downregulation were abrogated after concomitant treatment with the PKA inhibitor H89. Conclusion Collectively, our study results indicated that the TRPV1–PKA pathway contributed to cellular response under high levels of osmolarity stress; furthermore, the PKA inhibitor had a protective effect on RGCs exposed to this stress. Therefore, our findings may assist in the treatment of eye diseases involving RGC damage.
Collapse
Affiliation(s)
- Chih-Chien Hsu
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Ke-Hung Chien
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Ophthalmology, Tri-Service General Hospital and National Defense Medical Center, Taipei, 114, Taiwan
| | - Aliaksandr A Yarmishyn
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Waradee Buddhakosai
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Wen-Ju Wu
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Tai-Chi Lin
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Shih-Hwa Chiou
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Jiann-Torng Chen
- Department of Ophthalmology, Tri-Service General Hospital and National Defense Medical Center, Taipei, 114, Taiwan
| | - Chi-Hsien Peng
- Department of Ophthalmology, Shin Kong Wu Ho-Su Memorial Hospital and Fu-Jen Catholic University, Taipei, Taiwan
| | - De-Kuang Hwang
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Shih-Jen Chen
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan. .,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.
| | - Yuh-Lih Chang
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan. .,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan. .,Department of Pharmacy, Taipei Veterans General Hospital; Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
12
|
Gouin O, L'Herondelle K, Buscaglia P, Le Gall-Ianotto C, Philippe R, Legoux N, Mignen O, Buhé V, Leschiera R, Sakka M, Kerfant N, Carré JL, Le Garrec R, Lefeuvre L, Lebonvallet N, Misery L. Major Role for TRPV1 and InsP3R in PAR2-Elicited Inflammatory Mediator Production in Differentiated Human Keratinocytes. J Invest Dermatol 2018; 138:1564-1572. [DOI: 10.1016/j.jid.2018.01.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 01/24/2018] [Accepted: 01/28/2018] [Indexed: 11/15/2022]
|
13
|
Zhao R, Tsang SY. Versatile Roles of Intracellularly Located TRPV1 Channel. J Cell Physiol 2017; 232:1957-1965. [DOI: 10.1002/jcp.25704] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/22/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Rui Zhao
- School of Life Sciences; The Chinese University of Hong Kong; Hong Kong SAR China
| | - Suk Ying Tsang
- School of Life Sciences; The Chinese University of Hong Kong; Hong Kong SAR China
- State Key Laboratory of Agrobiotechnology; The Chinese University of Hong Kong; Hong Kong SAR China
- Key Laboratory for Regenerative Medicine, Ministry of Education; The Chinese University of Hong Kong; Hong Kong SAR China
- Centre for Novel Biomaterials; The Chinese University of Hong Kong; Hong Kong SAR China
| |
Collapse
|
14
|
Yamamoto M, Nishiyama M, Iizuka S, Suzuki S, Suzuki N, Aiso S, Nakahara J. Transient receptor potential vanilloid 1-immunoreactive signals in murine enteric glial cells. World J Gastroenterol 2016; 22:9752-9764. [PMID: 27956799 PMCID: PMC5124980 DOI: 10.3748/wjg.v22.i44.9752] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/19/2016] [Accepted: 10/10/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the possible involvement of transient receptor potential vanilloid 1 (TRPV1) in maturation of enteric glial cells (EGCs).
METHODS Immunohistochemical and immunocytochemical techniques were used to analyze EGC markers in myenteric plexus (MP) as well as cultured MP cells and EGCs using TRPV1 knockout (KO) mice.
RESULTS We detected TRPV1-immunoreactive signals in EGC in the MP of wild-type (WT) but not KO mice. Expression of glial fibrillary acidic protein (GFAP) immunoreactive signals was lower at postnatal day (PD) 6 in KO mice, though the difference was not clear at PD 13 and PD 21. When MP cells were isolated and cultured from isolated longitudinal muscle-MP preparation from WT and KO mice, the yield of KO EGC was lower than that of WT EGC, while the yield of KO and WT smooth muscle cells showed no difference. Addition of BCTC, a TRPV1 antagonist, to enriched EGC culture resulted in a decrease in the protein ratio of GFAP to S100B, another EGC/astrocyte-specific marker.
CONCLUSION These results address the possibility that TRPV1 may be involved in the maturation of EGC, though further studies are necessary to validate this possibility.
Collapse
|
15
|
Sukumaran P, Schaar A, Sun Y, Singh BB. Functional role of TRP channels in modulating ER stress and Autophagy. Cell Calcium 2016; 60:123-32. [PMID: 26995055 DOI: 10.1016/j.ceca.2016.02.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 02/07/2023]
Abstract
Intracellular calcium (Ca(2+)) levels play a vital role in regulating cellular fate. The coordination and interrelation among the cellular organelles, mainly the intracellular Ca(2+) stores in endoplasmic reticulum (ER), are crucial in maintaining cytosolic Ca(2+) levels and in general cellular homeostasis. Moreover, maintaining Ca(2+) homeostasis is essential for regulating diverse and sometimes opposing processes such as cell survival and cell death in disease conditions such as, neurodegeneration, cancer and aging. Ca(2+) is able to regulate opposing functions by either regulating the cellular "self-eating" phenomenon of autophagy to promote cell survival or by regulating the programmed cell death process of apoptosis. Autophagy is also important for cell survival especially after induction of ER stress and association between ER stress and autophagy may have relevance to numerous diseases. Moreover, a multitude of evidence is emerging that the functional regulation of TRP channels, their unique localization, and their interaction with other Ca(2+)-sensing elements define these diverse regulatory pathways. It is this unique function which allows individual TRP channels to contribute differently in the regulation of cell fate and, in turn, determines the precise effect of modulating Ca(2+) signaling via the particular channel. Thus, in this review we have focused on the aspects of TRP channel localization and function (Ca(2+) signaling) that affects the ER stress and autophagic process.
Collapse
Affiliation(s)
- Pramod Sukumaran
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201, United States
| | - Anne Schaar
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201, United States
| | - Yuyang Sun
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201, United States
| | - Brij B Singh
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201, United States.
| |
Collapse
|
16
|
Wollank Y, Ramer R, Ivanov I, Salamon A, Peters K, Hinz B. Inhibition of FAAH confers increased stem cell migration via PPARα. J Lipid Res 2015; 56:1947-60. [PMID: 26263913 DOI: 10.1194/jlr.m061473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 02/07/2023] Open
Abstract
Regenerative activity in tissues of mesenchymal origin depends on the migratory potential of mesenchymal stem cells (MSCs). The present study focused on inhibitors of the enzyme fatty acid amide hydrolase (FAAH), which catalyzes the degradation of endocannabinoids (anandamide, 2-arachidonoylglycerol) and endocannabinoid-like substances (N-oleoylethanolamine, N-palmitoylethanolamine). Boyden chamber assays, the FAAH inhibitors, URB597 and arachidonoyl serotonin (AA-5HT), were found to increase the migration of human adipose-derived MSCs. LC-MS analyses revealed increased levels of all four aforementioned FAAH substrates in MSCs incubated with either FAAH inhibitor. Following addition to MSCs, all FAAH substrates mimicked the promigratory action of FAAH inhibitors. Promigratory effects of FAAH inhibitors and substrates were causally linked to activation of p42/44 MAPKs, as well as to cytosol-to-nucleus translocation of the transcription factor, PPARα. Whereas PPARα activation by FAAH inhibitors and substrates became reversed upon inhibition of p42/44 MAPK activation, a blockade of PPARα left p42/44 MAPK phosphorylation unaltered. Collectively, these data demonstrate FAAH inhibitors and substrates to cause p42/44 MAPK phosphorylation, which subsequently activates PPARα to confer increased migration of MSCs. This novel pathway may be involved in regenerative effects of endocannabinoids whose degradation could be a target of pharmacological intervention by FAAH inhibitors.
Collapse
Affiliation(s)
- Yvonne Wollank
- Institute of Toxicology and Pharmacology, Rostock University Medical Center, D-18057 Rostock, Germany Department of Cell Biology, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Robert Ramer
- Institute of Toxicology and Pharmacology, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Igor Ivanov
- Institute of Toxicology and Pharmacology, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Achim Salamon
- Department of Cell Biology, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Kirsten Peters
- Department of Cell Biology, Rostock University Medical Center, D-18057 Rostock, Germany
| | - Burkhard Hinz
- Institute of Toxicology and Pharmacology, Rostock University Medical Center, D-18057 Rostock, Germany
| |
Collapse
|
17
|
Alptekin M, Eroglu S, Tutar E, Sencan S, Geyik MA, Ulasli M, Demiryurek AT, Camci C. Gene expressions of TRP channels in glioblastoma multiforme and relation with survival. Tumour Biol 2015; 36:9209-13. [PMID: 26088448 DOI: 10.1007/s13277-015-3577-x] [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] [Received: 03/25/2015] [Accepted: 05/15/2015] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal forms of cancer in humans, with a median survival of 10 to 12 months. Glioblastoma is highly malignant since the cells are supported by a great number of blood vessels. Although new treatments have been developed by increasing knowledge of molecular nature of the disease, surgical operation remains the standard of care. The TRP (transient receptor potential) superfamily consists of cation-selective channels that have roles in sensory physiology such as thermo- and osmosensation and in several complex diseases such as cancer, cardiovascular, and neuronal diseases. The aim of this study was to investigate the expression levels of TRP channel genes in patients with glioblastoma multiforme and to evaluate the relationship between TRP gene expressions and survival of the patients. Thirty-three patients diagnosed with glioblastoma were enrolled to the study. The expression levels of 21 TRP genes were quantified by using qRT-PCR with dynamic array 48 × 48 chip (BioMark HD System, Fluidigm, South San Francisco, CA, USA). TRPC1, TRPC6, TRPM2, TRPM3, TRPM7, TRPM8, TRPV1, and TRPV2 were found significantly higher in glioblastoma patients. Moreover, there was a significant relationship between the overexpression of TRP genes and the survival of the patients. These results demonstrate for the first time that TRP channels contribute to the progression and survival of the glioblastoma patients.
Collapse
Affiliation(s)
- M Alptekin
- Department of Neurosurgery, Faculty of Medicine, University of Gaziantep, Gaziantep, 27310, Turkey
| | - S Eroglu
- Department of Medical Biology, Faculty of Medicine, University of Gaziantep, Gaziantep, 27310, Turkey
| | - E Tutar
- Department of Pathology, Faculty of Medicine, University of Gaziantep, Gaziantep, 27310, Turkey
| | - S Sencan
- Department of Medical Biology, Faculty of Medicine, University of Gaziantep, Gaziantep, 27310, Turkey
| | - M A Geyik
- Department of Neurosurgery, Faculty of Medicine, University of Gaziantep, Gaziantep, 27310, Turkey
| | - M Ulasli
- Department of Medical Biology, Faculty of Medicine, University of Gaziantep, Gaziantep, 27310, Turkey
| | - A T Demiryurek
- Department of Pharmacology, Faculty of Medicine, University of Gaziantep, Gaziantep, 27310, Turkey
| | - C Camci
- Department of Medical Oncology, Faculty of Medicine, University of Gaziantep, Gaziantep, 27310, Turkey.
| |
Collapse
|
18
|
Endoplasmic reticulum stress in insulin resistance and diabetes. Cell Calcium 2014; 56:311-22. [PMID: 25239386 DOI: 10.1016/j.ceca.2014.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023]
Abstract
The endoplasmic reticulum is the main intracellular Ca(2+) store for Ca(2+) release during cell signaling. There are different strategies to avoid ER Ca(2+) depletion. Release channels utilize first Ca(2+)-bound to proteins and this minimizes the reduction of the free luminal [Ca(2+)]. However, if release channels stay open after exhaustion of Ca(2+)-bound to proteins, then the reduction of the free luminal ER [Ca(2+)] (via STIM proteins) activates Ca(2+) entry at the plasma membrane to restore the ER Ca(2+) load, which will work provided that SERCA pump is active. Nevertheless, there are several noxious conditions that result in decreased activity of the SERCA pump such as oxidative stress, inflammatory cytokines, and saturated fatty acids, among others. These conditions result in a deficient restoration of the ER [Ca(2+)] and lead to the ER stress response that should facilitate recovery of the ER. However, if the stressful condition persists then ER stress ends up triggering cell death and the ensuing degenerative process leads to diverse pathologies; particularly insulin resistance, diabetes and several of the complications associated with diabetes. This scenario suggests that limiting ER stress should decrease the incidence of diabetes and the mobility and mortality associated with this illness.
Collapse
|
19
|
Meng J, Dolly JO, Wang J. Selective cleavage of SNAREs in sensory neurons unveils protein complexes mediating peptide exocytosis triggered by different stimuli. Mol Neurobiol 2014; 50:574-88. [PMID: 24604356 DOI: 10.1007/s12035-014-8665-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 02/20/2014] [Indexed: 11/28/2022]
Abstract
Oligomerisation of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes is required for synaptic vesicle fusion and neurotransmitter release. How these regulate the release of pain peptides elicited by different stimuli from sensory neurons has not been established. Herein, K(+) depolarization was found to induce multiple sodium dodecyl sulfate (SDS)-resistant SNARE complexes in sensory neurons exposed to botulinum neurotoxins (BoNTs), with molecular weights ranging from 104-288 k (large) to 38-104 k (small). Isoform 1 of vesicle-associated membrane protein 1 (VAMP 1) assembled into stable complexes upon depolarisation and was required for the participation of intact synaptosome-associated protein of relative molecular mass 25 k (SNAP-25) or BoNT/A-truncated form (SNAP-25A) in the large functional and small inactive SDS-resistant SNARE complexes. Cleaving VAMP 1 decreased SNAP-25A in the functional complexes to a much greater extent than the remaining intact SNAP-25. Syntaxin 1 proved essential for the incorporation of intact and SNAP-25A into the large complexes. Truncation of syntaxin 1 by BoNT/C1 caused /A- and/or /C1-truncated SNAP-25 to appear in non-functional complexes and blocked the release of calcitonin gene-related peptide (CGRP) elicited by capsaicin, ionomycin, thapsigargin or K(+) depolarization. Only the latter two were susceptible to /A. Inhibition of CGRP release by BoNT/A was reversed by capsaicin and/or ionomycin, an effect overcome by BoNT/C1. Unlike BoNT/B, BoNT/D cleaved VAMP 1 in addition to 2 and 3 in rat sensory neurons and blocked both CGRP and substance P release. Thus, unlike SNAP-25, syntaxin 1 and VAMP 1 are more suitable targets to abolish functional SNARE complexes and pain peptide release evoked by any stimuli.
Collapse
Affiliation(s)
- Jianghui Meng
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | | | | |
Collapse
|
20
|
Chien CS, Ma KH, Lee HS, Liu PS, Li YH, Huang YS, Chueh SH. Dual effect of capsaicin on cell death in human osteosarcoma G292 cells. Eur J Pharmacol 2013; 718:350-60. [PMID: 24012930 DOI: 10.1016/j.ejphar.2013.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 07/23/2013] [Accepted: 08/24/2013] [Indexed: 12/11/2022]
Abstract
Thirty percent of osteosarcoma patients die within 5 years. New agents that induce apoptosis of osteosarcoma cells might be therapeutically useful. Here, we characterized the apoptotic mechanism induced by capsaicin in G292 osteosarcoma cells. Our results show that capsaicin induces an increase in the cytosolic Ca(2+) concentration which is independent of the extracellular Ca(2+) concentration and depletes intracellular Ca(2+) stores, suggesting the presence of endoplasmic reticulum transient receptor potential vanilloid receptor type 1. Capsaicin also activates the mitochondrial caspase 3-dependent death cascade. Rapamycin, an inhibitor of mammalian target of rapamycin, evokes autophagy, as do capsaicin or thapsigargin, a sarco(endo)plasmic reticulum Ca(2+) ATPase inhibitor that causes Ca(2+) store depletion. Capsaicin-induced cell death is completely inhibited by co-treatment with the pan-caspase inhibitor Z-VAD-fmk and increased by the autophagy inhibitor 3-methyladenine, suggesting the existence of an autophagy-dependent anti-apoptotic mechanism. Capsaicin also induces ERK phosphorylation, which acts as a downstream effector of autophagy. 3-Methyladenine or PD98059, an ERK kinase inhibitor, restores capsaicin-induced cell death in the presence of Z-VAD-fmk, suggesting that inhibition of autophagy activates a second cell death pathway that is caspase-independent. Taken together, our data show that capsaicin causes Ca(2+) depletion of intracellular Ca(2+) stores and simultaneously activates the mitochondrial caspase-dependent death cascade and autophagy-dependent ERK activation and that the latter counteracts a second death signaling pathway that is caspase-independent.
Collapse
Affiliation(s)
- Chi-Sheng Chien
- Department of Orthopaedics, Chi Mei Medical Center, Tainan, Taiwan, ROC.
| | | | | | | | | | | | | |
Collapse
|
21
|
Buckinx R, Van Nassauw L, Avula LR, Alpaerts K, Adriaensen D, Timmermans JP. Transient receptor potential vanilloid type 1 channel (TRPV1) immunolocalization in the murine enteric nervous system is affected by the targeted C-terminal epitope of the applied antibody. J Histochem Cytochem 2013; 61:421-32. [PMID: 23482327 DOI: 10.1369/0022155413484764] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The expression of transient receptor potential vanilloid type 1 channel (TRPV1) in the enteric nervous system is still the subject of debate. Although a number of studies have reported that TRPV1 is limited to extrinsic afferent fibers, other studies argue for an intrinsic expression of TRPV1. In the present study, reverse transcriptase PCR was employed to establish the expression of TRPV1 mRNA throughout the gastrointestinal tract. Using two antibodies directed against different epitopes of TRPV1, we were able to show at the protein level that the observed distribution pattern of TRPV1 is dependent on the antibody used in the immunohistochemical staining. A first antibody indeed mainly stained neuronal fibers, whereas a second antibody exclusively stained perikarya of enteric neurons throughout the mouse gastrointestinal tract. We argue that these different distribution patterns are due to the antibodies discriminating between different modulated forms of TRPV1 that influence the recognition of the targeted immunogen and as such distinguish intracellular from plasmalemmal forms of TRPV1. Our study is the first to directly compare these two antibodies within the same species and in identical conditions. Our observations underline that detailed knowledge of the epitope that is recognized by the antibodies employed in immunohistochemical procedures is a prerequisite for correctly interpreting experimental results.
Collapse
Affiliation(s)
- Roeland Buckinx
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | |
Collapse
|
22
|
Masumoto K, Tsukimoto M, Kojima S. Role of TRPM2 and TRPV1 cation channels in cellular responses to radiation-induced DNA damage. Biochim Biophys Acta Gen Subj 2013; 1830:3382-90. [PMID: 23458684 DOI: 10.1016/j.bbagen.2013.02.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/12/2013] [Accepted: 02/20/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND Radiation exposure causes DNA damage, and DNA repair systems are essential to rescue damaged cells. Although DNA damage or oxidative stress activates transient receptor potential melastatin 2 (TRPM2) and vanilloid 1 (TRPV1) cation channels, it has not been established whether these TRP channels are involved in cellular responses to radiation-induced DNA damage. Here, we investigated the contribution of TRPM2 and TRPV1 channels to γ-irradiation- and UVB-induced DNA damage responses in human lung cancer A549 cells. METHODS A549 cells were irradiated with γ-rays (2.0Gy) or UVB (5-10mJ/cm(2)). γH2AX foci, ATM activation, 53BP1 accumulation and EGFR expression were evaluated by immunofluorescence staining. Extracellular ATP concentration was measured by luciferin-luciferase assay. Knockdown of TRPM2 and TRPV1 expression was done by siRNA transfection. RESULTS γ-Irradiation-induced γH2AX focus formation, ATM activation, 53BP1 accumulation and EGFR nuclear translocation, which are all associated with DNA repair, were suppressed by knockdown of TRPM2 and TRPV1 channels in A549 cells. Release of ATP, which mediates DNA damage response-associated activation of P2Y receptors, was suppressed by pre-treatment with catalase or knockdown of TRPM2 channel, but not TRPV1 channel. Similarly, UVB-induced γH2AX focus formation was suppressed in TRPM2- and TRPV1-knockdown cells, while UVB-induced ATP release was blocked in TRPM2- but not TRPV1-knockdown cells. CONCLUSION Our results suggest that the activation of TRPM2 channel, which mediates ATP release, and TRPV1 channel plays significant roles in the cellular responses to DNA damage induced by γ-irradiation and UVB irradiation. GENERAL SIGNIFICANCE Our results provide a new insight into the function of TRP channels from the viewpoint of radiation biology.
Collapse
Affiliation(s)
- Kanako Masumoto
- Department of Radiation Biosciences, Tokyo University of Science, Noda-shi, Chiba, Japan
| | | | | |
Collapse
|
23
|
Szolcsányi J, Pintér E. Transient receptor potential vanilloid 1 as a therapeutic target in analgesia. Expert Opin Ther Targets 2013; 17:641-57. [PMID: 23421411 DOI: 10.1517/14728222.2013.772580] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The selective excitatory action of capsaicin followed by long-term chemoanalgesia due to an action on the 'capsaicin receptor' of C-polymodal nociceptors, cloned 15 years ago, opened up fascinating perspectives for a class of nociceptor blocking analgesics. AREAS COVERED The TRPV1/capsaicin receptor is an integrative, chemoceptive, noxious heat-gated cation channel also gated by several endogenous ligands and sensitized by phosphorylation through intracellular cascades triggered from receptors of bradykinin, prostanoids, NGF and interactions with TRPA1. In this review, types of sensory receptors and unique mechanisms for blocking nociceptor action, e.g., 'pore dilation' intracellular acidosis and the long-term function-related mitochondrial swelling at the nerve terminals and sensory neurons are summarized. In humans the 8% capsaicin dermal patch is already in usage for nondiabetic neuropathic pain and two topical preparations of civamide have also been approved recently for cluster headache and osteoarthritis. Evidence for epidermal nerve terminal loss in humans after topical applications and misleading results on sensory neuron death evoked by TRPV1 agonism in animals are discussed. EXPERT OPINION The unique 'multisteric' gating of TRPV1 channel which is opened and modulated in various conformational changes to natural stimuli differs from the operation of canonical ligand-gated channels and makes it suitable to initiate development of second generation of TRPV1 antagonists without on-target side effects of hyperthermia and risk of burn injury.
Collapse
Affiliation(s)
- János Szolcsányi
- University of Pécs Medical School, Department of Pharmacology and Pharmacotherapy , H-7624 Pécs, Szigeti u. 12 , Hungary.
| | | |
Collapse
|
24
|
Guinamard R, Simard C, Del Negro C. Flufenamic acid as an ion channel modulator. Pharmacol Ther 2013; 138:272-84. [PMID: 23356979 DOI: 10.1016/j.pharmthera.2013.01.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 12/26/2012] [Indexed: 12/29/2022]
Abstract
Flufenamic acid has been known since the 1960s to have anti-inflammatory properties attributable to the reduction of prostaglandin synthesis. Thirty years later, flufenamic acid appeared to be an ion channel modulator. Thus, while its use in medicine diminished, its use in ionic channel research expanded. Flufenamic acid commonly not only affects non-selective cation channels and chloride channels, but also modulates potassium, calcium and sodium channels with effective concentrations ranging from 10(-6)M in TRPM4 channel inhibition to 10(-3)M in two-pore outwardly rectifying potassium channel activation. Because flufenamic acid effects develop and reverse rapidly, it is a convenient and widely used tool. However, given the broad spectrum of its targets, experimental results have to be interpreted cautiously. Here we provide an overview of ion channels targeted by flufenamic acid to aid in interpreting its effects at the molecular, cellular, and system levels. If it is used with good practices, flufenamic acid remains a useful tool for ion channel research. Understanding the targets of FFA may help reevaluate its physiological impacts and revive interest in its therapeutic potential.
Collapse
|
25
|
Cong X, Zhang Y, Yang NY, Li J, Ding C, Ding QW, Su YC, Mei M, Guo XH, Wu LL, Yu GY. Occludin is required for TRPV1-modulated paracellular permeability in the submandibular gland. J Cell Sci 2013; 126:1109-21. [PMID: 23345400 DOI: 10.1242/jcs.111781] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Occludin plays an important role in maintaining tight junction barrier function in many types of epithelia. We previously reported that activation of transient receptor potential vanilloid subtype 1 (TRPV1) in rabbit submandibular gland promoted salivary secretion, partly by an increase in paracellular permeability. We have now explored the role of occludin in TRPV1-modulated paracellular permeability in a rat submandibular gland cell line SMG-C6. Both TRPV1 and occludin were expressed in SMG-C6 cells, and capsaicin induced redistribution of occludin, but not claudin-3, claudin-4 or E-cadherin, from the cell membrane into the cytoplasm. Capsaicin also decreased transepithelial electrical resistance (TER) and increased the Trypan Blue and FITC-dextran flux. Capsazepine (CPZ), a TRPV1 antagonist, inhibited the capsaicin-induced occludin redistribution and TER decrease. Moreover, occludin knockdown by shRNA suppressed, whereas occludin re-expression restored, the TER response to capsaicin. Mechanistically, TRPV1 activation increased ERK1/2 and MLC2 phosphorylation. PD98059, an ERK1/2 kinase inhibitor, abolished the capsaicin-induced MLC2 phosphorylation, whereas ML-7, an MLC2 kinase inhibitor, did not affect ERK1/2 phosphorylation, suggesting that ERK1/2 is the upstream signaling molecule of MLC2. Capsaicin also induced F-actin reorganization, which was abolished by CPZ, PD98059 and ML-7, indicating that TRPV1 activation altered F-actin organization in an ERK1/2- and MLC2-dependent manner. Furthermore, either PD98059 or ML-7 could abolish the capsaicin-induced TER response and occludin redistribution, whereas knockdown of ERK1/2 further confirmed that the TRPV1-modulated paracellular permeability was ERK1/2 dependent. Taken together, these results identified a crucial role of occludin in submandibular epithelial cells, and more importantly, demonstrated that occludin was required to mediate TRPV1-modulated paracellular permeability.
Collapse
Affiliation(s)
- Xin Cong
- Center for Salivary Gland Diseases of Peking University School and Hospital of Stomatology, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mechanism of capsaicin receptor TRPV1-mediated toxicity in pain-sensing neurons focusing on the effects of Na(+)/Ca(2+) fluxes and the Ca(2+)-binding protein calretinin. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:1680-91. [PMID: 22982061 DOI: 10.1016/j.bbamcr.2012.08.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/23/2012] [Accepted: 08/27/2012] [Indexed: 11/23/2022]
Abstract
Transient receptor potential vanilloid subtype 1 (TRPV1) receptor is a pain-sensing, ligand-gated, non-selective cation channel expressed in peripheral sensory neurons. Prolonged activation of TRPV1 by capsaicin leads to cell swelling and formation of membrane blebs in rat dorsal root ganglion (DRG) neurons. Similar results were obtained in NIH3T3 fibroblast cells stably expressing TRPV1. Here, we assessed the contribution of Ca(2+) and Na(+) ions to TRPV1-mediated changes. Cell swelling was caused by a substantial influx of extracellular Na(+) via TRPV1 channels, causing concomitant transport of water. In the absence of extracellular Na(+), the membrane blebbing was completely inhibited, but Ca(2+) influx did not change under these conditions. Na(+) influx was modulated by the intracellular Ca(2+) concentration ([Ca(2+)]i). Elevation of [Ca(2+)]i by ionomycin sensitized/activated TRPV1 channels causing cell swelling in TRPV1-positive cells. In the absence of extracellular Ca(2+), capsaicin caused only little increase in [Ca(2+)]i indicating that the increase in [Ca(2+)]i observed after capsaicin application is derived essentially from extracellular Ca(2+) and not from internal Ca(2+) stores. In the absence of extracellular Ca(2+) also the process of cell swelling was considerably slower. Calretinin is a Ca(2+) buffer protein, which is expressed in a subset of TRPV1-positive neurons. Calretinin decreased the amplitude, but slowed down the decay of Ca(2+) signals evoked by ionomycin. Cells co-expressing TRPV1 and calretinin were less sensitive to TRPV1-mediated, capsaicin-induced volume increases. In TRPV1-expressing NIH3T3 cells, calretinin decreased the capsaicin-induced Ca(2+) and Na(+) influx. Swelling and formation of membrane blebs resulted in impaired plasma membrane integrity finally leading to cell death. Our results hint towards a mechanistic explanation for the apoptosis-independent capsaicin-evoked neuronal loss and additionally reveal a protective effect of calretinin; we propose that the Ca(2+)-buffering capacity of calretinin reduces the susceptibility of calretinin-expressing DRG neurons against cell swelling/death caused by overstimulation of TRPV1 channels. This article is part of a Special Issue entitled:12th European Symposium on Calcium.
Collapse
|
27
|
Klasen K, Hollatz D, Zielke S, Gisselmann G, Hatt H, Wetzel CH. The TRPM8 ion channel comprises direct Gq protein-activating capacity. Pflugers Arch 2012; 463:779-97. [PMID: 22460725 DOI: 10.1007/s00424-012-1098-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 03/12/2012] [Accepted: 03/14/2012] [Indexed: 11/29/2022]
Abstract
The transient receptor potential (TRP) family of ion channels comprises receptors that are activated by a vast variety of physical as well as chemical stimuli. TRP channels interact in a complex manner with several intracellular signaling cascades, both up- and downstream of receptor activation. Investigating cascades stimulated downstream of the cold and menthol receptor TRPM8, we found evidence for both, functional and structural interaction of TRPM8 with Gαq. We demonstrated menthol-evoked increase in intracellular Ca(2+) under extracellular Ca(2+)-free conditions, which was blocked by the PLC inhibitors U73122 or edelfosine. This metabotropic Ca(2+) signal could be observed also in cells expressing a channel-dead (i.e. non-conducting) or a chloride-conducting TRPM8 pore mutant. However, this intracellular metabotropic Ca(2+) signal could not be detected in Gαq deficient cells or in the presence of dominant-negative GαqX. Evidence for a close spatial proximity necessary for physical interaction of TRPM8 and Gαq was provided by acceptor bleaching experiments demonstrating FRET between TRPM8-CFP and Gαq-YFP. A Gαq-YFP mobility assay (FRAP) revealed a restricted diffusion of Gαq-YFP under conditions when TRPM8 is immobilized in the plasma membrane. Moreover, a menthol-induced and TRPM8-mediated G protein activation could be demonstrated by FRET experiments monitoring the dissociation of Gαq-YFP from a Gβ/Gγ-CFP complex, and by the exchange of radioactive [(35)S]GTPγS for GDP. Our observations lead to a view that extends the operational range of the TRPM8 receptor from its function as a pure ion channel to a molecular switch with additional metabotropic capacity.
Collapse
Affiliation(s)
- Katharina Klasen
- Lehrstuhl für Zellphysiologie, Ruhr-Universität Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Shapovalov G, Lehen’kyi V, Skryma R, Prevarskaya N. TRP channels in cell survival and cell death in normal and transformed cells. Cell Calcium 2011; 50:295-302. [DOI: 10.1016/j.ceca.2011.05.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 05/05/2011] [Accepted: 05/05/2011] [Indexed: 12/29/2022]
|
29
|
TRPV channels in tumor growth and progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:947-67. [PMID: 21290335 DOI: 10.1007/978-94-007-0265-3_49] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Transient receptor potential (TRP) channels affect several physiological and pathological processes. In particular, TRP channels have been recently involved in the triggering of enhanced proliferation, aberrant differentiation, and resistance to apoptotic cell death leading to the uncontrolled tumor invasion. About thirty TRPs have been identified to date, and are classified in seven different families: TRPC (Canonical), TRPV (Vanilloid), TRPM (Melastatin), TRPML (Mucolipin), TRPP (Polycystin), and TRPA (Ankyrin transmembrane protein) and TRPN (NomPC-like). Among these channel families, the TRPC, TRPM, and TRPV families have been mainly correlated with malignant growth and progression. The aim of this review is to summarize data reported so far on the expression and the functional role of TRPV channels during cancer growth and progression. TRPV channels have been found to regulate cancer cell proliferation, apoptosis, angiogenesis, migration and invasion during tumor progression, and depending on the stage of the cancer, up- and down-regulation of TRPV mRNA and protein expression have been reported. These changes may have cancer promoting effects by increasing the expression of constitutively active TRPV channels in the plasma membrane of cancer cells by enhancing Ca(2+)-dependent proliferative response; in addition, an altered expression of TRPV channels may also offer a survival advantage, such as resistance of cancer cells to apoptotic-induced cell death. However, recently, a role of TRPV gene mutations in cancer development, and a relationship between the expression of specific TRPV gene single nucleotide polymorphisms and increased cancer risk have been reported. We are only at the beginning, a more deep studies on the physiopathology role of TRPV channels are required to understand the functional activity of these channels in cancer, to assess which TRPV proteins are associated with the development and progression of cancer and to develop further knowledge of TRPV proteins as valuable diagnostic and/or prognostic markers, as well as targets for pharmaceutical intervention and targeting in cancer.
Collapse
|
30
|
TRPV1: A Therapy Target That Attracts the Pharmaceutical Interests. TRANSIENT RECEPTOR POTENTIAL CHANNELS 2011; 704:637-65. [DOI: 10.1007/978-94-007-0265-3_34] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
31
|
Gees M, Colsoul B, Nilius B. The role of transient receptor potential cation channels in Ca2+ signaling. Cold Spring Harb Perspect Biol 2010; 2:a003962. [PMID: 20861159 DOI: 10.1101/cshperspect.a003962] [Citation(s) in RCA: 323] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The 28 mammalian members of the super-family of transient receptor potential (TRP) channels are cation channels, mostly permeable to both monovalent and divalent cations, and can be subdivided into six main subfamilies: the TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin), TRPML (mucolipin), and the TRPA (ankyrin) groups. TRP channels are widely expressed in a large number of different tissues and cell types, and their biological roles appear to be equally diverse. In general, considered as polymodal cell sensors, they play a much more diverse role than anticipated. Functionally, TRP channels, when activated, cause cell depolarization, which may trigger a plethora of voltage-dependent ion channels. Upon stimulation, Ca2+ permeable TRP channels generate changes in the intracellular Ca2+ concentration, [Ca2+]i, by Ca2+ entry via the plasma membrane. However, more and more evidence is arising that TRP channels are also located in intracellular organelles and serve as intracellular Ca2+ release channels. This review focuses on three major tasks of TRP channels: (1) the function of TRP channels as Ca2+ entry channels; (2) the electrogenic actions of TRPs; and (3) TRPs as Ca2+ release channels in intracellular organelles.
Collapse
Affiliation(s)
- Maarten Gees
- KU Leuven, Department of Molecular Cell Biology, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, bus 802, Leuven, Belgium
| | | | | |
Collapse
|
32
|
Peng G, Lu W, Li X, Chen Y, Zhong N, Ran P, Wang J. Expression of store-operated Ca2+ entry and transient receptor potential canonical and vanilloid-related proteins in rat distal pulmonary venous smooth muscle. Am J Physiol Lung Cell Mol Physiol 2010; 299:L621-30. [PMID: 20693314 DOI: 10.1152/ajplung.00176.2009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic hypoxia causes remodeling and alters contractile responses in both pulmonary arteries and pulmonary veins. Although pulmonary arteries have been studied extensively in these disorders, the mechanisms by which pulmonary veins respond to hypoxia and whether these responses contribute to chronic hypoxic pulmonary hypertension remain poorly understood. In pulmonary arterial smooth muscle, we have previously demonstrated that influx of Ca(2+) through store-operated calcium channels (SOCC) thought to be composed of transient receptor potential (TRP) proteins is likely to play an important role in development of chronic hypoxic pulmonary hypertension. To determine whether this mechanism could also be operative in pulmonary venous smooth muscle, we measured intracellular Ca(2+) concentration ([Ca(2+)](i)) by fura-2 fluorescence microscopy in primary cultures of pulmonary venous smooth muscle cells (PVSMC) isolated from rat distal pulmonary veins. In cells perfused with Ca(2+)-free media containing cyclopiazonic acid (10 μM) and nifedipine (5 μM) to deplete sarcoplasmic reticulum Ca(2+) stores and block voltage-dependent Ca(2+) channels, restoration of extracellular Ca(2+) (2.5 mM) caused marked increases in [Ca(2+)](i), whereas MnCl(2) (200 μM) quenched fura-2 fluorescence, indicating store-operated Ca(2+) entry (SOCE). SKF-96365 and NiCl(2), antagonists of SOCC, blocked SOCE at concentrations that did not alter Ca(2+) responses to 60 mM KCl. Of the seven known canonical TRP (TRPC1-7) and six vanilloid-related TRP channels (TRPV1-6), real-time PCR revealed mRNA expression of TRPC1 > TRPC6 > TRPC4 > TRPC2 ≈ TRPC5 > TRPC3, TRPV2 > TRPV4 > TRPV1 in distal PVSMC, and TRPC1 > TRPC6 > TRPC3 > TRPC4 ≈ TRPC5, TRPV2 ≈ TRPV4 > TRPV1 in rat distal pulmonary vein (PV) smooth muscle. Western blotting confirmed protein expression of TRPC1, TRPC6, TRPV2, and TRPV4 in both PVSMC and PV. Our results suggest that SOCE through Ca(2+) channels composed of TRP proteins may contribute to Ca(2+) signaling in rat distal PV smooth muscle.
Collapse
Affiliation(s)
- Gongyong Peng
- Guangzhou Institute of Respiratory Diseases, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, China
| | | | | | | | | | | | | |
Collapse
|
33
|
Sinkins WG, Estacion M, Prasad V, Goel M, Shull GE, Kunze DL, Schilling WP. Maitotoxin converts the plasmalemmal Ca(2+) pump into a Ca(2+)-permeable nonselective cation channel. Am J Physiol Cell Physiol 2009; 297:C1533-43. [PMID: 19794142 DOI: 10.1152/ajpcell.00252.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Maitotoxin (MTX) activates Ca(2+)-permeable nonselective cation channels and causes a dramatic increase in cytosolic free Ca(2+) concentration ([Ca(2+)](i)) in every cell examined to date, but the molecular identity of the channels involved remains unknown. A clue came from studies of a structurally related marine toxin called palytoxin (PTX). PTX binds to the plasmalemmal Na(+)-K(+)-ATPase (NKA) and converts the Na(+) pump into a nonselective cation channel. Given the high permeability of the MTX channel for Ca(2+), we considered the possibility that MTX may bind to the plasmalemmal Ca(2+)-ATPase (PMCA) pump, and like PTX, convert the pump into a channel. To test this hypothesis, the PMCA was overexpressed in Spodoptera frugiperda (Sf9) insect cells and in human embryonic kidneys (HEK) 293 cells. In both cell types, enhanced expression of the PMCA was associated with a significant increase in MTX-induced whole cell membrane currents. The effect of MTX on whole cell currents in both wild-type and PMCA overexpressing HEK cells was sensitive to pump ligands including Ca(2+) and ATP. MTX-induced currents were significantly reduced by knockdown of PMCA1 in HEK cells using small interfering RNA or in mouse embryonic fibroblasts from genetically modified mice with the PMCA1(+/-) PMCA4(-/-) genotype. Finally, PMCA catalytic activity (i.e., Ca(2+)-ATPase) in isolated membranes, or in purified PMCA preparations, was inhibited by MTX. Together, these results suggest that MTX binds to and converts the PMCA pump into a Ca(2+)-permeable nonselective cation channel.
Collapse
Affiliation(s)
- William G Sinkins
- Department of Physiology, Rammelkamp Center for Education and Research, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Mitchell JE, Campbell AP, New NE, Sadofsky LR, Kastelik JA, Mulrennan SA, Compton SJ, Morice AH. EXPRESSION AND CHARACTERIZATION OF THE INTRACELLULAR VANILLOID RECEPTOR (TRPV1) IN BRONCHI FROM PATIENTS WITH CHRONIC COUGH. Exp Lung Res 2009; 31:295-306. [PMID: 15962710 DOI: 10.1080/01902140590918803] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
TRPV1 is a modulator of noxious stimuli known to be important in the cough reflex. We have compared the expression of TRPV1 in normal human airways and those from patients with chronic cough and found that there is up regulation in airways smooth muscle in disease. This increased expression appears to be intracellular and we have therefore examined the role of intracellular rat and human TRPV1 activity was found using intracellular calcium signalling with human intracellular TRPV1 being located in a thapsigargin insensitive compartment. Increase in TRPV1 activity may have a role in the airway hypersensitivity seen in chronic cough.
Collapse
Affiliation(s)
- Jennifer E Mitchell
- Department of Cellular Pathology, Hull Royal Infirmary, Hull, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Matsumoto-Miyai K, Kagase A, Murakawa Y, Momota Y, Kawatani M. Extracellular Ca2+ regulates the stimulus-elicited ATP release from urothelium. Auton Neurosci 2009; 150:94-9. [PMID: 19525154 DOI: 10.1016/j.autneu.2009.05.253] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Revised: 05/21/2009] [Accepted: 05/27/2009] [Indexed: 01/12/2023]
Abstract
Accumulating evidence shows that the epithelial cells in urinary bladder (urothelium) serve as a sensory organ in micturition and/or in nociception pathway by releasing ATP in response to mechanical and/or chemical stimuli. Here, we compared the effects of capsaicin, acetylcholine, and prostaglandin E(2) receptor EP1 agonist (ONO-DI-004) on the urothelial ATP release in primary cultured mouse urothelial cells in low Ca(2+) medium. All of these chemicals induced a gradual ATP release from urothelium, implying that the downstream Ca(2+) release from endoplasmic reticulum could trigger the ATP release. Consistent with this suggestion, blockade of inositol 1,4,5-triphosphate receptor reduced the distention-induced ATP release from urothelial tissues. The distention-induced ATP release was not affected by tetrodotoxin. However, an increase in extracellular Ca(2+) diminished both chemical- and distention-induced ATP release from urothelium. Thus raising the extracellular Ca(2+) concentration was found to inhibit stimulation-evoked ATP urothelial release.
Collapse
|
36
|
Gkika D, Prevarskaya N. Molecular mechanisms of TRP regulation in tumor growth and metastasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:953-8. [DOI: 10.1016/j.bbamcr.2008.11.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 11/20/2008] [Accepted: 11/21/2008] [Indexed: 12/11/2022]
|
37
|
A small component of the endoplasmic reticulum is required for store-operated Ca2+ channel activation in liver cells: evidence from studies using TRPV1 and taurodeoxycholic acid. Biochem J 2009; 418:553-66. [PMID: 19007332 DOI: 10.1042/bj20081052] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The question of whether the activation of SOCs (store-operated Ca(2+) channels) requires the whole or part of the ER (endoplasmic reticulum) has not been fully resolved. The role of a putative sub-compartment of the ER in SOC activation in liver cells was investigated using ectopically expressed TRPV1 (transient receptor potential vanilloid 1), a non-selective cation channel, and TDCA (taurodeoxycholic acid), an activator of SOCs, to release Ca(2+) from different regions of the ER. TRPV1 was expressed in the ER and in the plasma membrane. The amount of Ca(2+) released from the ER by a TRPV1 agonist, measured using fura-2, was the same as that released by a SERCA (sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase) inhibitor, indicating that TRPV1 agonist-sensitive stores substantially overlap with SERCA inhibitor-sensitive stores. In contrast with SERCA inhibitors, TRPV1 agonists did not activate store-operated Ca(2+) entry. These findings were confirmed by patch-clamp recording. Using FFP-18, it was shown that SERCA inhibitors release Ca(2+) from the ER located closer to the plasma membrane than the region from which TRPV1 agonists release Ca(2+). In contrast with SERCA inhibitors, TRPV1 agonists did not induce a redistribution of STIM1 (stromal interaction molecule 1). TDCA caused the release of Ca(2+) from the ER, which was detected by FFP-18 but not by fura-2, and a redistribution of STIM1 to puncta similar to that caused by SERCA inhibitors. It is concluded that in liver cells, Ca(2+) release from a small component of the ER located near the plasma membrane is required to induce STIM1 redistribution and SOC activation.
Collapse
|
38
|
Korepanova A, Pereda-Lopez A, Solomon LR, Walter KA, Lake MR, Bianchi BR, McDonald HA, Neelands TR, Shen J, Matayoshi ED, Moreland RB, Chiu ML. Expression and purification of human TRPV1 in baculovirus-infected insect cells for structural studies. Protein Expr Purif 2008; 65:38-50. [PMID: 19121396 DOI: 10.1016/j.pep.2008.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 11/11/2008] [Accepted: 12/03/2008] [Indexed: 10/21/2022]
Abstract
TRPV1 is a ligand-gated cation channel that is involved in acute thermal nociception and neurogenic inflammation. By using the GP67 signal peptide, high levels of full-length human TRPV1 was expressed in High Five insect cells using the baculovirus expression system. The functional activity of the expressed TRPV1 was confirmed by whole-cell ligand-gated ion flux recordings in the presence of capsaicin and low pH and via specific ligand binding to the isolated cellular membranes. Efficient solubilization and purification protocols have resulted in milligram amounts of detergent-solubilized channel at 80-90% purity after Ni2+ IMAC chromatography and size exclusion chromatography. Western blot analysis of amino and carboxyl terminal domains and MS of tryptic digestions of purified protein confirmed the presence of the full-length human TRPV1. Specific ligand binding experiments confirmed the protein integrity of the purified human TRPV1.
Collapse
Affiliation(s)
- Alla Korepanova
- Department of Structural Biology, R46Y, Abbott Laboratories, Bldg. AP10-LL8, 100 Abbott Park Rd., Abbott Park, IL 60064-6098, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Huang W, Wang H, Galligan JJ, Wang DH. Transient receptor potential vanilloid subtype 1 channel mediated neuropeptide secretion and depressor effects: role of endoplasmic reticulum associated Ca2+ release receptors in rat dorsal root ganglion neurons. J Hypertens 2008; 26:1966-75. [PMID: 18806620 PMCID: PMC2669742 DOI: 10.1097/hjh.0b013e328309eff9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE This study tests the hypothesis that the transient receptor potential vanilloid subtype 1 channel induced neuropeptide secretion and depressor response are mediated by, at least in part, activation of endoplasmic reticulum associated Ca release receptors, leading to increased cytosolic Ca in dorsal root ganglion neurons. METHODS/RESULTS Bolus injection of capsaicin (10 or 50 microg/kg), a selective transient receptor potential vanilloid subtype 1 channel agonist, into anesthetized male Wistar rats caused a dose-dependent decrease in mean arterial pressure (P < 0.05). Capsaicin (50 microg/kg)-induced depressor effects and increase in plasma calcitonin gene related peptide (CGRP) levels (-29 +/- 2 mmHg, 82.2 +/- 5.0 pg/ml) were abolished by a selective transient receptor potential vanilloid subtype 1 channel antagonist, capsazepine (3 mg/kg, -4 +/- 1 mmHg, 41.8 +/- 4.4 pg/ml, P < 0.01), and attenuated by a selective ryanodine receptor antagonist, dantrolene (5 mg/kg, -12 +/- 1 mmHg, 57.2 +/- 2.6 pg/ml, P < 0.01), but unaffected by an inhibitor of endoplasmic reticulum Ca-ATPase, thapsigargin (50 microg/kg, -30 +/- 1 mmHg, 73.8 +/- 2.3 pg/ml, P > 0.05), or an antagonist of the inositol (1,4,5)-trisphosphate receptor, 2-aminoethoxydiphenyl borate (3 mg/kg, -34 +/- 5 mmHg, 69.0 +/- 3.7 pg/ml, P > 0.05). CGRP8-37 (1 mg/kg), a selective CGRP receptor antagonist, also blocked capsaicin-induced depressor effects. In contrast, dantrolene had no effect on CGRP (1 microg/kg)-induced depressor effects. In vitro, capsaicin (0.3 micromol/l) increased intracellular Ca concentrations and CGRP release from freshly isolated sensory neurons in dorsal root ganglion (P < 0.01), which were blocked by capsazepine (10 micromol/l) and attenuated by dantrolene but not thapsigargin or 2-aminoethoxydiphenyl borate. CONCLUSION Our results indicate that transient receptor potential vanilloid subtype 1 channel activation triggers ryanodine receptor but not inositol (1,4,5)-trisphosphate receptor dependent Ca release from endoplasmic reticulum in dorsal root ganglion neurons, leading to increased CGRP release and consequent depressor effects.
Collapse
Affiliation(s)
- Wei Huang
- Department of Medicine and Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan
- Department of Cardiology of the First Affiliated Hospital, Chongqing Medical University, China
| | - Hui Wang
- Department of Medicine and Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan
| | - James J. Galligan
- Department of Medicine and Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan
| | - Donna H. Wang
- Department of Medicine and Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
40
|
Roh EJ, Keller JM, Olah Z, Iadarola MJ, Jacobson KA. Structure-activity relationships of 1,4-dihydropyridines that act as enhancers of the vanilloid receptor 1 (TRPV1). Bioorg Med Chem 2008; 16:9349-58. [PMID: 18809334 DOI: 10.1016/j.bmc.2008.08.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Revised: 08/15/2008] [Accepted: 08/22/2008] [Indexed: 10/21/2022]
Abstract
Vanilloid agonists such as capsaicin activate ion flux through the TRPV1 channel, a heat- and ligand-gated cation channel that transduces painful chemical or thermal stimuli applied to peripheral nerve endings in skin or deep tissues. We have probed the SAR of a variety of 1,4-dihydropyridine (DHP) derivatives as novel 'enhancers' of TRPV1 activity by examining changes in capsaicin-induced elevations in (45)Ca(2+)-uptake in either cells ectopically expressing TRPV1 or in cultured dorsal root ganglion (DRG) neurons. The enhancers increased the maximal capsaicin effect on (45)Ca(2+)-uptake by typically 2- to 3-fold without producing an action when used alone. The DHP enhancers contained 6-aryl substitution and small alkyl groups at the 1 and 4 positions, and a 3-phenylalkylthioester was tolerated. Levels of free intracellular Ca(2+), as measured by calcium imaging, were also increased in DRG neurons when exposed to the combination of capsaicin and the most efficacious enhancer 23 compared to capsaicin alone. Thus, DHPs can modulate TRPV1 channels in a positive fashion.
Collapse
Affiliation(s)
- Eun Joo Roh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Building 8A, Room B1A-19 LBC, Bethesda, MD 20892-0810, USA
| | | | | | | | | |
Collapse
|
41
|
Jeske NA, Diogenes A, Ruparel NB, Fehrenbacher JC, Henry M, Akopian AN, Hargreaves KM. A-kinase anchoring protein mediates TRPV1 thermal hyperalgesia through PKA phosphorylation of TRPV1. Pain 2008; 138:604-616. [PMID: 18381233 DOI: 10.1016/j.pain.2008.02.022] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 12/17/2007] [Accepted: 02/20/2008] [Indexed: 11/27/2022]
Abstract
Certain phosphorylation events are tightly controlled by scaffolding proteins such as A-kinase anchoring protein (AKAP). On nociceptive terminals, phosphorylation of transient receptor potential channel type 1 (TRPV1) results in the sensitization to many different stimuli, contributing to the development of hyperalgesia. In this study, we investigated the functional involvement of AKAP150 in mediating sensitization of TRPV1, and found that AKAP150 is co-expressed in trigeminal ganglia (TG) neurons from rat and associates with TRPV1. Furthermore, siRNA-mediated knock-down of AKAP150 expression led to a significant reduction in PKA phosphorylation of TRPV1 in cultured TG neurons. In CHO cells, the PKA RII binding site on AKAP was necessary for PKA enhancement of TRPV1-mediated Ca2+-accumulation. In addition, AKAP150 knock-down in cultured TG neurons attenuated PKA sensitization of TRPV1 activity and in vivo administration of an AKAP antagonist significantly reduced prostaglandin E2 sensitization to thermal stimuli. These data suggest that AKAP150 functionally regulates PKA-mediated phosphorylation/sensitization of the TRPV1 receptor.
Collapse
Affiliation(s)
- Nathaniel A Jeske
- Department of Oral and Maxillofacial Surgery, University of Texas Health Science Center of San Antonio, MC 7908, 7703 Floyd Curl Dr., San Antonio, TX 78229-3900, USA Department of Endodontics, University of Texas Health Science Center of San Antonio, San Antonio, TX, USA Department of Cellular and Structural Biology, University of Texas Health Science Center of San Antonio, San Antonio, TX, USA Department of Pharmacology, University of Texas Health Science Center of San Antonio, San Antonio, TX, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Löf C, Blom T, Törnquist K. Overexpression of TRPC3 reduces the content of intracellular calcium stores in HEK-293 cells. J Cell Physiol 2008; 216:245-52. [DOI: 10.1002/jcp.21396] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
43
|
Hagenacker T, Ledwig D, Büsselberg D. Feedback mechanisms in the regulation of intracellular calcium ([Ca2+]i) in the peripheral nociceptive system: role of TRPV-1 and pain related receptors. Cell Calcium 2007; 43:215-27. [PMID: 17673288 DOI: 10.1016/j.ceca.2007.05.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 05/17/2007] [Accepted: 05/30/2007] [Indexed: 11/21/2022]
Abstract
Multimodal stimuli like heat, cold, bacterial or mechanical events are able to elicit pain, which is necessary to guarantee survival. However, the control of pain is of major clinical importance. The perception and transduction of pain is differentially modulated in the peripheral and central nervous system (CNS): while peripheral structures modulate these signals, the perception of pain occurs in the CNS. In recent years major advances have been made in the understanding of the processes which are involved in pain sensation. For the peripheral pain reception, the importance of specific pain receptors of the transition receptor pore (TRP)-family (e.g. the TRPV-1 receptor) has been analyzed. These receptors/channels are localized at the cell membrane of nociceptive neurones as well as in membranes of intracellular calcium stores like the endoplasmic reticulum. While the associated channel conducts different ions, a major proportion is calcium. Therefore, this review focuses on (1) the modulations of intracellular calcium ([Ca2+]i) initiated by the activation of pain receptors and (2) the consequences of [Ca2+]i changes for the processing of pain signals at the peripheral side. The possible interference of TRPV-1 induced [Ca2+]i modulations to the function of other membrane receptors and channels, like voltage gated calcium, sodium or potassium channels, or co-expressed CB1-receptors will be discussed. The latter interactions are of specific interest since the analgetic properties of endo- and exo-cannabinoids are mediated by CB1 receptors and their activation significantly modulates the calcium induced release of pain related transmitters. Furthermore, multiple cross links between different pain modulating intracellular pathways and their dependence on [Ca2+]i modulations will be illuminated. Overall, this review will summarize new insights resulting in the understanding of the prominent influence of [Ca2+]i for processes which are involved in pain sensation.
Collapse
Affiliation(s)
- T Hagenacker
- Universitätsklinikum Essen, Institut für Physiologie, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | |
Collapse
|
44
|
Abstract
Capacitative Ca2+ entry links the emptying of intracellular Ca2+ stores to the activation of store-operated Ca2+ channels in the plasma membrane. In the twenty years since the inception of the concept of capacitative Ca2+ entry, a number of activation mechanisms have been proposed, and there has been considerable interest in the possibility that TRP channels function as store-operated channels. However, in the past two years, two major players in both the signaling and permeation mechanisms for store-operated channels have been discovered: Stim1 and the Orai proteins. Stim1 is an endoplasmic reticulum Ca2+ sensor. It appears to act by redistributing within a small component of the endoplasmic reticulum, approaching the plasma membrane, but does not seem to translocate into the plasma membrane. Stim1 signals to plasma membrane Orai proteins, which constitute pore-forming subunits of store-operated channels.
Collapse
Affiliation(s)
- James W Putney
- National Institute of Environmental Health Sciences - NIH, PO Box 12233, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
45
|
Putney JW. Recent breakthroughs in the molecular mechanism of capacitative calcium entry (with thoughts on how we got here). Cell Calcium 2007; 42:103-10. [PMID: 17349691 PMCID: PMC1986648 DOI: 10.1016/j.ceca.2007.01.011] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Accepted: 01/29/2007] [Indexed: 11/24/2022]
Abstract
Activation of phospholipase C by G-protein-coupled receptors results in release of intracellular Ca(2+) and activation of Ca(2+) channels in the plasma membrane. The intracellular release of Ca(2+) is signaled by the second messenger, inositol 1,4,5-trisphosphate. Ca(2+) entry involves signaling from depleted intracellular stores to plasma membrane Ca(2+) channels, a process referred to as capacitative calcium entry or store-operated calcium entry. The electrophysiological current associated with capacitative calcium entry is the calcium-release-activated calcium current, or I(crac). In the 20 years since the inception of the concept of capacitative calcium entry, a variety of activation mechanisms have been proposed, and there has been considerable interest in the possibility of transient receptor potential channels functioning as store-operated channels. However, in the past 2 years, two major players in both the signaling and permeation mechanisms for store-operated channels have been discovered: Stim1 (and possibly Stim2) and the Orai proteins. Activation of store-operated channels involves an endoplasmic reticulum Ca(2+) sensor called Stim1. Stim1 acts by redistributing within a small component of the endoplasmic reticulum, approaching the plasma membrane, but does not appear to translocate into the plasma membrane. Stim1, either directly or indirectly, signals to plasma membrane Orai proteins which constitute pore-forming subunits of store-operated channels.
Collapse
Affiliation(s)
- James W Putney
- National Institute of Environmental Health Sciences, NIH, P.O. Box 12233, Research Triangle Park, NC 27709, United States.
| |
Collapse
|
46
|
Vetter I, Wyse BD, Monteith GR, Roberts-Thomson SJ, Cabot PJ. The mu opioid agonist morphine modulates potentiation of capsaicin-evoked TRPV1 responses through a cyclic AMP-dependent protein kinase A pathway. Mol Pain 2006; 2:22. [PMID: 16842630 PMCID: PMC1553434 DOI: 10.1186/1744-8069-2-22] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2006] [Accepted: 07/16/2006] [Indexed: 01/23/2023] Open
Abstract
Background The vanilloid receptor 1 (TRPV1) is critical in the development of inflammatory hyperalgesia. Several receptors including G-protein coupled prostaglandin receptors have been reported to functionally interact with the TRPV1 through a cAMP-dependent protein kinase A (PKA) pathway to potentiate TRPV1-mediated capsaicin responses. Such regulation may have significance in inflammatory pain. However, few functional receptor interactions that inhibit PKA-mediated potentiation of TRPV1 responses have been described. Results In the present studies we investigated the hypothesis that the μ opioid receptor (MOP) agonist morphine can modulate forskolin-potentiated capsaicin responses through a cAMP-dependent PKA pathway. HEK293 cells were stably transfected with TRPV1 and MOP, and calcium (Ca2+) responses to injection of the TRPV1 agonist capsaicin were monitored in Fluo-3-loaded cells. Pre-treatment with morphine did not inhibit unpotentiated capsaicin-induced Ca2+ responses but significantly altered capsaicin responses potentiated by forskolin. TRPV1-mediated Ca2+ responses potentiated by the direct PKA activator 8-Br-cAMP and the PKC activator Phorbol-12-myristate-13-acetatewere not modulated by morphine. Immunohistochemical studies confirmed that the TRPV1 and MOP are co-expressed on cultured Dorsal Root Ganglion neurones, pointing towards the existence of a functional relationship between the G-protein coupled MOP and nociceptive TRPV1. Conclusion The results presented here indicate that the opioid receptor agonist morphine acts via inhibition of adenylate cyclase to inhibit PKA-potentiated TRPV1 responses. Targeting of peripheral opioid receptors may therefore have therapeutic potential as an intervention to prevent potentiation of TRPV1 responses through the PKA pathway in inflammation.
Collapse
Affiliation(s)
- Irina Vetter
- The School of Pharmacy, The University of Queensland, Brisbane, 4072, Australia
| | - Bruce D Wyse
- The School of Pharmacy, The University of Queensland, Brisbane, 4072, Australia
| | - Gregory R Monteith
- The School of Pharmacy, The University of Queensland, Brisbane, 4072, Australia
| | | | - Peter J Cabot
- The School of Pharmacy, The University of Queensland, Brisbane, 4072, Australia
| |
Collapse
|
47
|
Lievremont JP, Numaga T, Vazquez G, Lemonnier L, Hara Y, Mori E, Trebak M, Moss SE, Bird GS, Mori Y, Putney JW. The role of canonical transient receptor potential 7 in B-cell receptor-activated channels. J Biol Chem 2005; 280:35346-51. [PMID: 16123040 DOI: 10.1074/jbc.m507606200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phospholipase C signaling stimulates Ca2+ entry across the plasma membrane through multiple mechanisms. Ca2+ store depletion stimulates store-operated Ca2+-selective channels, or alternatively, other phospholipase C-dependent events activate Ca2+-permeable non-selective cation channels. Transient receptor potential 7 (TRPC7) is a non-selective cation channel that can be activated by both mechanisms when ectopically expressed, but the regulation of native TRPC7 channels is not known. We knocked out TRPC7 in DT40 B-cells, which expresses both forms of Ca2+ entry. No difference in the store-operated current I(crac) was detected between TRPC7-/- and wild-type cells. Wild-type cells demonstrated nonstore-operated cation entry and currents in response to activation of the B-cell receptor or protease-activated receptor 2, intracellular dialysis with GTPgammaS, or application of the synthetic diacylglycerol oleyl-acetyl-glycerol. These responses were absent in TRPC7-/- cells but could be restored by transfection with human TRPC7. In conclusion, in B-lymphocytes, TRPC7 appeared to participate in the formation of ion channels that could be activated by phospholipase C-linked receptors. This represents the first demonstration of a physiological function for endogenous TRPC7 channels.
Collapse
Affiliation(s)
- Jean-Philippe Lievremont
- Department of Health and Human Services, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Goel M, Sinkins W, Keightley A, Kinter M, Schilling WP. Proteomic analysis of TRPC5- and TRPC6-binding partners reveals interaction with the plasmalemmal Na+/K+-ATPase. Pflugers Arch 2005; 451:87-98. [PMID: 16025302 DOI: 10.1007/s00424-005-1454-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Accepted: 04/21/2005] [Indexed: 10/25/2022]
Abstract
Mammalian transient receptor potential canonical (TRPC) genes encode a family of nonselective cation channels that are activated following stimulation of G-protein-coupled membrane receptors linked to phospholipase C. In Drosophila photoreceptor cells, TRP channels are found in large, multimolecular signaling complexes in association with the PDZ-containing scaffolding protein, INAD. A similar mammalian TRPC "signalplex" has been proposed, but has yet to be defined. In the present study, affinity-purified polyclonal antibodies against TRPC5 and TRPC6 were used to immunoprecipitate signalplex components from rat brain lysates. Immunoprecipitated proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, digested with trypsin, and sequenced by mass spectrometry. Proteins identified in the immunoprecipitates included cytoskeletal proteins spectrin, myosin, actin, drebrin, tubulin, and neurabin; endocytic vesicle-associated proteins clathrin, dynamin and AP-2; and the plasmalemmal Na(+)/K(+)-ATPase (NKA) pump. Several of these interactions were confirmed by reciprocal immunoprecipitation followed by Western blot analysis. In lysates from rat kidney, TRPC6, but not TRPC3, was found to coimmunoprecipitate with the NKA pump. Likewise, TRPC6, stably expressed in human embryonic kidney (HEK) cells, coimmunoprecipitated with endogenous NKA and colocalized with the pump to the plasmalemma when examined by immunofluorescence microscopy. Cell surface biotinylation experiments in intact HEK cells, confirmed that both the Na(+) pump and TRPC6 were present in the surface membrane and appeared to interact. Lastly, TRPC6 coimmunoprecipitated with the NKA pump when the proteins were coexpressed in Spodoptera frugiperda insect cells using recombinant baculoviruses. These observations suggest that TRPC6 and the Na(+) pump are part of a functional complex that may be involved in ion transport and homeostasis in both the brain and kidney.
Collapse
Affiliation(s)
- Monu Goel
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
In electrically nonexcitable cells, Ca2+influx is essential for regulating a host of kinetically distinct processes involving exocytosis, enzyme control, gene regulation, cell growth and proliferation, and apoptosis. The major Ca2+entry pathway in these cells is the store-operated one, in which the emptying of intracellular Ca2+stores activates Ca2+influx (store-operated Ca2+entry, or capacitative Ca2+entry). Several biophysically distinct store-operated currents have been reported, but the best characterized is the Ca2+release-activated Ca2+current, ICRAC. Although it was initially considered to function only in nonexcitable cells, growing evidence now points towards a central role for ICRAC-like currents in excitable cells too. In spite of intense research, the signal that relays the store Ca2+content to CRAC channels in the plasma membrane, as well as the molecular identity of the Ca2+sensor within the stores, remains elusive. Resolution of these issues would be greatly helped by the identification of the CRAC channel gene. In some systems, evidence suggests that store-operated channels might be related to TRP homologs, although no consensus has yet been reached. Better understood are mechanisms that inactivate store-operated entry and hence control the overall duration of Ca2+entry. Recent work has revealed a central role for mitochondria in the regulation of ICRAC, and this is particularly prominent under physiological conditions. ICRACtherefore represents a dynamic interplay between endoplasmic reticulum, mitochondria, and plasma membrane. In this review, we describe the key electrophysiological features of ICRACand other store-operated Ca2+currents and how they are regulated, and we consider recent advances that have shed insight into the molecular mechanisms involved in this ubiquitous and vital Ca2+entry pathway.
Collapse
Affiliation(s)
- Anant B Parekh
- Department of Physiology, University of Oxford, United Kingdom.
| | | |
Collapse
|
50
|
Vriens J, Janssens A, Prenen J, Nilius B, Wondergem R. TRPV channels and modulation by hepatocyte growth factor/scatter factor in human hepatoblastoma (HepG2) cells. Cell Calcium 2004; 36:19-28. [PMID: 15126053 DOI: 10.1016/j.ceca.2003.11.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Revised: 11/18/2003] [Accepted: 11/19/2003] [Indexed: 10/26/2022]
Abstract
Using patch clamp and Ca(2+) imaging techniques, we have studied Ca(2+) entry pathways in human hepatoblastoma (HepG2) cells. These cells express the mRNA of TRPV1, TRPV2, TRPV3 and TRPV4 channels, but not those of TRPV5 and TRPV6. Functional assessment showed that capsaicin (10 microM), 4alpha-phorbol-12,13-didecanoate (4alphaPDD, 1 microM), arachidonic acid (10 microM), hypotonic stress, and heat all stimulated increases in [Ca(2+)](i) within minutes. The increase in [Ca(2+)](i) depended on extracellular Ca(2+) and on the transmembrane potential, which indicated that both driving forces affected Ca(2+) entry. Capsaicin also stimulated an increase in [Ca(2+)](i) in nominally Ca(2+)-free solutions, which was compatible with the receptor functioning as a Ca(2+) release channel. Hepatocyte growth factor/scatter factor (HGF/SF) modulated Ca(2+) entry. Ca(2+) influx was greater in HepG2 cells incubated with HGF/SF (20 ng/ml for 20 h) compared with non-stimulated cells, but this occurred only in those cells with a migrating phenotype as determined by presence of a lamellipodium and trailing footplate. The effect of capsaicin on [Ca(2+)](i) was greater in migrating HGF/SF-treated cells, and this was inhibited by capsazepine. The difference between control and HGF/SF-treated cells was not found in Ca(2+)-free solutions. 4alphaPDD also had no greater effect on HGF/SF-treated cells. We conclude that TRPV1 and TRPV4 channels provide Ca(2+) entry pathways in HepG2 cells. HGF/SF increases Ca(2+) entry via TRPV1, but not via TRPV4. This rise in [Ca(2+)](i) may constitute an early response of a signalling cascade that gives rise to cell locomotion and the migratory phenotype.
Collapse
Affiliation(s)
- Joris Vriens
- Department of Physiology, Campus Gasthuisberg, Katholieke Universiteit, B-3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|