551
|
Nakatsuka M, Iwai Y. Expression of TRPV4 in the stimulated rat oral mucous membrane--nociceptive mechanisms of lingual conical papillae. Okajimas Folia Anat Jpn 2009; 86:45-54. [PMID: 19877445 DOI: 10.2535/ofaj.86.45] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The study was supported by 2006-2007 Aid Program for Overseas Training of the Promotion and Mutual Aid Corporation for Private School of Japan and International Exchange Grant, Osaka Dental University. We studied the function of TRPV4 expression and its neuronal activation in response to noxious stimulation of oral mucosa. The intermolar region of dorsal lingual eminence (IDLE) of rats was stimulated with 10 microl of either normal saline or 5% formalin. Immunohistological studies of the TRPV4, pERK and serotonin (5HT) expression in designated regions of tongues and brainstems were performed for studying the descending pain modulatory system in response to nociception. Specimens of the experimental IDLE demonstrated a significant increase of TRPV4 activity in particular in stratum basale of conical papillae (p < 0.01). pERK-IR positive neurons were significantly increased in the RMg (p < 0.05), Sp5C (p < 0.05) and Md (p < 0.01); TRPV4-IR neurons were found to show a similar distribution with pERK-IR cells in the peripheral Sp5C (p < 0.05). A significant increase of 5HT expression was observed in the RMg (p < 0.01), RPa (p < 0.01) and ROb (p < 0.05). The results suggest that TRPV4 in the oral mucosa is nociceptor of peripheral hyperalgesia, and pERK expression in the Sp5C is closely related with central hyperalgesia of the nociception. Furthermore, pERK-IR cells of the central 5HT nervous system are activated to accelerate 5HT release for neuronal modulation of the descending pain modulatory system in response to nociception.
Collapse
Affiliation(s)
- Michiko Nakatsuka
- Dept. of Oral Anatomy, Osaka Dental University, Kuzuha Hanazono-cho 8-1, Hirakata, Osaka 573-1121, Japan.
| | | |
Collapse
|
552
|
Watanabe H. Pathological role of TRP channels in cardiovascular and respiratory diseases. Nihon Yakurigaku Zasshi 2009; 134:127-30. [PMID: 19749483 DOI: 10.1254/fpj.134.127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
553
|
Inoue R, Mori M, Kawarabayashi Y, Jian Z. Chemical-mechanical synergism for cardiovascular TRPC6 channel activation via PLC/diacylglycerol and PLA2/omega-hydroxylase/20-HETE pathways. Nihon Yakurigaku Zasshi 2009; 134:116-21. [PMID: 19749481 DOI: 10.1254/fpj.134.116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
554
|
Hu H, Tian J, Zhu Y, Wang C, Xiao R, Herz JM, Wood JD, Zhu MX. Activation of TRPA1 channels by fenamate nonsteroidal anti-inflammatory drugs. Pflugers Arch 2009; 459:579-92. [PMID: 19888597 DOI: 10.1007/s00424-009-0749-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 10/12/2009] [Accepted: 10/14/2009] [Indexed: 11/24/2022]
Abstract
Transient receptor potential A1 (TRPA1) forms nonselective cation channels implicated in acute inflammatory pain and nociception. The mechanism of ligand activation of TRPA1 may involve either covalent modification of cysteine residues or conventional reversible ligand-receptor interactions. For certain electrophilic prostaglandins, covalent modification has been considered as the main mechanism involved in their stimulatory effect on TRPA1. Because some nonsteroidal anti-inflammatory drugs (NSAIDs) are structural analogs of prostaglandins, we examined several nonelectrophilic NSAIDs on TRPA1 activation using electrophysiological techniques and intracellular Ca(2+) measurements and found that a selected group of NSAIDs can act as TRPA1 agonists. Extracellularly applied flufenamic, niflumic, and mefenamic acid, as well as flurbiprofen, ketoprofen, diclofenac, and indomethacin, rapidly activated rat TRPA1 expressed in Xenopus oocytes and human TRPA1 endogenously expressed in WI-38 fibroblasts. Similarly, the NSAID ligands activated human TRPA1 inducibly expressed in HEK293 cells, but the responses were absent in uninduced and parental HEK293 cells. The response to fenamate agonists was blocked by TRPA1 antagonists, AP-18, HC-030031, and ruthenium red. At subsaturating concentrations, the fenamate NSAIDs also potentiate the activation of TRPA1 by allyl isothiocyanate, cinnamaldehyde, and cold, demonstrating positive synergistic interactions with other well-characterized TRPA1 activators. Importantly, among several thermosensitive TRP channels, the stimulatory effect is specific to TRPA1 because flufenamic acid inhibited TRPV1, TRPV3, and TRPM8. We conclude that fenamate NSAIDs are a novel class of potent and reversible direct agonists of TRPA1. This selective group of TRPA1-stimulating NSAIDs should provide a structural basis for developing novel ligands that noncovalently interact with TRPA1 channels.
Collapse
Affiliation(s)
- Hongzhen Hu
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
555
|
Identification and characterization of novel TRPV4 modulators. Biochem Biophys Res Commun 2009; 389:490-4. [DOI: 10.1016/j.bbrc.2009.09.007] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 09/02/2009] [Indexed: 11/19/2022]
|
556
|
Phelps CB, Wang RR, Choo SS, Gaudet R. Differential regulation of TRPV1, TRPV3, and TRPV4 sensitivity through a conserved binding site on the ankyrin repeat domain. J Biol Chem 2009; 285:731-40. [PMID: 19864432 DOI: 10.1074/jbc.m109.052548] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transient receptor potential vanilloid (TRPV) channels, which include the thermosensitive TRPV1-V4, have large cytoplasmic regions flanking the transmembrane domain, including an N-terminal ankyrin repeat domain. We show that a multiligand binding site for ATP and calmodulin previously identified in the TRPV1 ankyrin repeat domain is conserved in TRPV3 and TRPV4, but not TRPV2. Accordingly, TRPV2 is insensitive to intracellular ATP, while, as previously observed with TRPV1, a sensitizing effect of ATP on TRPV4 required an intact binding site. In contrast, ATP reduced TRPV3 sensitivity and potentiation by repeated agonist stimulations. Thus, ATP and calmodulin, acting through this conserved binding site, are key players in generating the different sensitivity and adaptation profiles of TRPV1, TRPV3, and TRPV4. Our results suggest that competing interactions of ATP and calmodulin influence channel sensitivity to fluctuations in calcium concentration and perhaps even metabolic state. Different feedback mechanisms likely arose because of the different physiological stimuli or temperature thresholds of these channels.
Collapse
Affiliation(s)
- Christopher B Phelps
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | | | | | | |
Collapse
|
557
|
Non-CB1, Non-CB2 Receptors for Endocannabinoids, Plant Cannabinoids, and Synthetic Cannabimimetics: Focus on G-protein-coupled Receptors and Transient Receptor Potential Channels. J Neuroimmune Pharmacol 2009; 5:103-21. [DOI: 10.1007/s11481-009-9177-z] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 09/24/2009] [Indexed: 12/24/2022]
|
558
|
Everaerts W, Nilius B, Owsianik G. The vanilloid transient receptor potential channel TRPV4: from structure to disease. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2009; 103:2-17. [PMID: 19835908 DOI: 10.1016/j.pbiomolbio.2009.10.002] [Citation(s) in RCA: 264] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 10/07/2009] [Indexed: 12/19/2022]
Abstract
The Transient Receptor Potential Vanilloid 4 channel, TRPV4, is a Ca(2+) and Mg(2+) permeable non-selective cation channel involved in many different cellular functions. It is activated by a variety of physical and chemical stimuli, including heat, mechano-stimuli, endogenous substances such as arachidonic acid and its cytochrome P450-derived metabolites (epoxyeicosatrienoic acids), endocannabinoids (anandamide and 2-arachidonoylglycerol), as well as synthetic alpha-phorbol derivatives. Recently, TRPV4 has been characterized as an important player modulating osteoclast differentiation in bone remodelling and as a urothelial mechanosensor that controls normal voiding. Several TRPV4 gain-of-function mutations are shown to cause autosomal-dominant bone dysplasias such as brachyolmia and Koszlowski disease. In this review we comprehensively describe the structural, biophysical and (patho)physiological properties of the TRPV4 channel and we summarize the current knowledge about the role of TRPV4 in the pathogenesis of several diseases.
Collapse
Affiliation(s)
- Wouter Everaerts
- Department of Molecular Cell Biology, Laboratory Ion Channel Research, Campus Gasthuisberg, KULeuven, Herestraat 49, bus 802, B-3000 Leuven, Belgium
| | | | | |
Collapse
|
559
|
Fan HC, Zhang X, McNaughton PA. Activation of the TRPV4 ion channel is enhanced by phosphorylation. J Biol Chem 2009; 284:27884-27891. [PMID: 19661060 PMCID: PMC2788839 DOI: 10.1074/jbc.m109.028803] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Revised: 08/04/2009] [Indexed: 11/06/2022] Open
Abstract
The TRPV4 (transient receptor potential vanilloid 4) ion channel, a member of the vanilloid subfamily of the transient receptor potential channels, is activated by membrane stretch, by non-noxious warm temperatures, and by a range of chemical activators. In the present study we examined the role of phosphorylation in modulating the activation of TRPV4. We expressed TRPV4 in HEK293 cells and activated the channel by cell swelling in a hypotonic solution. TRPV4 channel activation and serine phosphorylation were enhanced by exposure to the protein kinase C (PKC) activator phorbol 12-myristate 13-acetate or by application of bradykinin, which activates PKC via a G-protein-coupled mechanism. The enhancement was inhibited by the PKC inhibitors staurosporine, bisindolylmaleimide I, and rottlerin or by mutation of the serine/threonine residues Ser(162), Thr(175), and Ser(189). The adenylate cyclase activator forskolin also enhanced activation of TRPV4, and the enhancement was antagonized by the selective cyclic AMP-dependent protein kinase (PKA) inhibitor H89 or by mutation of serine residue Ser(824). Sensitization of TRPV4 by both PKC and PKA depended on the scaffolding protein AKAP79, because channel activation and phosphorylation were enhanced by co-transfection of AKAP79 and were antagonized by removal of AKAP79 using small interfering RNA. We conclude that the serine/threonine kinases PKC and PKA enhance activation of the TRPV4 ion channel by phosphorylation at specific sites and that phosphorylation depends on assembly of PKC and PKA by AKAP79 into a signaling complex with TRPV4.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom; Department of Pediatrics, Tri-Service General Hospital and National Defense Medical Center, Taipei 11490, Taiwan
| | - Xuming Zhang
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom
| | - Peter A McNaughton
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom.
| |
Collapse
|
560
|
Xiao R, Xu XZS. Function and regulation of TRP family channels in C. elegans. Pflugers Arch 2009; 458:851-60. [PMID: 19421772 PMCID: PMC2857680 DOI: 10.1007/s00424-009-0678-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2009] [Revised: 04/25/2009] [Accepted: 04/28/2009] [Indexed: 12/26/2022]
Abstract
Seventeen transient receptor potential (TRP) family proteins are encoded by the C. elegans genome, and they cover all of the seven TRP subfamilies, including TRPC, TRPV, TRPM, TRPN, TRPA, TRPP, and TRPML. Classical forward and reverse genetic screens have isolated mutant alleles in every C. elegans trp gene, and their characterizations have revealed novel functions and regulatory mechanisms of TRP channels. For example, the TRPC channels TRP-1 and TRP-2 control nicotine-dependent behavior, while TRP-3, a sperm TRPC channel, is regulated by sperm activation and required for sperm-egg interactions during fertilization. Similar to their vertebrate counterparts, C. elegans TRPs function in sensory physiology. For instance, the TRPV channels OSM-9 and OCR-2 act in chemosensation, osmosensation, and touch sensation, the TRPA member TRPA-1 regulates touch sensation, while the TRPN channel TRP-4 mediates proprioception. Some C. elegans TRPM, TRPP, and TRPML members exhibit cellular functions similar to their vertebrate homologues and have provided insights into human diseases, including polycystic kidney disease, hypomagnesemia, and mucolipidosis type IV. The availability of a complete set of trp gene mutants in conjunction with its facile genetics makes C. elegans a powerful model for studying the function and regulation of TRP family channels in vivo.
Collapse
Affiliation(s)
- Rui Xiao
- Life Sciences Institute and Department of Molecular & Integrative Physiology, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
561
|
Falck JR, Kodela R, Manne R, Atcha KR, Puli N, Dubasi N, Manthati VL, Capdevila JH, Yi XY, Goldman DH, Morisseau C, Hammock BD, Campbell WB. 14,15-Epoxyeicosa-5,8,11-trienoic acid (14,15-EET) surrogates containing epoxide bioisosteres: influence upon vascular relaxation and soluble epoxide hydrolase inhibition. J Med Chem 2009; 52:5069-75. [PMID: 19653681 PMCID: PMC2888647 DOI: 10.1021/jm900634w] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
All-cis-14,15-epoxyeicosa-5,8,11-trienoic acid (14,15-EET) is a labile, vasodilatory eicosanoid generated from arachidonic acid by cytochrome P450 epoxygenases. A series of robust, partially saturated analogues containing epoxide bioisosteres were synthesized and evaluated for relaxation of precontracted bovine coronary artery rings and for in vitro inhibition of soluble epoxide hydrolase (sEH). Depending upon the bioisostere and its position along the carbon chain, varying levels of vascular relaxation and/or sEH inhibition were observed. For example, oxamide 16 and N-iPr-amide 20 were comparable (ED(50) 1.7 microM) to 14,15-EET as vasorelaxants but were approximately 10-35 times less potent as sEH inhibitors (IC(50) 59 and 19 microM, respectively); unsubstituted urea 12 showed useful activity in both assays (ED(50) 3.5 microM, IC(50) 16 nM). These data reveal differential structural parameters for the two pharmacophores that could assist the development of potent and specific in vivo drug candidates.
Collapse
Affiliation(s)
- J R Falck
- Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
562
|
Tian W, Fu Y, Garcia-Elias A, Fernández-Fernández JM, Vicente R, Kramer PL, Klein RF, Hitzemann R, Orwoll ES, Wilmot B, McWeeney S, Valverde MA, Cohen DM. A loss-of-function nonsynonymous polymorphism in the osmoregulatory TRPV4 gene is associated with human hyponatremia. Proc Natl Acad Sci U S A 2009; 106:14034-9. [PMID: 19666518 PMCID: PMC2729015 DOI: 10.1073/pnas.0904084106] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Indexed: 12/24/2022] Open
Abstract
Disorders of water balance are among the most common and morbid of the electrolyte disturbances, and are reflected clinically as abnormalities in the serum sodium concentration. The transient receptor potential vanilloid 4 (TRPV4) channel is postulated to comprise an element of the central tonicity-sensing mechanism in the mammalian hypothalamus, and is activated by hypotonic stress in vitro. A nonsynonymous polymorphism in the TRPV4 gene gives rise to a Pro-to-Ser substitution at residue 19. We show that this polymorphism is significantly associated with serum sodium concentration and with hyponatremia (serum sodium concentration < or =135 mEq/L) in 2 non-Hispanic Caucasian male populations; in addition, mean serum sodium concentration is lower among subjects with the TRPV4(P19S) allele relative to the wild-type allele. Subjects with the minor allele were 2.4-6.4 times as likely to exhibit hyponatremia as subjects without the minor allele (after inclusion of key covariates). Consistent with these observations, a human TRPV4 channel mutated to incorporate the TRPV4(P19S) polymorphism showed diminished response to hypotonic stress (relative to the wild-type channel) and to the osmotransducing lipid epoxyeicosatrienoic acid in heterologous expression studies. These data suggest that this polymorphism affects TRPV4 function in vivo and likely influences systemic water balance on a population-wide basis.
Collapse
Affiliation(s)
- Wei Tian
- Department of Medicine, Divisions of Nephrology and Hypertension, and
- Behavioral Neuroscience, and
| | - Yi Fu
- Department of Medicine, Divisions of Nephrology and Hypertension, and
- Portland VA Medical Center, Portland, OR 97239; and
| | - Anna Garcia-Elias
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - José M. Fernández-Fernández
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rubén Vicente
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Robert F. Klein
- Endocrinology and Metabolism
- Portland VA Medical Center, Portland, OR 97239; and
| | - Robert Hitzemann
- Behavioral Neuroscience, and
- Portland VA Medical Center, Portland, OR 97239; and
| | - Eric S. Orwoll
- Endocrinology and Metabolism
- Portland VA Medical Center, Portland, OR 97239; and
| | - Beth Wilmot
- Public Health and Preventive Medicine, Oregon Health and Science University, Portland, OR 97239
| | - Shannon McWeeney
- Public Health and Preventive Medicine, Oregon Health and Science University, Portland, OR 97239
| | - Miguel A. Valverde
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David M. Cohen
- Department of Medicine, Divisions of Nephrology and Hypertension, and
- Portland VA Medical Center, Portland, OR 97239; and
| |
Collapse
|
563
|
Earley S, Pauyo T, Drapp R, Tavares MJ, Liedtke W, Brayden JE. TRPV4-dependent dilation of peripheral resistance arteries influences arterial pressure. Am J Physiol Heart Circ Physiol 2009; 297:H1096-102. [PMID: 19617407 DOI: 10.1152/ajpheart.00241.2009] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channels have been implicated as mediators of calcium influx in both endothelial and vascular smooth muscle cells and are potentially important modulators of vascular tone. However, very little is known about the functional roles of TRPV4 in the resistance vasculature or how these channels influence hemodynamic properties. In the present study, we examined arterial vasomotor activity in vitro and recorded blood pressure dynamics in vivo using TRPV4 knockout (KO) mice. Acetylcholine-induced hyperpolarization and vasodilation were reduced by approximately 75% in mesenteric resistance arteries from TRPV4 KO versus wild-type (WT) mice. Furthermore, 11,12-epoxyeicosatrienoic acid (EET), a putative endothelium-derived hyperpolarizing factor, activated a TRPV4-like cation current and hyperpolarized the membrane of vascular smooth muscle cells, resulting in the dilation of mesenteric arteries from WT mice. In contrast, 11,12-EET had no effect on membrane potential, diameter, or ionic currents in the mesenteric arteries from TRPV4 KO mice. A disruption of the endothelium reduced 11,12-EET-induced hyperpolarization and vasodilatation by approximately 50%. A similar inhibition of these responses was observed following the block of endothelial (small and intermediate conductance) or smooth muscle (large conductance) K(+) channels, suggesting a link between 11,12-EET activity, TRPV4, and K(+) channels in endothelial and smooth muscle cells. Finally, we found that hypertension induced by the inhibition of nitric oxide synthase was greater in TRPV4 KO compared with WT mice. These results support the conclusion that both endothelial and smooth muscle TRPV4 channels are critically involved in the vasodilation of mesenteric arteries in response to endothelial-derived factors and suggest that in vivo this mechanism opposes the effects of hypertensive stimuli.
Collapse
Affiliation(s)
- Scott Earley
- Department of Pharmacology, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | | | | | |
Collapse
|
564
|
Bishara NB, Triggle CR, Hill MA. Cytochrome P450 Products and Arachidonic Acid–Induced, Non–Store-Operated, Ca2+Entry in Cultured Bovine Endothelial Cells. ACTA ACUST UNITED AC 2009; 12:153-61. [PMID: 16162437 DOI: 10.1080/10623320500227036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Endothelial cells possess multiple mechanisms for the control of Ca2+ influx during agonist and mechanical stimulation. Increased intracellular Ca2+ during such events is important in the production of vasoactive substances including NO, prostacyclin, and, possibly, endothelium-derived hyperpolarizing factor(s). The present studies examined the effect of arachidonic acid on cellular Ca2+ entry and the underlying mechanisms by which this fatty acid regulates entry. Studies were conducted in cultured bovine aortic endothelial cells (passages 3 to 6) with changes in intracellular Ca2+ determined using the fluorescent Ca2+-sensitive indicator fura 2. Arachidonic acid (1 to 50 microM) stimulated Ca2+ entry from the superfusate without affecting Ca2+ release from intracellular stores. 2-aminoethoxydiphenyl borate (2APB) (100 microM) added at the peak of Ca2+ entry did not inhibit arachidonic acid-induced Ca2+ entry but, in contrast, significantly inhibited entry stimulated by ATP (1 microM). Arachidonic acid-induced Ca2+ entry was inhibited by econazole (1 microM), but not indomethacin (10 microM) or nordihydroguairetic acid (10 microM), suggesting the involvement of cytochrome P450 monooxygenase metabolite of arachidonic acid. Oleic acid (10 microM) was ineffective in inducing Ca2+ entry, whereas linoleic acid (10 microM) stimulated Ca2+ entry but by a mechanism insensitive to econazole. Collectively the data demonstrate that primary cultured aortic endothelial cells possess a Ca2+ entry mechanism modulated by arachidonic acid. This mode of Ca2+ entry appears to operate independently of store depletion-mediated mechanisms.
Collapse
Affiliation(s)
- Nour B Bishara
- Microvascular Biology Group, School of Medical Sciences, RMIT University, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
565
|
Vignali M, Benfenati V, Caprini M, Anderova M, Nobile M, Ferroni S. The endocannabinoid anandamide inhibits potassium conductance in rat cortical astrocytes. Glia 2009; 57:791-806. [PMID: 19031444 DOI: 10.1002/glia.20807] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Endocannabinoids are a family of endogenous signaling molecules that modulate neuronal excitability in the central nervous system (CNS) by interacting with cannabinoid (CB) receptors. In spite of the evidence that astroglial cells also possess CB receptors, there is no information on the role of endocannabinoids in regulating CNS function through the modulation of ion channel-mediated homeostatic mechanisms in astroglial cells. We provide electrophysiological evidence that the two brain endocannabinoids anandamide (AEA) and 2-arachidonylglycerol (2-AG) markedly depress outward conductance mediated by delayed outward rectifier potassium current (IK(DR)) in primary cultured rat cortical astrocytes. Pharmacological experiments suggest that the effect of AEA does not result from the activation of known CB receptors. Moreover, neither the production of AEA metabolites nor variations in free cytosolic calcium are involved in the negative modulation of IK(DR). We show that the action of AEA is mediated by its interaction with the extracellular leaflet of the plasma membrane. Similar experiments performed in situ in cortical slices indicate that AEA downregulates IK(DR) in complex and passive astroglial cells. Moreover, IK(DR) is also inhibited by AEA in NG2 glia. Collectively, these results support the notion that endocannabinoids may exert their modulation of CNS function via the regulation of homeostatic function of the astroglial syncytium mediated by ion channel activity.
Collapse
Affiliation(s)
- M Vignali
- Department of Human and General Physiology, University of Bologna, 40127 Bologna, Italy
| | | | | | | | | | | |
Collapse
|
566
|
Inoue R, Jian Z, Kawarabayashi Y. Mechanosensitive TRP channels in cardiovascular pathophysiology. Pharmacol Ther 2009; 123:371-85. [PMID: 19501617 DOI: 10.1016/j.pharmthera.2009.05.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 05/14/2009] [Indexed: 12/22/2022]
Abstract
Transient receptor potential (TRP) proteins constitute a large non-voltage-gated cation channel superfamily, activated polymodally by various physicochemical stimuli, and are implicated in a variety of cellular functions. Known activators for TRP include not only chemical stimuli such as receptor stimulation, increased acidity and pungent/cooling agents, but temperature change and various forms of mechanical stimuli such as osmotic stress, membrane stretch, and shear force. Recent investigations have revealed that at least ten mammalian TRPs exhibit mechanosensitivity (TRPC1, 5, 6; TRPV1, 2, 4; TRPM3, 7; TRPA1; TRPP2), but the mechanisms underlying it appear considerably divergent and complex. The proposed mechanisms are associated with lipid bilayer mechanics, specialized force-transducing structures, biochemical reactions, membrane trafficking and transcriptional regulation. Many of mechanosensitive (MS)-TRP channel likely undergo multiple regulations via these mechanisms. In the cardiovascular system in which hemodynamic forces constantly operate, the impact of mechanical stress may be particularly significant. Extensive morphological and functional studies have indicated that several MS-TRP channels are expressed in cardiac muscle, vascular smooth muscle, endothelium and vasosensory neurons, each differentially contributing to cardiovascular (CV) functions. To further complexity, the recent evidence suggests that mechanical stress may synergize with neurohormonal mechanisms thereby amplifying otherwise marginal responses. Furthermore, the currently available data suggest that MS-TRP channels may be involved in CV pathophysiology such as cardiac arrhythmia, cardiac hypertrophy/myopathy, hypertension and aneurysms. This review will overview currently known mechanisms for mechanical activation/modulation of TRPs and possible connections of MS-TRP channels to CV disorders.
Collapse
Affiliation(s)
- Ryuji Inoue
- Department of Physiology, Graduate School of Medical Sciences, Fukuoka University, Nanakuma 7-45-1, Jonan-ku, Fukuoka 814-0180, Japan.
| | | | | |
Collapse
|
567
|
Chen L, Liu C, Liu L. Changes in osmolality modulate voltage-gated sodium channels in trigeminal ganglion neurons. Neurosci Res 2009; 64:199-207. [PMID: 19428701 PMCID: PMC2684961 DOI: 10.1016/j.neures.2009.02.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2008] [Revised: 01/24/2009] [Accepted: 02/26/2009] [Indexed: 02/01/2023]
Abstract
Voltage-gated sodium channels (VGSCs) are important channels which participate in many physiological functions. Whether VGSCs can be modulated by changes in osmolality in trigeminal ganglion (TG) neurons remains unknown. In this study, by using whole-cell patch clamp techniques, we tested the effects of hypo- and hypertonicity on VGSCs in cultured TG neurons. Our data show that tetrodotoxin-resistant sodium current (TTX-R current) was inhibited in the presence of hypo- and hypertonic solutions. In hypertonic solutions both voltage-dependent activation and inactivation curves shifted to the hyperpolarizing direction, while in hypotonic solutions only inactivation curve shifted to the hyperpolarizing direction. Transient Receptor Potential Vanilloid 4 (TRPV4) receptor activator mimicked the inhibition of TTX-R current by hypotonicity and the inhibition by hypotonicity was markedly attenuated by TRPV4 receptor blocker and in TRPV4(-/-) mice TG neurons. We also demonstrate that the inhibition of PKA selectively attenuated hypotonicity-induced inhibition, whereas antagonism of PLC and PI3K selectively attenuated hypertonicity-induced inhibition. We conclude that although hypo- and hypertonicity have similar effect on VGSCs, receptor and intracellular signaling pathways are different for hypo- versus hypertonicity-induced inhibition of TTX-R current.
Collapse
Affiliation(s)
- Lei Chen
- Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Department of Neurobiology, Duke University, Durham, North Carolina, USA
- Department of Physiology, Nanjing Medical University, Nanjing, P.R. China
| | - Changjin Liu
- Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Lieju Liu
- Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Department of Anesthesiology, Duke University, Durham, North Carolina, USA
- Institution of nutrition, Wuhan Polytechnic University, Wuhan, P.R. China
| |
Collapse
|
568
|
Vriens J, Appendino G, Nilius B. Pharmacology of vanilloid transient receptor potential cation channels. Mol Pharmacol 2009; 75:1262-79. [PMID: 19297520 DOI: 10.1124/mol.109.055624] [Citation(s) in RCA: 313] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Depending on their primary structure, the 28 mammalian transient receptor potential (TRP) cation channels identified so far can be sorted into 6 subfamilies: TRPC ("Canonical"), TRPV ("Vanilloid"), TRPM ("Melastatin"), TRPP ("Polycystin"), TRPML ("Mucolipin"), and TRPA ("Ankyrin"). The TRPV subfamily (vanilloid receptors) comprises channels critically involved in nociception and thermosensing (TRPV1, TRPV2, TRPV3, and TRPV4), whereas TRPV5 and TRPV6 are involved in renal Ca(2+) absorption/reabsorption. Apart from TRPV1, the pharmacology of these channels is still insufficiently known. Furthermore, only few small-molecule ligands for non-TRPV1 vanilloid receptors have been identified, and little is known of their endogenous ligands, resulting in a substantial "orphan" state for these channels. In this review, we summarize the pharmacological properties of members of the TRPV subfamily, highlighting the critical issues and challenges facing their "deorphanization" and clinical exploitation.
Collapse
Affiliation(s)
- Joris Vriens
- Department of Molecular Cell Biology, Katholieke Universiteit Leuven, Belgium
| | | | | |
Collapse
|
569
|
Zhu G, ICGN Investigators, Gulsvik A, Bakke P, Ghatta S, Anderson W, Lomas DA, Silverman EK, Pillai SG. Association of TRPV4 gene polymorphisms with chronic obstructive pulmonary disease. Hum Mol Genet 2009; 18:2053-62. [PMID: 19279160 DOI: 10.1093/hmg/ddp111] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by airway epithelial damage, bronchoconstriction, parenchymal destruction and mucus hypersecretion. Upon activation by a broad range of stimuli, transient receptor potential vanilloid 4 (TRPV4) functions to control airway epithelial cell volume and epithelial and endothelial permeability; it also triggers bronchial smooth muscle contraction and participates in autoregulation of mucociliary transport. These functions of TRPV4 may be important for the regulation of COPD pathogenesis, so TRPV4 is a candidate gene for COPD. We genotyped 20 single nucleotide polymorphisms (SNPs) in TRPV4, and tested qualitative COPD and quantitative FEV(1) and FEV(1)/(F)VC phenotypes in two independent large populations. The family population had 606 pedigrees including 1891 individuals, and the case-control sample included 953 COPD cases and 956 controls. Family-based association tests were performed in the family data. Logistic regression and linear models were used in the case-control data to replicate the association results. In the family data, seven out of 20 SNPs tested were associated with COPD (2.5 x 10(-4) < or = P < or = 0.04) and six SNPs were associated with FEV(1)/VC (0.02 < or = P < or = 0.03) from family-based association tests (PBAT) analysis. Four out of the seven SNPs associated with COPD demonstrated replicated associations with the same effect directions in the case-control population (0.02 < or = P < or = 0.03). Significant haplotype associations supported the results of single SNP analyses. Thus, polymorphisms in the TRPV4 gene are associated with COPD.
Collapse
Affiliation(s)
- Guohua Zhu
- Genetics, GlaxoSmithKline R&D, 5 Moore Drive, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | | | | | | | |
Collapse
Collaborators
Alvar Agusti, Peter M A Calverley, Claudio F Donner, Robert D Levy, Barry J Make, Peter D Paré, Stephen I Rennard, Jørgen Vestbo, Emiel F M Wouters,
Collapse
|
570
|
Buczynski MW, Dumlao DS, Dennis EA. Thematic Review Series: Proteomics. An integrated omics analysis of eicosanoid biology. J Lipid Res 2009; 50:1015-38. [PMID: 19244215 PMCID: PMC2681385 DOI: 10.1194/jlr.r900004-jlr200] [Citation(s) in RCA: 418] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 02/23/2009] [Indexed: 11/20/2022] Open
Abstract
Eicosanoids have been implicated in a vast number of devastating inflammatory conditions, including arthritis, atherosclerosis, pain, and cancer. Currently, over a hundred different eicosanoids have been identified, with many having potent bioactive signaling capacity. These lipid metabolites are synthesized de novo by at least 50 unique enzymes, many of which have been cloned and characterized. Due to the extensive characterization of eicosanoid biosynthetic pathways, this field provides a unique framework for integrating genomics, proteomics, and metabolomics toward the investigation of disease pathology. To facilitate a concerted systems biology approach, this review outlines the proteins implicated in eicosanoid biosynthesis and signaling in human, mouse, and rat. Applications of the extensive genomic and lipidomic research to date illustrate the questions in eicosanoid signaling that could be uniquely addressed by a thorough analysis of the entire eicosanoid proteome.
Collapse
Affiliation(s)
| | | | - Edward A. Dennis
- Department of Chemistry and Biochemistry, Department of Pharmacology, and School of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
571
|
Su Z, Zhou X, Loukin SH, Haynes WJ, Saimi Y, Kung C. The use of yeast to understand TRP-channel mechanosensitivity. Pflugers Arch 2009; 458:861-7. [PMID: 19462180 DOI: 10.1007/s00424-009-0680-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Accepted: 04/30/2009] [Indexed: 01/04/2023]
Abstract
Mechanosensitive (MS) ion channels likely underlie myriad force-sensing processes, from basic osmotic regulation to specified sensations of animal hearing and touch. Albeit important, the molecular identities of many eukaryotic MS channels remain elusive, let alone their working mechanisms. This is in stark contrast to our advanced knowledge on voltage- or ligand-sensitive channels. Several members of transient receptor potential (TRP) ion channel family have been implicated to function in mechanosensation and are recognized as promising candidate MS channels. The yeast TRP homolog, TRPY1, is clearly a first-line force transducer. It can be activated by hypertonic shock in vivo and by membrane stretch force in excised patches under patch clamp, making it a useful model for understanding TRP channel mechanosensitivity in general. TRPY1 offers two additional research advantages: (1) It has a large ( approximately 300 pS) unitary conductance and therefore a favorable S/N ratio. (2) Budding yeast allows convenient and efficient genetic and molecular manipulations. In this review, we focus on the current research of TRPY1 and discuss its prospect. We also describe the use of yeast as a system to express and characterize animal TRP channels.
Collapse
Affiliation(s)
- Zhenwei Su
- Laboratory of Molecular Biology, 305 R.M. Bock Laboratories, 1525 Linden Drive, Madison, WI 53706, USA.
| | | | | | | | | | | |
Collapse
|
572
|
Inoue R, Jensen LJ, Jian Z, Shi J, Hai L, Lurie AI, Henriksen FH, Salomonsson M, Morita H, Kawarabayashi Y, Mori M, Mori Y, Ito Y. Synergistic activation of vascular TRPC6 channel by receptor and mechanical stimulation via phospholipase C/diacylglycerol and phospholipase A2/omega-hydroxylase/20-HETE pathways. Circ Res 2009; 104:1399-409. [PMID: 19443836 DOI: 10.1161/circresaha.108.193227] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
TRPC6 is a non-voltage-gated Ca(2+) entry/depolarization channel associated with vascular tone regulation and remodeling. Expressed TRPC6 channel responds to both neurohormonal and mechanical stimuli, the mechanism for which remains controversial. In this study, we examined the possible interactions of receptor and mechanical stimulations in activating this channel using the patch clamp technique. In HEK293 cells expressing TRPC6, application of mechanical stimuli (hypotonicity, shear, 2,4,6-trinitrophenol) caused, albeit not effective by themselves, a prominent potentiation of cationic currents (I(TRPC6)) induced by a muscarinic receptor agonist carbachol. This effect was insensitive to a tarantula toxin GsMTx-4 (5 mumol/L). A similar extent of mechanical potentiation was observed after activation of I(TRPC6) by GTPgammaS or a diacylglycerol analog 1-oleoyl-2-acetyl-sn-glycerol (OAG). Single TRPC6 channel activity evoked by carbachol was also enhanced by a negative pressure added in the patch pipette. Mechanical potentiation of carbachol- or OAG-induced I(TRPC6) was abolished by small interfering RNA knockdown of cytosolic phospholipase A(2) or pharmacological inhibition of omega-hydroxylation of arachidonic acid into 20-HETE (20-hydroxyeicosatetraenoic acid). Conversely, direct application of 20-HETE enhanced both OAG-induced macroscopic and single channel TRPC6 currents. Essentially the same results were obtained for TRPC6-like cation channel in A7r5 myocytes, where its activation by noradrenaline or Arg8 vasopressin was greatly enhanced by mechanical stimuli via 20-HETE production. Furthermore, myogenic response of pressurized mesenteric artery was significantly enhanced by weak receptor stimulation dependently on 20-HETE production. These results collectively suggest that simultaneous operation of receptor and mechanical stimulations may synergistically amplify transmembrane Ca(2+) mobilization through TRPC6 activation, thereby enhancing the vascular tone via phospholipase C/diacylglycerol and phospholipase A(2)/omega-hydroxylase/20-HETE pathways.
Collapse
Affiliation(s)
- Ryuji Inoue
- Department of Physiology, Graduate School of Medical Sciences, Fukuoka University, Nanakuma 7-45-1, Jonan-ku, Fukuoka 814-0180, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
573
|
Potenzieri C, Brink TS, Simone DA. Excitation of cutaneous C nociceptors by intraplantar administration of anandamide. Brain Res 2009; 1268:38-47. [PMID: 19285051 PMCID: PMC2749687 DOI: 10.1016/j.brainres.2009.02.061] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Revised: 02/16/2009] [Accepted: 02/17/2009] [Indexed: 11/20/2022]
Abstract
Anandamide has been characterized as both an endocannabinoid and endovanilloid. Consistent with its actions as an endovanilloid, previous studies have demonstrated that anandamide can excite primary sensory neurons in vitro via transient receptor potential vanilloid type one (TRPV1) receptors. In the present study, we sought to determine if anandamide excited cutaneous C nociceptors in vivo and if this excitation correlated with nocifensive behaviors. Using teased-fiber electrophysiological methods in the rat, C nociceptors isolated from the tibial nerve with receptive fields (RFs) on the plantar surface of the hindpaw were studied. Injection of anandamide into the RF dose-dependently excited nociceptors at doses of 10 and 100 microg. The TRPV1 receptor antagonists, capsazepine or SB 366791, were applied to the RF to determine if excitation by anandamide was mediated through TRPV1 receptors. Intraplantar injection of either capsazepine (10 microg) or SB 366791 (3 microg) attenuated the excitation produced by 100 microg anandamide. We also determined whether excitation of C nociceptors by anandamide was associated with nocifensive behaviors. Intraplantar injection of 100 microg anandamide produced nocifensive behaviors that were attenuated by pre-treatment with either capsazepine or SB 366791. Furthermore, we determined if intraplantar injection of anandamide altered withdrawal responses to radiant heat. Neither intraplantar injection of anandamide nor vehicle produced antinociception or hyperalgesia to radiant heat. Our results indicate that anandamide excited cutaneous C nociceptors and produced nocifensive behaviors via activation of TRPV1 receptors.
Collapse
Affiliation(s)
- Carl Potenzieri
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN, USA; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Thaddeus S Brink
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN, USA; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
574
|
van Rossum DB, Patterson RL. PKC and PLA2: probing the complexities of the calcium network. Cell Calcium 2009; 45:535-45. [PMID: 19345415 DOI: 10.1016/j.ceca.2009.02.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 02/24/2009] [Accepted: 02/26/2009] [Indexed: 11/15/2022]
Abstract
Lipid signaling and phosphorylation cascades are fundamental to calcium signaling networks. In this review, we will discuss the recent laboratory findings for the phospholipase A(2) (PLA(2))/protein kinase C (PKC) pathway within cellular calcium networks. The complexity and connectivity of these ubiquitous cellular signals make interpretation of experimental results extremely challenging. We present here computational methods which have been developed to conquer such complex data, and how they can be used to make models capable of accurately predicting cellular responses within multiple calcium signaling pathways. We propose that information obtained from network analysis and computational techniques provides a rich source of knowledge which can be directly translated to the laboratory benchtop.
Collapse
Affiliation(s)
- Damian B van Rossum
- Department of Biology, The Pennsylvania State University, PA, United States.
| | | |
Collapse
|
575
|
Sudhahar V, Shaw S, Imig JD. Mechanisms involved in oleamide-induced vasorelaxation in rat mesenteric resistance arteries. Eur J Pharmacol 2009; 607:143-50. [PMID: 19326479 PMCID: PMC2664517 DOI: 10.1016/j.ejphar.2009.02.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Fatty acid amides are a new class of signaling lipids that have been implicated in diverse physiological and pathological conditions. Oleamide is a fatty acid amide that induces vasorelaxation. Here, we investigated the mechanisms behind the vasorelaxation effect of oleamide in rat mesenteric resistance arteries. Oleamide-induced concentration dependent (0.01 microM-10 microM) vasorelaxation in mesenteric resistance arteries. This relaxation was unaffected by the presence of the fatty acid amide hydrolase (FAAH) inhibitors. The cannabinoid type 1 (CB1) receptor antagonist, AM251 and the non-CB1/CB2 cannabinoid receptor antagonist, O-1918, attenuated the oleamide vasodilatory response, however the cannabinoid CB2 receptor antagonist, AM630, did not affect the vascular response. Moreover, inhibition of the transient receptor potential vanilloid (TRPV) 1 receptor with capsazepine shifted the oleamide-induced vasorelaxation response to the right. In agreement with the vascular functional data, the cannabinoid CB1 and TRPV1 receptor proteins were expressed in mesenteric resistance arteries but cannabinoid CB2 receptors and the FAAH enzyme were not. In endothelium-denuded arteries, the oleamide-mediated vasorelaxation was attenuated and cannabinoid CB1 or non-CB1/CB2 cannabinoid receptor blockade did not further reduce the dilatory response whereas TRPV1 antagonism further decreased the response. These findings indicate that cannabinoid receptors on the endothelium and endothelium-independent TRPV1 receptors contribute to the oleamide vasodilatory response. Taken together, these results demonstrate that the oleamide-induced vasorelaxation is mediated, in part, by cannabinoid CB1 receptors, non-CB1/CB2 cannabinoid receptors, and TRPV1 receptors in rat mesenteric resistance arteries. These mechanisms are overlapping in respect to oleamide-induced mesenteric resistance artery dilation.
Collapse
MESH Headings
- Animals
- Dose-Response Relationship, Drug
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Hypnotics and Sedatives/administration & dosage
- Hypnotics and Sedatives/pharmacology
- Male
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/metabolism
- Oleic Acids/administration & dosage
- Oleic Acids/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, Cannabinoid/drug effects
- Receptors, Cannabinoid/metabolism
- TRPV Cation Channels/drug effects
- TRPV Cation Channels/metabolism
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Varadarajan Sudhahar
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Sean Shaw
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - John D. Imig
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI
- Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
576
|
Cioffi DL, Lowe K, Alvarez DF, Barry C, Stevens T. TRPing on the lung endothelium: calcium channels that regulate barrier function. Antioxid Redox Signal 2009; 11:765-76. [PMID: 18783312 PMCID: PMC2850299 DOI: 10.1089/ars.2008.2221] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rises in cytosolic calcium are sufficient to initiate the retraction of endothelial cell borders and to increase macromolecular permeability. Although endothelial cell biologists have recognized the importance of shifts in cytosolic calcium for several decades, only recently have we gained a rudimentary understanding of the membrane calcium channels that change cell shape. Members of the transient receptor potential family (TRP) are chief among the molecular candidates for permeability-coupled calcium channels. Activation of calcium entry through store-operated calcium entry channels, most notably TRPC1 and TRPC4, increases lung endothelial cell permeability, as does activation of calcium entry through the TRPV4 channel. However, TRPC1 and TRPC4 channels appear to influence the lung extraalveolar endothelial barrier most prominently, whereas TRPV4 channels appear to influence the lung capillary endothelial barrier most prominently. Thus, phenotypic heterogeneity in ion channel expression and function exists within the lung endothelium, along the arterial-capillary-venous axis, and is coupled to discrete control of endothelial barrier function.
Collapse
Affiliation(s)
- Donna L Cioffi
- Center for Lung Biology, University of South Alabama, Mobile, Alabama 36688, USA
| | | | | | | | | |
Collapse
|
577
|
Christianson JA, Bielefeldt K, Altier C, Cenac N, Davis BM, Gebhart GF, High KW, Kollarik M, Randich A, Undem B, Vergnolle N. Development, plasticity and modulation of visceral afferents. BRAIN RESEARCH REVIEWS 2009; 60:171-86. [PMID: 19150371 PMCID: PMC2841801 DOI: 10.1016/j.brainresrev.2008.12.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 12/29/2008] [Indexed: 12/25/2022]
Abstract
Visceral pain is the most common reason for doctor visits in the US. Like somatic pain, virtually all visceral pain sensations begin with the activation of primary sensory neurons innervating the viscera and/or the blood vessels associated with these structures. Visceral afferents also play a central role in tissue homeostasis. Recent studies show that in addition to monitoring the state of the viscera, they perform efferent functions through the release of small molecules (e.g. peptides like CGRP) that can drive inflammation, thereby contributing to the development of visceral pathologies (e.g. diabetes Razavi, R., Chan, Y., Afifiyan, F.N., Liu, X.J., Wan, X., Yantha, J., Tsui, H., Tang, L., Tsai, S., Santamaria, P., Driver, J.P., Serreze, D., Salter, M.W., Dosch, H.M., 2006. TRPV1+ sensory neurons control beta cell stress and islet inflammation in autoimmune diabetes, Cell 127 1123-1135). Visceral afferents are heterogeneous with respect to their anatomy, neurochemistry and function. They are also highly plastic in that their cellular environment continuously influences their response properties. This plasticity makes them susceptible to long-term changes that may contribute significantly to the development of persistent pain states such as those associated with irritable bowel syndrome, pancreatitis, and visceral cancers. This review examines recent insights into visceral afferent anatomy and neurochemistry and how neonatal insults can affect the function of these neurons in the adult. New approaches to the treatment of visceral pain, which focus on primary afferents, will also be discussed.
Collapse
Affiliation(s)
- Julie A. Christianson
- University of Pittsburgh School of Medicine, Pittsburgh Center for Pain Research, 200 Lothrop St., Pittsburgh, PA 16261, USA
| | - Klaus Bielefeldt
- University of Pittsburgh School of Medicine, Pittsburgh Center for Pain Research, 200 Lothrop St., Pittsburgh, PA 16261, USA
| | - Christophe Altier
- University of Calgary, Department of Pharmacology and Therapeutics, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Nicolas Cenac
- University of Calgary, Department of Pharmacology and Therapeutics, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Brian M. Davis
- University of Pittsburgh School of Medicine, Pittsburgh Center for Pain Research, 200 Lothrop St., Pittsburgh, PA 16261, USA
| | - Gerald F. Gebhart
- University of Pittsburgh School of Medicine, Pittsburgh Center for Pain Research, 200 Lothrop St., Pittsburgh, PA 16261, USA
| | - Karin W. High
- Department of Physiology, MS-609 Chandler Medical Center, 800 Rose St., University of Kentucky, Lexington, KY 40536-0298, USA
| | - Marian Kollarik
- Johns Hopkins School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | - Alan Randich
- University of Alabama, Birmingham, Department of Psychology, 1300 University Blvd., Birmingham, AL 35294-1170, USA
| | - Brad Undem
- Department of Physiology, MS-609 Chandler Medical Center, 800 Rose St., University of Kentucky, Lexington, KY 40536-0298, USA
| | - Nathalie Vergnolle
- University of Calgary, Department of Pharmacology and Therapeutics, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| |
Collapse
|
578
|
Kauer JA, Gibson HE. Hot flash: TRPV channels in the brain. Trends Neurosci 2009; 32:215-24. [PMID: 19285736 DOI: 10.1016/j.tins.2008.12.006] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Revised: 12/08/2008] [Accepted: 12/09/2008] [Indexed: 01/27/2023]
Abstract
TRPV1 (transient receptor potential, vanilloid) channels belong to a family of ligand-gated ion channels gated not only by the binding of certain lipophilic molecules but also by extracellular protons and physical stimuli such as heat or osmotic pressure changes. These nonselective cation channels are permeable to Na(+) and K(+) and are also very Ca(2+) permeable; in fact, TRPV1 is as Ca(2+) permeable as the NMDA receptor channel and can, thus, act as a trigger for Ca(2+)-mediated cell signaling. Although these channels are highly expressed in primary sensory afferents, accumulating evidence indicates that TRPV family channels are also present in the brain. Here, we review evidence that TRPV channels in the central nervous system might contribute to many basic neuronal functions including resting membrane potential, neurotransmitter release and synaptic plasticity.
Collapse
Affiliation(s)
- Julie A Kauer
- Brown University, Department of Molecular Pharmacology, Physiology and Biotechnology, Providence, RI 02912, USA.
| | | |
Collapse
|
579
|
Zhang DX, Mendoza SA, Bubolz AH, Mizuno A, Ge ZD, Li R, Warltier DC, Suzuki M, Gutterman DD. Transient receptor potential vanilloid type 4-deficient mice exhibit impaired endothelium-dependent relaxation induced by acetylcholine in vitro and in vivo. Hypertension 2009; 53:532-8. [PMID: 19188524 PMCID: PMC2694062 DOI: 10.1161/hypertensionaha.108.127100] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2008] [Revised: 12/22/2008] [Accepted: 01/06/2009] [Indexed: 11/16/2022]
Abstract
Agonist-induced Ca2+ entry is important for the synthesis and release of vasoactive factors in endothelial cells. The transient receptor potential vanilloid type 4 (TRPV4) channel, a Ca2+-permeant cation channel, is expressed in endothelial cells and involved in the regulation of vascular tone. Here we investigated the role of TRPV4 channels in acetylcholine-induced vasodilation in vitro and in vivo using the TRPV4 knockout mouse model. The expression of TRPV4 mRNA and protein was detected in both conduit and resistance arteries from wild-type mice. In small mesenteric arteries from wild-type mice, the TRPV4 activator 4alpha-phorbol-12,13-didecanoate increased endothelial [Ca2+]i in situ, which was reversed by the TRPV4 blocker ruthenium red. In wild-type animals, acetylcholine dilated small mesenteric arteries that involved both NO and endothelium-derived hyperpolarizing factors. In TRPV4-deficient mice, the NO component of the relaxation was attenuated and the endothelium-derived hyperpolarizing factor component was largely eliminated. Compared with their wild-type littermates, TRPV4-deficient mice demonstrated a blunted endothelial Ca2+ response to acetylcholine in mesenteric arteries and reduced NO release in carotid arteries. Acetylcholine (5 mg/kg, IV) decreased blood pressure by 37.0+/-6.2 mm Hg in wild-type animals but only 16.6+/-2.7 mm Hg in knockout mice. We conclude that acetylcholine-induced endothelium-dependent vasodilation is reduced both in vitro and in vivo in TRPV4 knockout mice. These findings may provide novel insight into mechanisms of Ca2+ entry evoked by chemical agonists in endothelial cells.
Collapse
Affiliation(s)
- David X Zhang
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
580
|
Iliff JJ, Alkayed NJ. Soluble Epoxide Hydrolase Inhibition: Targeting Multiple Mechanisms of Ischemic Brain Injury with a Single Agent. FUTURE NEUROLOGY 2009; 4:179-199. [PMID: 19779591 DOI: 10.2217/14796708.4.2.179] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Soluble epoxide hydrolase (sEH) is a key enzyme in the metabolic conversion and degradation of P450 eicosanoids called epoxyeicosatrienoic acids (EETs). Genetic variations in the sEH gene, designated EPHX2, are associated with ischemic stroke risk. In experimental studies, sEH inhibition and gene deletion reduce infarct size after focal cerebral ischemia in mice. Although the precise mechanism of protection afforded by sEH inhibition remains under investigation, EETs exhibit a wide array of potentially beneficial actions in stroke, including vasodilation, neuroprotection, promotion of angiogenesis and suppression of platelet aggregation, oxidative stress and post-ischemic inflammation. Herein we argue that by capitalizing on this broad protective profile, sEH inhibition represents a prototype "combination therapy" targeting multiple mechanisms of stroke injury with a single agent.
Collapse
Affiliation(s)
- Jeffrey J Iliff
- Department of Anesthesiology and Peri-Operative Medicine, Oregon Health and Science University, Portland OR 97239
| | | |
Collapse
|
581
|
Functional interaction of the cation channel transient receptor potential vanilloid 4 (TRPV4) and actin in volume regulation. Eur J Cell Biol 2009; 88:141-52. [DOI: 10.1016/j.ejcb.2008.10.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 10/03/2008] [Accepted: 10/06/2008] [Indexed: 11/19/2022] Open
|
582
|
Hoffmann EK, Lambert IH, Pedersen SF. Physiology of cell volume regulation in vertebrates. Physiol Rev 2009; 89:193-277. [PMID: 19126758 DOI: 10.1152/physrev.00037.2007] [Citation(s) in RCA: 1061] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The ability to control cell volume is pivotal for cell function. Cell volume perturbation elicits a wide array of signaling events, leading to protective (e.g., cytoskeletal rearrangement) and adaptive (e.g., altered expression of osmolyte transporters and heat shock proteins) measures and, in most cases, activation of volume regulatory osmolyte transport. After acute swelling, cell volume is regulated by the process of regulatory volume decrease (RVD), which involves the activation of KCl cotransport and of channels mediating K(+), Cl(-), and taurine efflux. Conversely, after acute shrinkage, cell volume is regulated by the process of regulatory volume increase (RVI), which is mediated primarily by Na(+)/H(+) exchange, Na(+)-K(+)-2Cl(-) cotransport, and Na(+) channels. Here, we review in detail the current knowledge regarding the molecular identity of these transport pathways and their regulation by, e.g., membrane deformation, ionic strength, Ca(2+), protein kinases and phosphatases, cytoskeletal elements, GTP binding proteins, lipid mediators, and reactive oxygen species, upon changes in cell volume. We also discuss the nature of the upstream elements in volume sensing in vertebrate organisms. Importantly, cell volume impacts on a wide array of physiological processes, including transepithelial transport; cell migration, proliferation, and death; and changes in cell volume function as specific signals regulating these processes. A discussion of this issue concludes the review.
Collapse
Affiliation(s)
- Else K Hoffmann
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | | | | |
Collapse
|
583
|
Loukin SH, Su Z, Kung C. Hypotonic shocks activate rat TRPV4 in yeast in the absence of polyunsaturated fatty acids. FEBS Lett 2009; 583:754-8. [PMID: 19174160 PMCID: PMC2825150 DOI: 10.1016/j.febslet.2009.01.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 01/14/2009] [Accepted: 01/16/2009] [Indexed: 01/23/2023]
Abstract
Transient-receptor-potential channels (TRPs) underlie the sensing of chemicals, heat, and mechanical force. We expressed the rat TRPV1 and TRPV4 subtypes in yeast and monitored their activities in vivo as Ca(2+) rise using transgenic aequorin. Heat and capsaicin activate TRPV1 but not TRPV4 in yeast. Hypotonic shocks activate TRPV4 but not TRPV1. Osmotic swelling is modeled to activate enzyme(s), producing polyunsaturated fatty acids (PUFAs) to open TRPV4 in mammalian cells. This model relegates mechanosensitivity to the enzyme and not the channel. Yeast has only a single Delta9 fatty-acid monodesaturase and cannot make PUFAs suggesting an alternative mechanism for TRPV4 activation. We discuss possible explanations of this difference.
Collapse
Affiliation(s)
- Stephen H Loukin
- Laboratory of Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive Madison, WI 53706, USA
| | | | | |
Collapse
|
584
|
Cao DS, Yu SQ, Premkumar LS. Modulation of transient receptor potential Vanilloid 4-mediated membrane currents and synaptic transmission by protein kinase C. Mol Pain 2009; 5:5. [PMID: 19208258 PMCID: PMC2650694 DOI: 10.1186/1744-8069-5-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Accepted: 02/10/2009] [Indexed: 11/10/2022] Open
Abstract
Background Transient receptor potential Vanilloid (TRPV) receptors are involved in nociception and are expressed predominantly in sensory neurons. TRPV1, a non-selective cation channel has been extensively studied and is responsible for inflammatory thermal hypersensitivity. In this study, the expression and function of TRPV4 have been characterized and compared with those of TRPV1. Results Immunohistochemical studies revealed that both TRPV1 and TRPV4 were co-expressed in dorsal root ganglion (DRG) neuronal cell bodies and in the central terminals of laminae I and II of the spinal dorsal horn (DH). In Ca2+ fluorescence imaging and whole-cell patch-clamp experiments, TRPV1- and TRPV4-mediated responses were observed in a population of the same DRG neurons. Sensitization of TRPV1 has been shown to be involved in inflammatory pain conditions. Incubation with phorbol 12, 13-dibutyrate (PDBu), a PKC activator, resulted in a significant potentiation of TRPV4 currents in DRG neurons. In TRPV4 expressing HEK 293T cells, PDBu increased 4α-phorbol 12, 13-didecanoate (4α-PDD)-induced single-channel activity in cell-attached patches, which was abrogated by bisindolylmaleimide (BIM), a selective PKC inhibitor. TRPV4 is also expressed at the central terminals of sensory neurons. Activation of TRPV4 by 4α-PDD increased the frequency of miniature excitatory post synaptic currents (mEPSCs) in DRG-DH neuronal co-cultures. 4α-PDD-induced increase in the frequency of mEPSCs was further enhanced by PDBu. The expression of TRP channels has been shown in other areas of the CNS; application of 4α-PDD significantly increased the mEPSC frequency in cultured hippocampal neurons, which was further potentiated by PDBu, whereas, TRPV1 agonist capsaicin did not modulate synaptic transmission. Conclusion These results indicate that TRPV4 and TRPV1 are co-expressed in certain DRG neurons and TRPV4 can be sensitized by PKC not only in DRG neuronal cell bodies, but also in the central sensory and non-sensory nerve terminals. Co-expression of TRPV1 and TRPV4 ion channels, their modulation of synaptic transmission and their sensitization by PKC may synergistically play a role in nociception.
Collapse
Affiliation(s)
- De-Shou Cao
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | | | | |
Collapse
|
585
|
Gao F, Sui D, Garavito RM, Worden RM, Wang DH. Salt intake augments hypotensive effects of transient receptor potential vanilloid 4: functional significance and implication. Hypertension 2009; 53:228-35. [PMID: 19075100 PMCID: PMC2729143 DOI: 10.1161/hypertensionaha.108.117499] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Accepted: 11/18/2008] [Indexed: 11/16/2022]
Abstract
To test the hypothesis that activation of the transient receptor potential vanilloid 4 (TRPV4) channel conveys a hypotensive effect that is enhanced during salt load, male Wistar rats fed a normal-sodium (0.5%) or high-sodium (HS; 4%) diet for 3 weeks were given 4 alpha-phorbol 12,13-didecanoate (4 alpha-PDD), a specific TRPV4 activator, in the presence or absence of capsazepine, a selective TRPV1 blocker, ruthenium red, a TRPV4 blocker, or TRPV4 small hairpin RNA that selectively knockdowns TRPV4. 4 alpha-PDD (1, 2.5, or 5 mg/kg IV) dose-dependently decreased mean arterial pressure (P<0.05). HS enhanced 4 alpha-PDD-induced depressor effects as well as 4 alpha-PDD-mediated release of calcitonin gene-related peptide and substance P (P<0.001). Ruthenium red markedly blunted (P<0.001), whereas capsazepine slightly attenuated (P<0.05) 4 alpha-PDD-induced depressor effects in HS and normal-sodium diet rats. Ruthenium red alone increased baseline mean arterial pressure in both HS and normal-sodium diet rats with a greater magnitude in the former (P<0.05). Western blot analysis showed that HS increased TRPV4 expression in dorsal root ganglia and mesenteric arteries (P<0.05) but not the renal cortex and medulla. Gene-silencing approach revealed that TRPV4 small hairpin RNA downregulated TRPV4 expression leading to blunted 4 alpha-PDD-induced hypotension (P<0.05). Thus, TRPV4 activation decreases blood pressure in rats given a normal-sodium diet. HS enhances TRPV4 expression in sensory nerves/mesenteric arteries and TRPV4-mediated depressor effects and calcitonin gene-related peptide/substance P release such that HS causes a greater increase in blood pressure when TRPV4 is blocked. Our data indicate that TRPV4 activation may constitute a compensatory mechanism in preventing salt-induced increases in blood pressure.
Collapse
Affiliation(s)
- Feng Gao
- Department of Medicine, Michigan State University
| | - Dexin Sui
- Department of Biochemistry and Molecular Biology, Michigan State University
| | | | - R. Mark Worden
- Department of Chemical Engineering and Materials Science, Michigan State University
| | - Donna H. Wang
- Department of Medicine, Michigan State University
- Neuroscience Program, Michigan State University
- Cell & Molecular Biology Program, Michigan State University
| |
Collapse
|
586
|
Akopian AN, Ruparel NB, Jeske NA, Patwardhan A, Hargreaves KM. Role of ionotropic cannabinoid receptors in peripheral antinociception and antihyperalgesia. Trends Pharmacol Sci 2009; 30:79-84. [PMID: 19070372 PMCID: PMC2863326 DOI: 10.1016/j.tips.2008.10.008] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 10/23/2008] [Accepted: 10/29/2008] [Indexed: 12/17/2022]
Abstract
Despite the wealth of information on cannabinoid-induced peripheral antihyperalgesic and antinociceptive effects in many pain models, the molecular mechanism(s) for these actions remains unknown. Although metabotropic cannabinoid receptors have important roles in many pharmacological actions of cannabinoids, recent studies have led to the recognition of a family of at least five ionotropic cannabinoid receptors (ICRs). The known ICRs are members of the family of transient receptor potential (TRP) channels and include TRPV1, TRPV2, TRPV4, TRPM8 and TRPA1. Cannabinoid activation of ICRs can result in desensitization of the TRPA1 and TRPV1 channel activities, inhibition of nociceptors and antihyperalgesia and antinociception in certain pain models. Thus, cannabinoids activate both metabotropic and ionotropic mechanisms to produce peripheral analgesic effects. Here, we provide an overview of the pharmacology of TRP channels as ICRs.
Collapse
Affiliation(s)
- Armen N Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio, TX 78229, USA.
| | | | | | | | | |
Collapse
|
587
|
Watanabe H, Murakami M, Ohba T, Ono K, Ito H. The Pathological Role of Transient Receptor Potential Channels in Heart Disease. Circ J 2009; 73:419-27. [DOI: 10.1253/circj.cj-08-1153] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Hiroyuki Watanabe
- Second Department of Internal Medicine, Akita University School of Medicine
| | - Manabu Murakami
- Department of Physiology, Akita University School of Medicine
| | - Takayoshi Ohba
- Department of Physiology, Akita University School of Medicine
| | - Kyoichi Ono
- Department of Physiology, Akita University School of Medicine
| | - Hiroshi Ito
- Second Department of Internal Medicine, Akita University School of Medicine
| |
Collapse
|
588
|
Godlewski G, Offertáler L, Osei-Hyiaman D, Mo FM, Harvey-White J, Liu J, Davis MI, Zhang L, Razdan RK, Milman G, Pacher P, Mukhopadhyay P, Lovinger DM, Kunos G. The endogenous brain constituent N-arachidonoyl L-serine is an activator of large conductance Ca2+-activated K+ channels. J Pharmacol Exp Ther 2009; 328:351-61. [PMID: 18923087 PMCID: PMC2605781 DOI: 10.1124/jpet.108.144717] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Accepted: 10/07/2008] [Indexed: 12/20/2022] Open
Abstract
The novel endocannabinoid-like lipid N-arachidonoyl L-serine (ARA-S) causes vasodilation through both endothelium-dependent and -independent mechanisms. We have analyzed the vasorelaxant effect of ARA-S in isolated vascular preparations and its effects on Ca(2+)-activated K(+) currents in human embryonic kidney cells stably transfected with the alpha-subunit of the human, large conductance Ca(+)-activated K(+) (BK(Ca)) channel [human embryonic kidney (HEK) 293hSlo cells]. ARA-S caused relaxation of rat isolated, intact and denuded, small mesenteric arteries preconstricted with (R)-(-)-1-(3-hydroxyphenyl)-2-methylaminoethanol hydrochloride (pEC(50), 5.49 and 5.14, respectively), whereas it caused further contraction of vessels preconstricted with KCl (pEC(50), 5.48 and 4.82, respectively). Vasorelaxation by ARA-S was inhibited by 100 nM iberiotoxin. In human embryonic kidney cells stably transfected with the alpha-subunit of the human BK(Ca) channel cells, ARA-S and its enantiomer, N-arachidonoyl-D-serine, enhanced the whole-cell outward K(+) current with similar potency (pEC(50), 5.63 and 5.32, respectively). The potentiation was not altered by the beta(1) subunit or mediated by ARA-S metabolites, stimulation of known cannabinoid receptors, G proteins, protein kinases, or Ca(2+)-dependent processes; it was lost after patch excision or after membrane cholesterol depletion but was restored after cholesterol reconstitution. BK(Ca) currents were also enhanced by N-arachidonoyl ethanolamide (pEC(50), 5.27) but inhibited by another endocannabinoid, O-arachidonoyl ethanolamine (pIC(50), 6.35), or by the synthetic cannabinoid O-1918 [(-)-1,3-dimethoxy-2-(3-3,4-trans-p-menthadien-(1,8)-yl)-orcinol] (pIC(50), 6.59), which blocks ARA-S-induced vasodilation. We conclude the following. 1) ARA-S directly activates BK(Ca) channels. 2) This interaction does not involve cannabinoid receptors or cytosolic factors but is dependent on the presence of membrane cholesterol. 3) Direct BK(Ca) channel activation probably contributes to the endothelium-independent component of ARA-S-induced mesenteric vasorelaxation. 4) O-1918 is a BK(Ca) channel inhibitor.
Collapse
Affiliation(s)
- Grzegorz Godlewski
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Bethesda, MD 20892-9413, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
589
|
Wagner TFJ, Loch S, Lambert S, Straub I, Mannebach S, Mathar I, Düfer M, Lis A, Flockerzi V, Philipp SE, Oberwinkler J. Transient receptor potential M3 channels are ionotropic steroid receptors in pancreatic beta cells. Nat Cell Biol 2008; 10:1421-30. [PMID: 18978782 DOI: 10.1038/ncb1801] [Citation(s) in RCA: 297] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Accepted: 10/03/2008] [Indexed: 11/09/2022]
Abstract
Transient receptor potential (TRP) cation channels are renowned for their ability to sense diverse chemical stimuli. Still, for many members of this large and heterogeneous protein family it is unclear how their activity is regulated and whether they are influenced by endogenous substances. On the other hand, steroidal compounds are increasingly recognized to have rapid effects on membrane surface receptors that often have not been identified at the molecular level. We show here that TRPM3, a divalent-permeable cation channel, is rapidly and reversibly activated by extracellular pregnenolone sulphate, a neuroactive steroid. We show that pregnenolone sulphate activates endogenous TRPM3 channels in insulin-producing beta cells. Application of pregnenolone sulphate led to a rapid calcium influx and enhanced insulin secretion from pancreatic islets. Our results establish that TRPM3 is an essential component of an ionotropic steroid receptor enabling unanticipated crosstalk between steroidal and insulin-signalling endocrine systems.
Collapse
Affiliation(s)
- Thomas F J Wagner
- Emmy Noether Research Group Toxikologie, Universität des Saarlandes, D-66421 Homburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
590
|
Gomis A, Soriano S, Belmonte C, Viana F. Hypoosmotic- and pressure-induced membrane stretch activate TRPC5 channels. J Physiol 2008; 586:5633-49. [PMID: 18832422 DOI: 10.1113/jphysiol.2008.161257] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transient receptor potential (TRP) channels mediate a wide array of sensory functions. We investigated the role of TRPC5, a poorly characterized channel widely expressed in the central and peripheral nervous system, as a potential osmosensory protein. Here we show that hypoosmotic stimulation activates TRPC5 channels resulting in a large calcium influx. The response to osmotically induced membrane stretch is blocked by GsMTx-4, an inhibitor of stretch activated ion channels. Direct hypoosmotic activation of TRPC5 is independent of phospholipase C function. However, the osmotic response is inhibited in a cell line in which PIP(2) levels are reduced by regulated overexpression of a lipid phosphatase. The response was restored by increasing intracellular PIP(2) levels through the patch pipette. The mechano-sensitivity of the channel was probed in the whole-cell configuration by application of steps of positive pressure through the patch pipette. Pressure-induced membrane stretch also activated TRPC5 channels, suggesting its role as a transducer of osmo-mechanical stimuli. We also demonstrated the expression of TRPC5 in sensory neurones which together with the osmo-mechanical characteristics of TRPC5 channels suggest its putative role in mechanosensory transduction events.
Collapse
Affiliation(s)
- Ana Gomis
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández. Av. Ramón y Cajal s/n. 03550 Sant Joan d'Alacant, Alicante, Spain.
| | | | | | | |
Collapse
|
591
|
Garcia-Elias A, Lorenzo IM, Vicente R, Valverde MA. IP3 receptor binds to and sensitizes TRPV4 channel to osmotic stimuli via a calmodulin-binding site. J Biol Chem 2008; 283:31284-8. [PMID: 18826956 DOI: 10.1074/jbc.c800184200] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of the non-selective cation channel TRPV4 by mechanical and osmotic stimuli requires the involvement of phospholipase A2 and the subsequent production of the arachidonic acid metabolites, epoxieicosatrienoic acids (EET). Previous studies have shown that inositol trisphosphate (IP3) sensitizes TRPV4 to mechanical, osmotic, and direct EET stimulation. We now search for the IP3 receptor-binding site on TRPV4 and its relevance to IP3-mediated sensitization. Three putative sites involved in protein-protein interactions were evaluated: a proline-rich domain (PRD), a calmodulin (CaM)-binding site, and the last four amino acids (DAPL) that show a PDZ-binding motif-like. TRPV4-DeltaCaM-(Delta812-831) channels preserved activation by hypotonicity, 4alpha-phorbol 12,13-didecanoate, and EET but lost their physical interaction with IP3 receptor 3 and IP3-mediated sensitization. Deletion of a PDZ-binding motif-like (TRPV4-DeltaDAPL) did not affect channel activity or IP3-mediated sensitization, whereas TRPV4-DeltaPRD-(Delta132-144) resulted in loss of channel function despite correct trafficking. We conclude that IP3-mediated sensitization requires IP3 receptor binding to a TRPV4 C-terminal domain that overlaps with a previously described calmodulin-binding site.
Collapse
Affiliation(s)
- Anna Garcia-Elias
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Edifici PRBB, C/Dr. Aiguader 88, Barcelona 08003, Spain
| | | | | | | |
Collapse
|
592
|
Becker D, Müller M, Leuner K, Jendrach M. The C-terminal domain of TRPV4 is essential for plasma membrane localization. Mol Membr Biol 2008; 25:139-51. [PMID: 18307101 DOI: 10.1080/09687680701635237] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Many members of the TRP superfamily oligomerize in the ER before trafficking to the plasma membrane. For membrane localization of the non-selective cation channel TRPV4 specific domains in the N-terminus are required, but the role of the C-terminus in the oligomerization and trafficking process has been not determined until now. Therefore, the localization of recombinant TRPV4 in two cell models was analyzed: HaCaT keratinocytes that express TRPV4 endogenously were compared to CHO cells that are devoid of endogenous TRPV4. When deletions were introduced in the C-terminal domain three states of TRPV4 localization were defined: a truncated TRPV4 protein of 855 amino acids was exported to the plasma membrane like the full-length channel (871 aa) and was also functional. Mutants with a length of 828 to 844 amino acids remained in the ER of CHO cells, but in HaCaT cells plasma membrane localization was partially rescued by oligomerization with endogenous TRPV4. This was confirmed by coexpression of recombinant full-length TRPV4 together with these deletion mutants, which resulted in an almost complete plasma membrane localization of both proteins and significant FRET in the plasma membrane and the ER. All deletions upstream of amino acid 828 resulted in total ER retention that could not rescued by coexpression with the full-length protein. However, these deletion mutants did not impair export of full-length TRPV4, implying that no oligomerization took place. These data indicate that the C-terminus of TRPV4 is required for oligomerization, which takes place in the ER and precedes plasma membrane trafficking.
Collapse
Affiliation(s)
- Daniel Becker
- Kinematic Cell Research, Institute for Cell Biology and Neuroscience, JW Goethe University, Frankfurt/Main, Germany
| | | | | | | |
Collapse
|
593
|
Chen JK, Chen J, Imig JD, Wei S, Hachey DL, Guthi JS, Falck JR, Capdevila JH, Harris RC. Identification of novel endogenous cytochrome p450 arachidonate metabolites with high affinity for cannabinoid receptors. J Biol Chem 2008; 283:24514-24. [PMID: 18606824 PMCID: PMC2528993 DOI: 10.1074/jbc.m709873200] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 07/01/2008] [Indexed: 12/20/2022] Open
Abstract
Arachidonic acid is an essential constituent of cell membranes that is esterified to the sn-2-position of glycerophospholipids and is released from selected lipid pools by phospholipase cleavage. The released arachidonic acid can be metabolized by three enzymatic pathways: the cyclooxygenase pathway forming prostaglandins and thromboxanes, the lipoxygenase pathway generating leukotrienes and lipoxins, and the cytochrome P450 (cP450) pathway producing epoxyeicosatrienoic acids and hydroxyeicosatetraenoic acids. The present study describes a novel group of cP450 epoxygenase-dependent metabolites of arachidonic acid, termed 2-epoxyeicosatrienoylglycerols (2-EG), including two regioisomers, 2-(11,12-epoxyeicosatrienoyl)glycerol (2-11,12-EG) and 2-(14,15-epoxyeicosatrienoyl)glycerol (2-14,15-EG), which are both produced in the kidney and spleen, whereas 2-11,12-EG is also detected in the brain. Both 2-11,12-EG and 2-14,15-EG activated the two cannabinoid (CB) receptor subtypes, CB1 and CB2, with high affinity and elicited biological responses in cultured cells expressing CB receptors and in intact animals. In contrast, the parental arachidonic acid and epoxyeicosatrienoic acids failed to activate CB1 or CB2 receptors. Thus, these cP450 epoxygenase-dependent metabolites are a novel class of endogenously produced, biologically active lipid mediators with the characteristics of endocannabinoids. This is the first evidence of a cytochrome P450-dependent arachidonate metabolite that can activate G-protein-coupled cell membrane receptors and suggests a functional link between the cytochrome P450 enzyme system and the endocannabinoid system.
Collapse
Affiliation(s)
- Jian-Kang Chen
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| | - Jianchun Chen
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| | - John D. Imig
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| | - Shouzuo Wei
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| | - David L. Hachey
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| | - Jagadeesh Setti Guthi
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| | - John R. Falck
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| | - Jorge H. Capdevila
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| | - Raymond C. Harris
- Departments of Medicine and
Biochemistry, Vanderbilt University, Nashville,
Tennessee 37232, the Department of Veterans Affairs,
Nashville, Tennessee 37212, the Vascular Biology
Center, Medical College of Georgia, Augusta, Georgia 30912 and the
Department of Biochemistry, University of Texas
Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
594
|
Shioya T, Sato K, Sano M, Watanabe H. [Transient receptor potential (TRP) channel and cough]. Nihon Yakurigaku Zasshi 2008; 131:417-22. [PMID: 18552442 DOI: 10.1254/fpj.131.417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
595
|
Cenac N, Altier C, Chapman K, Liedtke W, Zamponi G, Vergnolle N. Transient receptor potential vanilloid-4 has a major role in visceral hypersensitivity symptoms. Gastroenterology 2008; 135:937-46, 946.e1-2. [PMID: 18565335 DOI: 10.1053/j.gastro.2008.05.024] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2007] [Revised: 04/23/2008] [Accepted: 05/01/2008] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS The transient receptor potential vanilloid-4 (TRPV4) is an osmosensitive channel that responds to mechanical stimulation. We hypothesized that TRPV4 could be important in visceral nociception and in the development of hypersensitivity. METHODS TRPV4 expression was investigated by immunohistochemistry and reverse transcription-polymerase chain reaction. Calcium signaling and patch-clamp studies were performed in dorsal root ganglia (DRG) neurons validating the use of 4alphaPDD as a selective TRPV4 agonist. The effects of TRPV4 activation on visceral nociception were evaluated in mice that received intracolonically TRPV4 agonist (4 alpha-phorbol 12,13-didecanoate [4alphaPDD]) and in TRPV4-deficient mice in which abdominal muscle contractions in response to colorectal distention (CRD) were recorded. Intervertebral injections of TRPV4 or mismatch small interfering RNA (siRNA) were used to specifically down-regulate TRPV4 expression in sensory neurons and to investigate the role of TRPV4 in basal visceral nociception or in protease-activated receptor 2 (PAR(2)) activation-induced visceral hypersensitivity. RESULTS TRPV4 agonist 4alphaPDD specifically activated a cationic current and calcium influx in colonic projections of DRG neurons and caused dose-dependent visceral hypersensitivity. TRPV4-targeted but not mismatched siRNA intervertebral treatments were effective at reducing basal visceral nociception, as well as 4alphaPDD or PAR(2) agonist-induced hypersensitivity. Effects of the TRPV4 ligand were lost in TRPV4-deficient mice. CONCLUSIONS 4alphaPDD selectively activates TRPV4 in sensory neurons projecting from the colon, and TRPV4 activation causes visceral hypersensitivity. TRPV4 activation is implicated in the nociceptive response to CRD in basal conditions and in PAR(2) agonist-induced hypersensitivity. These results suggest a pivotal role for TRPV4 in visceral nociception and hypersensitivity.
Collapse
Affiliation(s)
- Nicolas Cenac
- Department of Pharmacology and Therapeutics, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
596
|
Guibert C, Ducret T, Savineau JP. Voltage-independent calcium influx in smooth muscle. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2008; 98:10-23. [DOI: 10.1016/j.pbiomolbio.2008.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
597
|
Rock MJ, Prenen J, Funari VA, Funari TL, Merriman B, Nelson SF, Lachman RS, Wilcox WR, Reyno S, Quadrelli R, Vaglio A, Owsianik G, Janssens A, Voets T, Ikegawa S, Nagai T, Rimoin DL, Nilius B, Cohn DH. Gain-of-function mutations in TRPV4 cause autosomal dominant brachyolmia. Nat Genet 2008; 40:999-1003. [PMID: 18587396 PMCID: PMC3525077 DOI: 10.1038/ng.166] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Accepted: 04/23/2008] [Indexed: 12/16/2022]
Abstract
The brachyolmias constitute a clinically and genetically heterogeneous group of skeletal dysplasias characterized by a short trunk, scoliosis and mild short stature. Here, we identify a locus for an autosomal dominant form of brachyolmia on chromosome 12q24.1-12q24.2. Among the genes in the genetic interval, we selected TRPV4, which encodes a calcium permeable cation channel of the transient receptor potential (TRP) vanilloid family, as a candidate gene because of its cartilage-selective gene expression pattern. In two families with the phenotype, we identified point mutations in TRPV4 that encoded R616Q and V620I substitutions, respectively. Patch clamp studies of transfected HEK cells showed that both mutations resulted in a dramatic gain of function characterized by increased constitutive activity and elevated channel activation by either mechano-stimulation or agonist stimulation by arachidonic acid or the TRPV4-specific agonist 4alpha-phorbol 12,13-didecanoate (4alphaPDD). This study thus defines a previously unknown mechanism, activation of a calcium-permeable TRP ion channel, in skeletal dysplasia pathogenesis.
Collapse
Affiliation(s)
- Matthew J Rock
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
598
|
Shimizu T, Janssens A, Voets T, Nilius B. Regulation of the murine TRPP3 channel by voltage, pH, and changes in cell volume. Pflugers Arch 2008; 457:795-807. [PMID: 18663466 DOI: 10.1007/s00424-008-0558-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 07/08/2008] [Accepted: 07/10/2008] [Indexed: 01/10/2023]
Abstract
Transient receptor potential (TRP) polycystin 3 (TRPP3) is a member of the TRP superfamily of cation channels. Murine TRPP3 has been reported to form an acid-activated cation channel on the plasma membrane when coexpressed with the polycystin 1-like protein 3 (PKD1L3); however, the function and biophysical properties of TRPP3-dependent channels have not yet been characterized in detail. Here we show that overexpression of murine TRPP3 channel in HEK293 cells, without coexpression of PDK1-like proteins, leads to robust channel activity. These channels exhibit a high single-channel conductance of 184 pS at negative potentials, are Ca2+-permeable, and relatively nonselective between cations. Whole-cell experiments showed a characteristic form of voltage-dependent gating of TRPP3 channels, whereby repolarization after depolarization caused large transient inward TRPP3 tail currents. Moreover, we found that TRPP3 activity was increased upon cell swelling and by alkalization. Taken together, our results demonstrate that TRPP3, on its own, can act as a voltage-dependent, pH- and volume-sensitive plasma membrane cation channel.
Collapse
Affiliation(s)
- Takahiro Shimizu
- Department of Molecular Cell Biology, Laboratory of Ion Channel Research, Katholieke Universiteit Leuven, Campus Gasthuisberg, Herestraat 49, Bus 802, B-3000, Leuven, Belgium
| | | | | | | |
Collapse
|
599
|
Abstract
Arachidonic acid (AA), a polyunsaturated fatty acid with four double bonds, has multiple actions on living cells. Many of these effects are mediated by an action of AA or its metabolites on ion channels. During the last 10 years, new types of ion channels, transient receptor potential (TRP) channels, store-operated calcium entry (SOCE) channels and non-SOCE channels have been studied. This review summarizes our current knowledge about the effects of AA on TRP and non-SOCE channels as well as classical ion channels. It aims to distinguish between effects of AA itself and effects of AA metabolites. Lipid mediators are of clinical interest because some of them (for example, leukotrienes) play a role in various diseases, others (such as prostaglandins) are targets for pharmacological therapeutic intervention.
Collapse
|
600
|
|