501
|
Kim KS, Shin DH, Nam JH, Park KS, Zhang YH, Kim WK, Kim SJ. Functional Expression of TRPV4 Cation Channels in Human Mast Cell Line (HMC-1). THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2010; 14:419-25. [PMID: 21311684 PMCID: PMC3034123 DOI: 10.4196/kjpp.2010.14.6.419] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 11/15/2010] [Accepted: 11/17/2010] [Indexed: 11/15/2022]
Abstract
Mast cells are activated by specific allergens and also by various nonspecific stimuli, which might induce physical urticaria. This study investigated the functional expression of temperature sensitive transient receptor potential vanilloid (TRPV) subfamily in the human mast cell line (HMC-1) using whole-cell patch clamp techniques. The temperature of perfusate was raised from room temperature (RT, 23~25℃ to a moderately high temperature (MHT, 37~39℃ to activate TRPV3/4, a high temperature (HT, 44~46℃ to activate TRPV1, or a very high temperature (VHT, 53~55℃ to activate TRPV2. The membrane conductance of HMC-1 was increased by MHT and HT in about 50% (21 of 40) of the tested cells, and the I/V curves showed weak outward rectification. VHT-induced current was 10-fold larger than those induced by MHT and HT. The application of the TRPV4 activator 4α-phorbol 12,13-didecanoate (4αPDD, 1µM) induced weakly outward rectifying currents similar to those induced by MHT. However, the TRPV3 agonist camphor or TRPV1 agonist capsaicin had no effect. RT-PCR analysis of HMC-1 demonstrated the expression of TRPV4 as well as potent expression of TRPV2. The [Ca(2+)](c) of HMC-1 cells was also increased by MHT or by 4αPDD. In summary, our present study indicates that HMC-1 cells express Ca(2+)-permeable TRPV4 channels in addition to the previously reported expression of TRPV2 with a higher threshold of activating temperature.
Collapse
Affiliation(s)
- Kyung Soo Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Dong Hoon Shin
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Kyungju 780-714, Korea
| | - Kyung Sun Park
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Yin Hua Zhang
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Woo Kyung Kim
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Seoul 100-715, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, Korea
- Ischemia/Hypoxia Disease Institute, Medical Research Center, Seoul National University, Seoul 110-799, Korea
| |
Collapse
|
502
|
Connor M, Vaughan CW, Vandenberg RJ. N-acyl amino acids and N-acyl neurotransmitter conjugates: neuromodulators and probes for new drug targets. Br J Pharmacol 2010; 160:1857-71. [PMID: 20649585 DOI: 10.1111/j.1476-5381.2010.00862.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The myriad functions of lipids as signalling molecules is one of the most interesting fields in contemporary pharmacology, with a host of compounds recognized as mediators of communication within and between cells. The N-acyl conjugates of amino acids and neurotransmitters (NAANs) have recently come to prominence because of their potential roles in the nervous system, vasculature and the immune system. NAAN are compounds such as glycine, GABA or dopamine conjugated with long chain fatty acids. More than 70 endogenous NAAN have been reported although their physiological role remains uncertain, with various NAAN interacting with a low affinity at G protein coupled receptors (GPCR) and ion channels. Regardless of their potential physiological function, NAAN are of great interest to pharmacologists because of their potential as flexible tools to probe new sites on GPCRs, transporters and ion channels. NAANs are amphipathic molecules, with a wide variety of potential fatty acid and headgroup moieties, a combination which provides a rich source of potential ligands engaging novel binding sites and mechanisms for modulation of membrane proteins such as GPCRs, ion channels and transporters. The unique actions of subsets of NAAN on voltage-gated calcium channels and glycine transporters indicate that the wide variety of NAAN may provide a readily exploitable resource for defining new pharmacological targets. Investigation of the physiological roles and pharmacological potential of these simple lipid conjugates is in its infancy, and we believe that there is much to be learnt from their careful study.
Collapse
Affiliation(s)
- Mark Connor
- Brain and Mind Research Institute, University of Sydney, Camperdown, NSW, Australia.
| | | | | |
Collapse
|
503
|
Sokabe T, Tominaga M. The TRPV4 cation channel: A molecule linking skin temperature and barrier function. Commun Integr Biol 2010; 3:619-21. [PMID: 21331258 DOI: 10.4161/cib.3.6.13461] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 08/30/2010] [Indexed: 11/19/2022] Open
Abstract
The skin barrier function is indispensable for terrestrial animals to avoid dehydration. The function is achieved by a hydrophobic cornified layer consisting of dead keratinocytes and lipids, and by an intercellular junction barrier formed among differentiated keratinocytes. A recent report demonstrated that TRPV4, one of the temperature-sensitive cation channels, contributes to the formation and maintenance of the intercellular junction-dependent barrier in the skin. TRPV4 associates with the E-cadherin complex via β-catenin, and thereby participates in the promotion of cell-cell junction development. TRPV4 allows influx of Ca(2+) ions from the extracellular space at physiological skin temperatures. The Ca(2+) influx induces Rho activation and promotes actin fiber organization and junction formation, thereby augmenting barrier integrity. Indeed, the intercellular junction structures and the skin barrier function were impaired in TRPV4-deficeint mice. This novel role of TRPV4 in keratinocytes may explain the significant correlation between temperature and the condition of skin.>
Collapse
Affiliation(s)
- Takaaki Sokabe
- Division of Cell Signaling; Okazaki Institute for Integrative Bioscience; National Institutes of Natural Sciences
| | | |
Collapse
|
504
|
Endogenous lipid-derived ligands for sensory TRP ion channels and their pain modulation. Arch Pharm Res 2010; 33:1509-20. [DOI: 10.1007/s12272-010-1004-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 08/25/2010] [Accepted: 08/27/2010] [Indexed: 10/18/2022]
|
505
|
Gees M, Colsoul B, Nilius B. The role of transient receptor potential cation channels in Ca2+ signaling. Cold Spring Harb Perspect Biol 2010; 2:a003962. [PMID: 20861159 DOI: 10.1101/cshperspect.a003962] [Citation(s) in RCA: 327] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The 28 mammalian members of the super-family of transient receptor potential (TRP) channels are cation channels, mostly permeable to both monovalent and divalent cations, and can be subdivided into six main subfamilies: the TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin), TRPML (mucolipin), and the TRPA (ankyrin) groups. TRP channels are widely expressed in a large number of different tissues and cell types, and their biological roles appear to be equally diverse. In general, considered as polymodal cell sensors, they play a much more diverse role than anticipated. Functionally, TRP channels, when activated, cause cell depolarization, which may trigger a plethora of voltage-dependent ion channels. Upon stimulation, Ca2+ permeable TRP channels generate changes in the intracellular Ca2+ concentration, [Ca2+]i, by Ca2+ entry via the plasma membrane. However, more and more evidence is arising that TRP channels are also located in intracellular organelles and serve as intracellular Ca2+ release channels. This review focuses on three major tasks of TRP channels: (1) the function of TRP channels as Ca2+ entry channels; (2) the electrogenic actions of TRPs; and (3) TRPs as Ca2+ release channels in intracellular organelles.
Collapse
Affiliation(s)
- Maarten Gees
- KU Leuven, Department of Molecular Cell Biology, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, bus 802, Leuven, Belgium
| | | | | |
Collapse
|
506
|
Lo HY, Man CC, Fleck RW, Farrow NA, Ingraham RH, Kukulka A, Proudfoot JR, Betageri R, Kirrane T, Patel U, Sharma R, Hoermann MA, Kabcenell A, Lombaert SD. Substituted pyrazoles as novel sEH antagonist: investigation of key binding interactions within the catalytic domain. Bioorg Med Chem Lett 2010; 20:6379-83. [PMID: 20934334 DOI: 10.1016/j.bmcl.2010.09.095] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 09/14/2010] [Accepted: 09/16/2010] [Indexed: 10/19/2022]
Abstract
A novel series of pyrazole sEH inhibitors is reported. Lead optimization efforts to replace the aniline core are also described. In particular, 2-pyridine, 3-pyridine and pyridazine analogs are potent sEH inhibitors with favorable CYP3A4 inhibitory and microsomal stability profiles.
Collapse
Affiliation(s)
- Ho Yin Lo
- Boehringer Ingelheim Pharmaceuticals Inc., Biomolecular Screening, 900 Ridgebury Rd., PO Box 368, Ridgefield, CT 06877, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
507
|
Higashimori H, Blanco VM, Tuniki VR, Falck JR, Filosa JA. Role of epoxyeicosatrienoic acids as autocrine metabolites in glutamate-mediated K+ signaling in perivascular astrocytes. Am J Physiol Cell Physiol 2010; 299:C1068-78. [PMID: 20844244 DOI: 10.1152/ajpcell.00225.2010] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Epoxyeicosatrienoic acids (EETs), synthesized and released by astrocytes in response to glutamate, are known to play a pivotal role in neurovascular coupling. In vascular smooth muscle cells (VSMC), EETs activate large-conductance, Ca(2+)-activated K(+) (BK) channels resulting in hyperpolarization and vasodilation. However, the functional role and mechanism of action for glial-derived EETs are still to be determined. In this study, we evaluated the effect of the synthetic EET analog 11-nonyloxy-undec-8(Z)-enoic acid (NUD-GA) on outward K(+) currents mediated by calcium-activated K(+) channels. Addition of NUD-GA significantly increased intracellular Ca(2+) and outward K(+) currents in perivascular astrocytes. NUD-GA-induced currents were significantly inhibited by BK channel blockers paxilline and tetraethylammonium (TEA) (23.4 ± 2.4%; P < 0.0005). Similarly, NUD-GA-induced currents were also significantly inhibited in the presence of the small-conductance Ca(2+)-activated K(+) channel inhibitor apamin along with a combination of blockers against glutamate receptors (12.8 ± 2.70%; P < 0.05). No changes in outward currents were observed in the presence of the channel blocker for intermediate-conductance K(+) channels TRAM-34. Blockade of the endogenous production of EETs with N-methylsulfonyl-6-(2-propargyloxyphenyl)hexanamide (MS-PPOH) significantly blunted (dl)-1-aminocyclopentane-trans-1,3-dicarboxylic acid (t-ACPD)-induced outward K(+) currents (P < 0.05; n = 6). Both NUD-GA and t-ACPD significantly increased BK channel single open probability; the later was blocked following MS-PPOH incubation. Our data supports the idea that EETs are potent K(+) channel modulators in cortical perivascular astrocytes and further suggest that these metabolites may participate in NVC by modulating the levels of K(+) released at the gliovascular space.
Collapse
Affiliation(s)
- Haruki Higashimori
- Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | | | |
Collapse
|
508
|
Wu LJ, Sweet TB, Clapham DE. International Union of Basic and Clinical Pharmacology. LXXVI. Current progress in the mammalian TRP ion channel family. Pharmacol Rev 2010; 62:381-404. [PMID: 20716668 PMCID: PMC2964900 DOI: 10.1124/pr.110.002725] [Citation(s) in RCA: 440] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transient receptor potential (TRP) channels are a large family of ion channel proteins, surpassed in number in mammals only by voltage-gated potassium channels. TRP channels are activated and regulated through strikingly diverse mechanisms, making them suitable candidates for cellular sensors. They respond to environmental stimuli such as temperature, pH, osmolarity, pheromones, taste, and plant compounds, and intracellular stimuli such as Ca(2+) and phosphatidylinositol signal transduction pathways. However, it is still largely unknown how TRP channels are activated in vivo. Despite the uncertainties, emerging evidence using TRP channel knockout mice indicates that these channels have broad function in physiology. Here we review the recent progress on the physiology, pharmacology and pathophysiological function of mammalian TRP channels.
Collapse
Affiliation(s)
- Long-Jun Wu
- Howard Hughes Medical Institute, Department of Cardiology, Children's Hospital Boston, 320 Longwood Avenue, Boston, MA 02115, USA
| | | | | |
Collapse
|
509
|
Dadon D, Minke B. Cellular functions of transient receptor potential channels. Int J Biochem Cell Biol 2010; 42:1430-45. [PMID: 20399884 PMCID: PMC2910227 DOI: 10.1016/j.biocel.2010.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 04/08/2010] [Accepted: 04/08/2010] [Indexed: 11/26/2022]
Abstract
Transient Receptor Potential channels are polymodal cellular sensors involved in a wide variety of cellular processes, mainly by increasing cellular Ca(2+). In this review we focus on the roles of these channels in: (i) cell death (ii) proliferation and differentiation and (iii) transmitter release. Cell death: Ca(2+) influx participates in apoptotic and necrotic cell death. The Ca(2+) permeability and high sensitivity of part of these channels to oxidative/metabolic stress make them important participants in cell death. Several examples are given. Transient Receptor Potential Melastatin 2 is activated by H(2)O(2), inducing cell death through an increase in cellular Ca(2+) and activation of Poly ADP-Ribose Polymerase. Exposure of cultured cortical neurons to oxygen-glucose deprivation, in vitro, causes cell death via cation influx, mediated by Transient Receptor Potential Melastatin 7. Metabolic stress constitutively activates the Ca(2+) permeable Transient Receptor Potential channels of Drosophila photoreceptor in the dark, potentially leading to retinal degeneration. Similar sensitivity to metabolic stress characterizes several mammalian Transient Receptor Potential Canonical channels. Proliferation and differentiation: The rise in cytosolic Ca(2+) induces cell growth, differentiation and proliferation via activation of several transcription factors. Activating a variety of store operated and Transient Receptor Potential channels cause a rise in cytosolic Ca(2+), making these channels components involved in proliferation and differentiation. Transmitter release: Transient Receptor Potential Melastatin 7 channels reside in synaptic vesicles and regulate neurotransmitter release by a mechanism that is not entirely clear. All the above features of Transient Receptor Potential channels make them crucial components in important, sometimes conflicting, cellular processes that still need to be explored.
Collapse
Affiliation(s)
- Daniela Dadon
- Department of Medical Neurobiology, The Institute of Medical Research Israel-Canada and the Kühne Minerva Center for Studies of Visual Transduction, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
| | - Baruch Minke
- Department of Medical Neurobiology, The Institute of Medical Research Israel-Canada and the Kühne Minerva Center for Studies of Visual Transduction, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
| |
Collapse
|
510
|
Hamanaka K, Jian MY, Townsley MI, King JA, Liedtke W, Weber DS, Eyal FG, Clapp MM, Parker JC. TRPV4 channels augment macrophage activation and ventilator-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2010; 299:L353-62. [PMID: 20562229 PMCID: PMC2951075 DOI: 10.1152/ajplung.00315.2009] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 06/11/2010] [Indexed: 12/24/2022] Open
Abstract
We have previously implicated transient receptor potential vanilloid 4 (TRPV4) channels and alveolar macrophages in initiating the permeability increase in response to high peak inflation pressure (PIP) ventilation. Alveolar macrophages were harvested from TRPV4(-/-) and TRPV4(+/+) mice and instilled in the lungs of mice of the opposite genotype. Filtration coefficients (K(f)) measured in isolated perfused lungs after ventilation with successive 30-min periods of 9, 25, and 35 cmH(2)O PIP did not significantly increase in lungs from TRPV4(-/-) mice but increased >2.2-fold in TRPV4(+/+) lungs, TRPV4(+/+) lungs instilled with TRPV4(-/-) macrophages, and TRPV4(-/-) lungs instilled with TRPV4(+/+) macrophages after ventilation with 35 cmH(2)O PIP. Activation of TRPV4 with 4-alpha-phorbol didecanoate (4alphaPDD) significantly increased intracellular calcium, superoxide, and nitric oxide production in TRPV4(+/+) macrophages but not TRPV4(-/-) macrophages. Cross-sectional areas increased nearly 3-fold in TRPV4(+/+) macrophages compared with TRPV4(-/-) macrophages after 4alphaPDD. Immunohistochemistry staining of lung tissue for nitrotyrosine revealed increased amounts in high PIP ventilated TRPV4(+/+) lungs compared with low PIP ventilated TRPV4(+/+) or high PIP ventilated TRPV4(-/-) lungs. Thus TRPV4(+/+) macrophages restored susceptibility of TRPV4(-/-) lungs to mechanical injury. A TRPV4 agonist increased intracellular calcium and reactive oxygen and nitrogen species in harvested TRPV4(+/+) macrophages but not TRPV4(-/-) macrophages. K(f) increases correlated with tissue nitrotyrosine, a marker of peroxynitrite production.
Collapse
Affiliation(s)
- Kazutoshi Hamanaka
- Department of Physiology, University of South Alabama, Mobile, Alabama 36688, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
511
|
Ceppa E, Cattaruzza F, Lyo V, Amadesi S, Pelayo JC, Poole DP, Vaksman N, Liedtke W, Cohen DM, Grady EF, Bunnett NW, Kirkwood KS. Transient receptor potential ion channels V4 and A1 contribute to pancreatitis pain in mice. Am J Physiol Gastrointest Liver Physiol 2010; 299:G556-71. [PMID: 20539005 PMCID: PMC2950679 DOI: 10.1152/ajpgi.00433.2009] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The mechanisms of pancreatic pain, a cardinal symptom of pancreatitis, are unknown. Proinflammatory agents that activate transient receptor potential (TRP) channels in nociceptive neurons can cause neurogenic inflammation and pain. We report a major role for TRPV4, which detects osmotic pressure and arachidonic acid metabolites, and TRPA1, which responds to 4-hydroxynonenal and cyclopentenone prostaglandins, in pancreatic inflammation and pain in mice. Immunoreactive TRPV4 and TRPA1 were detected in pancreatic nerve fibers and in dorsal root ganglia neurons innervating the pancreas, which were identified by retrograde tracing. Agonists of TRPV4 and TRPA1 increased intracellular Ca(2+) concentration ([Ca(2+)](i)) in these neurons in culture, and neurons also responded to the TRPV1 agonist capsaicin and are thus nociceptors. Intraductal injection of TRPV4 and TRPA1 agonists increased c-Fos expression in spinal neurons, indicative of nociceptor activation, and intraductal TRPA1 agonists also caused pancreatic inflammation. The effects of TRPV4 and TRPA1 agonists on [Ca(2+)](i), pain and inflammation were markedly diminished or abolished in trpv4 and trpa1 knockout mice. The secretagogue cerulein induced pancreatitis, c-Fos expression in spinal neurons, and pain behavior in wild-type mice. Deletion of trpv4 or trpa1 suppressed c-Fos expression and pain behavior, and deletion of trpa1 attenuated pancreatitis. Thus TRPV4 and TRPA1 contribute to pancreatic pain, and TRPA1 also mediates pancreatic inflammation. Our results provide new information about the contributions of TRPV4 and TRPA1 to inflammatory pain and suggest that channel antagonists are an effective therapy for pancreatitis, when multiple proinflammatory agents are generated that can activate and sensitize these channels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wolfgang Liedtke
- 3Department of Medicine and Neurobiology, Duke University Medical Center, Durham, North Carolina; and
| | - David M. Cohen
- 4Portland Veterans Affairs Medical Center, Portland, Oregon
| | | | - Nigel W. Bunnett
- Departments of 1Surgery and ,2Physiology, University of California, San Francisco, San Francisco, California;
| | | |
Collapse
|
512
|
Loukin S, Zhou X, Su Z, Saimi Y, Kung C. Wild-type and brachyolmia-causing mutant TRPV4 channels respond directly to stretch force. J Biol Chem 2010; 285:27176-27181. [PMID: 20605796 PMCID: PMC2930716 DOI: 10.1074/jbc.m110.143370] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 06/24/2010] [Indexed: 11/06/2022] Open
Abstract
Whether animal ion channels functioning as mechanosensors are directly activated by stretch force or indirectly by ligands produced by the stretch is a crucial question. TRPV4, a key molecular model, can be activated by hypotonicity, but the mechanism of activation is unclear. One model has this channel being activated by a downstream product of phospholipase A(2), relegating mechanosensitivity to the enzymes or their regulators. We expressed rat TRPV4 in Xenopus oocytes and repeatedly examined >200 excised patches bathed in a simple buffer. We found that TRPV4 can be activated by tens of mm Hg pipette suctions with open probability rising with suction even in the presence of relevant enzyme inhibitors. Mechanosensitivity of TRPV4 provides the simplest explanation of its various force-related physiological roles, one of which is in the sensing of weight load during bone development. Gain-of-function mutants cause heritable skeletal dysplasias in human. We therefore examined the brachyolmia-causing R616Q gain-of-function channel and found increased whole-cell current densities compared with wild-type channels. Single-channel analysis revealed that R616Q channels maintain mechanosensitivity but have greater constitutive activity and no change in unitary conductance or rectification.
Collapse
MESH Headings
- Animals
- Bone Diseases, Developmental/genetics
- Bone Diseases, Developmental/metabolism
- Bone Diseases, Developmental/pathology
- Bone Diseases, Developmental/physiopathology
- Disease Models, Animal
- Genetic Diseases, Inborn/genetics
- Genetic Diseases, Inborn/metabolism
- Genetic Diseases, Inborn/pathology
- Genetic Diseases, Inborn/physiopathology
- Humans
- Mutation, Missense
- Oocytes
- Phospholipases A2/genetics
- Phospholipases A2/metabolism
- Rats
- TRPV Cation Channels/genetics
- TRPV Cation Channels/metabolism
- Weight-Bearing
- Xenopus
Collapse
Affiliation(s)
- Stephen Loukin
- Laboratory of Molecular Biology, University of Wisconsin, Madison, Wisconsin 53706.
| | - Xinliang Zhou
- Laboratory of Molecular Biology, University of Wisconsin, Madison, Wisconsin 53706
| | - Zhenwei Su
- Laboratory of Molecular Biology, University of Wisconsin, Madison, Wisconsin 53706
| | - Yoshiro Saimi
- Laboratory of Molecular Biology, University of Wisconsin, Madison, Wisconsin 53706
| | - Ching Kung
- Laboratory of Molecular Biology, University of Wisconsin, Madison, Wisconsin 53706; Department of Genetics, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
513
|
The Role of Cannabinoid Receptors in the Descending Modulation of Pain. Pharmaceuticals (Basel) 2010; 3:2661-2673. [PMID: 27713370 PMCID: PMC4033943 DOI: 10.3390/ph3082661] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 07/30/2010] [Accepted: 08/16/2010] [Indexed: 11/27/2022] Open
Abstract
The endogenous antinociceptive descending pathway represents a circuitry of the supraspinal central nervous system whose task is to counteract pain. It includes the periaqueductal grey (PAG)-rostral ventromedial medulla (RVM)-dorsal horn (DH) axis, which is the best characterized pain modulation system through which pain is endogenously inhibited. Thus, an alternative rational strategy for silencing pain is the activation of this anatomical substrate. Evidence of the involvement of cannabinoid receptors (CB) in the supraspinal modulation of pain can be found in several studies in which intra-cerebral microinjections of cannabinoid ligands or positive modulators have proved to be analgesic in different pain models, whereas cannabinoid receptor antagonists or antisense nucleotides towards CB1 receptors have facilitated pain. Like opioids, cannabinoids produce centrally-mediated analgesia by activating a descending pathway which includes PAG and its projection to downstream RVM neurons, which in turn send inhibitory projections to the dorsal horn of the spinal cord. Indeed, several studies underline a supraspinal regulation of cannabinoids on γ-aminobutyric acid (GABA) and glutamate release which inhibit and enhance the antinociceptive descending pathway, respectively. Cannabinoid receptor activation expressed on presynaptic GABAergic terminals reduces the probability of neurotransmitter release thus dis-inhibiting the PAG-RVM-dorsal horn antinociceptive pathway. Cannabinoids seem to increase glutamate release (maybe as consequence of GABA decrease) and to require glutamate receptor activation to induce antinociception. The consequent outcome is behavioral analgesia, which is reproduced in several pain conditions, from acute to chronic pain models such as inflammatory and neuropathic pain. Taken together these findings would suggest that supraspinal cannabinoid receptors have broad applications, from pain control to closely related central nervous system diseases such as anxiety and depression.
Collapse
|
514
|
Kang L, Gao J, Schafer WR, Xie Z, Xu XZS. C. elegans TRP family protein TRP-4 is a pore-forming subunit of a native mechanotransduction channel. Neuron 2010; 67:381-91. [PMID: 20696377 PMCID: PMC2928144 DOI: 10.1016/j.neuron.2010.06.032] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2010] [Indexed: 11/30/2022]
Abstract
Mechanotransduction channels mediate several common sensory modalities such as hearing, touch, and proprioception; however, very little is known about the molecular identities of these channels. Many TRP family channels have been implicated in mechanosensation, but none have been demonstrated to form a mechanotransduction channel, raising the question of whether TRP proteins simply play indirect roles in mechanosensation. Using Caenorhabditis elegans as a model, here we have recorded a mechanosensitive conductance in a ciliated mechanosensory neuron in vivo. This conductance develops very rapidly upon mechanical stimulation with its latency and activation time constant reaching the range of microseconds, consistent with mechanical gating of the conductance. TRP-4, a TRPN (NOMPC) subfamily channel, is required for this conductance. Importantly, point mutations in the predicted pore region of TRP-4 alter the ion selectivity of the conductance. These results indicate that TRP-4 functions as an essential pore-forming subunit of a native mechanotransduction channel.
Collapse
Affiliation(s)
- Lijun Kang
- Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
515
|
Westlund KN, Kochukov MY, Lu Y, McNearney TA. Impact of central and peripheral TRPV1 and ROS levels on proinflammatory mediators and nociceptive behavior. Mol Pain 2010; 6:46. [PMID: 20691059 PMCID: PMC2924298 DOI: 10.1186/1744-8069-6-46] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 08/06/2010] [Indexed: 02/07/2023] Open
Abstract
Background Transient receptor potential vanilloid 1 (TRPV1) channels are important membrane sensors on peripheral nerve endings and on supportive non-neuronal synoviocytes in the knee joint. TRPV 1 ion channels respond with activation of calcium and sodium fluxes to pH, thermal, chemical, osmotic, mechanical and other stimuli abundant in inflamed joints. In the present study, the kaolin/carrageenan (k/c) induced knee joint arthritis model in rats, as well as primary and clonal human synoviocyte cultures were used to understand the reciprocal interactions between reactive nitroxidative species (ROS) and functional TRPV1 channels. ROS generation was monitored with ROS sensitive dyes using live cell imaging in vitro and in spinal tissue histology, as well as with measurement of ROS metabolites in culture media using HPLC. Results Functional responses in the experimental arthritis model, including increased nociceptive responses (thermal and mechanical hyperalgesia and allodynia), knee joint temperature reflecting local blood flow, and spinal cord ROS elevations were reduced by the ROS scavenger PBN after intraperitoneal pretreatment. Increases in TRPV1 and ROS, generated by synoviocytes in vitro, were reciprocally blocked by TRPV1 antagonists and the ROS scavenger. Further evidence is presented that synoviocyte responses to ROS and TRPV1 activation include increases in TNFα and COX-2, both measured as an indicator of the inflammation in vitro. Conclusions The results demonstrate that contributions of ROS to pronociceptive responses and neurogenic inflammation are mediated both centrally and peripherally. Responses are mediated by TRPV1 locally in the knee joint by synoviocytes, as well as by ROS-induced sensitization in the spinal cord. These findings and those of others reported in the literature indicate reciprocal interactions between TRPV1 and ROS play critical roles in the pathological and nociceptive responses active during arthritic inflammation.
Collapse
Affiliation(s)
- Karin N Westlund
- Department of Physiology, University of Kentucky, Lexington, KY 40536-0298, USA.
| | | | | | | |
Collapse
|
516
|
Cantero-Recasens G, Gonzalez JR, Fandos C, Duran-Tauleria E, Smit LAM, Kauffmann F, Antó JM, Valverde MA. Loss of function of transient receptor potential vanilloid 1 (TRPV1) genetic variant is associated with lower risk of active childhood asthma. J Biol Chem 2010; 285:27532-5. [PMID: 20639579 DOI: 10.1074/jbc.c110.159491] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Transient receptor potential cation channels of the vanilloid subfamily (TRPV) participate in the generation of Ca(2+) signals at different locations of the respiratory system, thereby controlling its correct functioning. TRPV1 expression and activity appear to be altered under pathophysiological conditions such as chronic cough and airway hypersensitivity, whereas TRPV4 single nucleotide polymorphisms (SNP) are associated with chronic obstructive pulmonary disease. However, to date, there is no information about the genetic impact of either TRPV1 or TRPV4 on asthma pathophysiology. We now report on the association of two functional SNPs, TRPV1-I585V and TRPV4-P19S, with childhood asthma. Both SNPs were genotyped in a population of 470 controls without respiratory symptoms and 301 asthmatics. Although none of the SNPs modified the risk of suffering from asthma, carriers of the TRPV1-I585V genetic variant showed a lower risk of current wheezing (odds ratio = 0.51; p = 0.01), a characteristic of active asthma, or cough (odds ratio = 0.57; p = 0.02). Functional analysis of TRPV1-I585V, using the Ca(2+)-sensitive dye fura-2 to measure intracellular [Ca(2+)] concentrations, revealed a decreased channel activity in response to two typical TRPV1 stimuli, heat and capsaicin. On the other hand, TRPV4-P19S, despite its loss-of-channel function, showed no significant association with asthma or the presence of wheezing. Our data suggest that genetically determined level of TRPV1 activity is relevant for asthma pathophysiology.
Collapse
Affiliation(s)
- Gerard Cantero-Recasens
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona 08003, Spain
| | | | | | | | | | | | | | | |
Collapse
|
517
|
Gradilone SA, Masyuk TV, Huang BQ, Banales JM, Lehmann GL, Radtke BN, Stroope A, Masyuk AI, Splinter PL, LaRusso NF. Activation of Trpv4 reduces the hyperproliferative phenotype of cystic cholangiocytes from an animal model of ARPKD. Gastroenterology 2010; 139:304-14.e2. [PMID: 20399209 PMCID: PMC2902576 DOI: 10.1053/j.gastro.2010.04.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 03/17/2010] [Accepted: 04/08/2010] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS In polycystic liver diseases, cyst formation involves cholangiocyte hyperproliferation. In polycystic kidney (PCK) rats, an animal model of autosomal-recessive polycystic kidney disease (ARPKD), decreased intracellular calcium [Ca(2+)](i) in cholangiocytes is associated with hyperproliferation. We recently showed transient receptor potential vanilloid 4 (Trpv4), a calcium-entry channel, is expressed in normal cholangiocytes and its activation leads to [Ca(2+)](i) increase. Thus, we hypothesized that pharmacologic activation of Trpv4 might reverse the hyperproliferative phenotype of PCK cholangiocytes. METHODS Trpv4 expression was examined in liver of normal and PCK rats, normal human beings, and patients with autosomal-dominant polycystic kidney disease or ARPKD. Trpv4 activation effect on cell proliferation and cyst formation was assessed in cholangiocytes derived from normal and PCK rats. The in vivo effects of Trpv4 activation on kidney and liver cysts was analyzed in PCK rats. RESULTS Trpv4 was overexpressed both at messenger RNA (8-fold) and protein (3-fold) levels in PCK cholangiocytes. Confocal and immunogold electron microscopy supported Trpv4 overexpression in the livers of PCK rats and ARPKD or autosomal-dominant polycystic kidney disease patients. Trpv4 activation in PCK cholangiocytes increased [Ca(2+)](i) by 30%, inhibiting cell proliferation by approximately 25%-50% and cyst growth in 3-dimensional culture (3-fold). Trpv4-small interfering RNA silencing blocked effects of Trpv4 activators by 70%. Trpv4 activation was associated with Akt phosphorylation and beta-Raf and Erk1/2 inhibition. In vivo, Trpv4 activation induced a significant decrease in renal cystic area and a nonsignificant decrease in liver cysts. CONCLUSIONS Taken together, our in vitro and in vivo data suggest that increasing intracellular calcium by Trpv4 activation may represent a potential therapeutic approach in PKD.
Collapse
Affiliation(s)
- Sergio A Gradilone
- Miles and Shirley Fiterman Center for Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
518
|
Vriens J, Nilius B, Vennekens R. Herbal compounds and toxins modulating TRP channels. Curr Neuropharmacol 2010; 6:79-96. [PMID: 19305789 PMCID: PMC2645550 DOI: 10.2174/157015908783769644] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Revised: 07/19/2007] [Accepted: 08/15/2007] [Indexed: 01/13/2023] Open
Abstract
Although the benefits are sometimes obvious, traditional or herbal medicine is regarded with skepticism, because the mechanism through which plant compounds exert their powers are largely elusive. Recent studies have shown however that many of these plant compounds interact with specific ion channels and thereby modulate the sensing mechanism of the human body. Especially members of the Transient Receptor Potential (TRP) channels have drawn large attention lately as the receptors for plant-derived compounds such as capsaicin and menthol. TRP channels constitute a large and diverse family of channel proteins that can serve as versatile sensors that allow individual cells and entire organisms to detect changes in their environment. For this family, a striking number of empirical views have turned into mechanism-based actions of natural compounds. In this review we will give an overview of herbal compounds and toxins, which modulate TRP channels.
Collapse
Affiliation(s)
- Joris Vriens
- Laboratory of Ion Channel Research, Department of Mol. Cell Biology, Division of Physiology, Campus Gasthuisberg, KU Leuven, Herestraat 49, B-3000 LEUVEN, Belgium
| | | | | |
Collapse
|
519
|
Story GM. The emerging role of TRP channels in mechanisms of temperature and pain sensation. Curr Neuropharmacol 2010; 4:183-96. [PMID: 18615141 DOI: 10.2174/157015906778019482] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 05/17/2006] [Indexed: 01/13/2023] Open
Abstract
Pain is universal and vital to survival. It is an essential component of our sense of touch; together, touch and pain have evolved to enable our awareness of the intricacies of our environment and to warn us of danger and possible injury. There is a clear link between temperature sensation and pain-painful temperature sensations occur acutely and are a hallmark of inflammatory and chronic pain disorders of the nervous system. Mounting evidence suggests a subset of Transient Receptor Potential (TRP) ion channels activated by temperature (thermoTRPs) are important molecular players in acute, inflammatory and chronic pain states. Varying degrees of heat activate four of these channels (TRPV1-4), while cooling temperatures ranging from pleasant to painful activate two distantly related thermoTRP channels (TRPM8 and TRPA1). ThermoTRP channels are also chemosensitive, being activated and or modulated by plant-derived small molecules and endogenous inflammatory mediators. All thermoTRPs are expressed in tissues essential to cutaneous thermal and pain sensation. This review examines the contribution of thermoTRP channels to our understanding of temperature and pain transduction at the molecular level.
Collapse
Affiliation(s)
- Gina M Story
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Avenue, Campus Box 8054, St. Louis, MO 63110, USA.
| |
Collapse
|
520
|
Lee EJ, Shin SH, Chun J, Hyun S, Kim Y, Kang SS. The modulation of TRPV4 channel activity through its Ser 824 residue phosphorylation by SGK1. Anim Cells Syst (Seoul) 2010. [DOI: 10.1080/19768354.2010.486939] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
521
|
Zhu Y, Parsons SP, Huizinga JD. Measurement of intracellular chloride ion concentration in ICC in situ and in explant culture. Neurogastroenterol Motil 2010; 22:704-9. [PMID: 20403100 DOI: 10.1111/j.1365-2982.2010.01501.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Chloride channels are proposed to play a central role in the electrical pacemaking mechanism of interstitial cells of Cajal (ICC). A key unknown factor in the consideration of this role is the chloride equilibrium potential (E(Cl)), as determined by the relative concentrations of intra- ([Cl(-)](i)) and extracellular ([Cl(-)](o)) chloride ions. METHODS To calculate the E(Cl) of ICC, [Cl(-)](i) was measured with the fluorescent chloride indicator N-(6-methoxyquinolyl) acetoethyl ester (MQAE). Pacemaker ICC in explant cultures or in situ, i.e. ICC associated with the myenteric plexus of the small intestine, were loaded with MQAE and fluorescence was measured by laser scanning confocal microscopy. The dye fluorescence was then calibrated against known [Cl(-)](i) by treating the explants or in situ preparations with chloride ionophore and varying bath chloride concentrations. KEY RESULTS In explants, ICC [Cl(-)](i) was measured as 13 mmol L(-1) with [Cl(-)](o) of 100 mmol L(-1), giving an E(Cl) of -52 mV [corrected]. With [Cl(-)](o) at 166 mmol L(-1), [Cl(-)](i) was 26 mmol L(-1), giving an E(Cl) of -47 mV[corrected]. In situ, ICC [Cl(-)](i) was measured as 26 mmol L(-1) with [Cl(-)](o) of 130 mmol L(-1), giving an E(Cl) of -41 mV [corrected]. Importantly ICC compensate for changes in extracellular chloride by changing [Cl(-)](i) and thus maintain E(Cl). In ICC explant clusters, [Cl(-)](i) was seen to fluctuate, possibly evoked by rhythmic changes in intracellular calcium. CONCLUSIONS & INFERENCES The intracellular chloride concentration in ICC fluctuates to keep its equilibrium potential constant. The identification of E(Cl) as positive to the resting membrane potential of ICC indicates that opening of chloride channels will depolarize ICC.
Collapse
Affiliation(s)
- Y Zhu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | | | | |
Collapse
|
522
|
Zimoń M, Baets J, Auer-Grumbach M, Berciano J, Garcia A, Lopez-Laso E, Merlini L, Hilton-Jones D, McEntagart M, Crosby AH, Barisic N, Boltshauser E, Shaw CE, Landouré G, Ludlow CL, Gaudet R, Houlden H, Reilly MM, Fischbeck KH, Sumner CJ, Timmerman V, Jordanova A, Jonghe PD. Dominant mutations in the cation channel gene transient receptor potential vanilloid 4 cause an unusual spectrum of neuropathies. Brain 2010; 133:1798-809. [PMID: 20460441 PMCID: PMC2912694 DOI: 10.1093/brain/awq109] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Revised: 03/23/2010] [Accepted: 04/01/2010] [Indexed: 11/13/2022] Open
Abstract
Hereditary neuropathies form a heterogeneous group of disorders for which over 40 causal genes have been identified to date. Recently, dominant mutations in the transient receptor potential vanilloid 4 gene were found to be associated with three distinct neuromuscular phenotypes: hereditary motor and sensory neuropathy 2C, scapuloperoneal spinal muscular atrophy and congenital distal spinal muscular atrophy. Transient receptor potential vanilloid 4 encodes a cation channel previously implicated in several types of dominantly inherited bone dysplasia syndromes. We performed DNA sequencing of the coding regions of transient receptor potential vanilloid 4 in a cohort of 145 patients with various types of hereditary neuropathy and identified five different heterozygous missense mutations in eight unrelated families. One mutation arose de novo in an isolated patient, and the remainder segregated in families. Two of the mutations were recurrent in unrelated families. Four mutations in transient receptor potential vanilloid 4 targeted conserved arginine residues in the ankyrin repeat domain, which is believed to be important in protein-protein interactions. Striking phenotypic variability between and within families was observed. The majority of patients displayed a predominantly, or pure, motor neuropathy with axonal characteristics observed on electrophysiological testing. The age of onset varied widely, ranging from congenital to late adulthood onset. Various combinations of additional features were present in most patients including vocal fold paralysis, scapular weakness, contractures and hearing loss. We identified six asymptomatic mutation carriers, indicating reduced penetrance of the transient receptor potential vanilloid 4 defects. This finding is relatively unusual in the context of hereditary neuropathies and has important implications for diagnostic testing and genetic counselling.
Collapse
Affiliation(s)
- Magdalena Zimoń
- 1 Neurogenetics Group, VIB Department of Molecular Genetics, University of Antwerp, 2610 Antwerpen, Belgium
- 2 Neurogenetics Laboratory, Institute Born-Bunge, University of Antwerp, 2610 Antwerpen, Belgium
| | - Jonathan Baets
- 1 Neurogenetics Group, VIB Department of Molecular Genetics, University of Antwerp, 2610 Antwerpen, Belgium
- 2 Neurogenetics Laboratory, Institute Born-Bunge, University of Antwerp, 2610 Antwerpen, Belgium
- 3 Division of Neurology, University Hospital Antwerp, 2650 Antwerpen, Belgium
| | - Michaela Auer-Grumbach
- 4 Institute of Medical Biology and Department of Internal Medicine, Diabetes and Metabolism, Medical University Graz, 8010 Graz, Austria
| | - José Berciano
- 5 Service of Neurology, University Hospital ‘Marqués de Valdecilla’, ‘Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)’ and University of Cantabria (UC), 39008 Santander, Spain
| | - Antonio Garcia
- 6 Service of Clinical Neurophysiology, University Hospital ‘Marqués de Valdecilla’, ‘Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED)’ and University of Cantabria (UC), 39008 Santander, Spain
| | - Eduardo Lopez-Laso
- 7 Paediatric Neurology Unit, Department of Paediatrics, University Hospital Reina Sofia, University of Córdoba, 14005 Córdoba, Spain
| | - Luciano Merlini
- 8 Muscle Unit, Division of Medical Genetics, Department of Experimental and Diagnostic Medicine, University of Ferrara, 44100 Ferrara, Italy
- 9 Laboratory of Musculoskeletal Cell Biology, IOR-IRCCS, 40136 Bologna, Italy
| | | | - Meriel McEntagart
- 11 Department of Medical Genetics, St. George’s Hospital, Medical School, SW17 0RE London, UK
| | - Andrew H. Crosby
- 11 Department of Medical Genetics, St. George’s Hospital, Medical School, SW17 0RE London, UK
| | - Nina Barisic
- 12 Department of Paediatrics, University of Zagreb, Medical School, University Hospital Centre Zagreb, 10000 Zagreb,Croatia
| | - Eugen Boltshauser
- 13 Department of Paediatric Neurology, University of Zürich, Children's Hospital, CH-8032 Zürich, Switzerland
| | - Christopher E. Shaw
- 14 MRC Centre for Neurodegeneration Research, Department of Clinical Neuroscience, King's College London, Institute of Psychiatry, SE5 8AF London, UK
| | - Guida Landouré
- 15 University College London, Departments of Medicine and Neuroscience, WC1N 3BG London, UK
- 16 Neurogenetics Branch, NINDS, NIH, 3705 Bethesda, MD, USA
| | - Christy L. Ludlow
- 17 Laboratory of Neural Bases of Communication and Swallowing, James Madison University, 22807 Harrisonburg, VA, USA
| | - Rachelle Gaudet
- 18 Department of Molecular and Cellular Biology, Harvard University, 02138 Cambridge, MA, USA
| | - Henry Houlden
- 19 Department of Molecular Neuroscience, UCL Institute of Neurology, WC1N 3BG London, UK
- 20 MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, WC1N 3BG London, UK
| | - Mary M. Reilly
- 20 MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, WC1N 3BG London, UK
| | | | - Charlotte J. Sumner
- 21 Department of Neurology, Johns Hopkins University, 21287 Baltimore, MD, USA
| | - Vincent Timmerman
- 2 Neurogenetics Laboratory, Institute Born-Bunge, University of Antwerp, 2610 Antwerpen, Belgium
- 22 Peripheral Neuropathy Group, VIB Department of Molecular Genetics, University of Antwerp, 2610 Antwerpen, Belgium
| | - Albena Jordanova
- 1 Neurogenetics Group, VIB Department of Molecular Genetics, University of Antwerp, 2610 Antwerpen, Belgium
- 2 Neurogenetics Laboratory, Institute Born-Bunge, University of Antwerp, 2610 Antwerpen, Belgium
| | - Peter De Jonghe
- 1 Neurogenetics Group, VIB Department of Molecular Genetics, University of Antwerp, 2610 Antwerpen, Belgium
- 2 Neurogenetics Laboratory, Institute Born-Bunge, University of Antwerp, 2610 Antwerpen, Belgium
- 3 Division of Neurology, University Hospital Antwerp, 2650 Antwerpen, Belgium
| |
Collapse
|
523
|
Interdomain interactions control Ca2+-dependent potentiation in the cation channel TRPV4. PLoS One 2010; 5:e10580. [PMID: 20485495 PMCID: PMC2867956 DOI: 10.1371/journal.pone.0010580] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Accepted: 04/20/2010] [Indexed: 11/24/2022] Open
Abstract
Several Ca2+-permeable channels, including the non-selective cation channel TRPV4, are subject to Ca2+-dependent facilitation. Although it has been clearly demonstrated in functional experiments that calmodulin (CaM) binding to intracellular domains of TRP channels is involved in this process, the molecular mechanism remains elusive. In this study, we provide experimental evidence for a comprehensive molecular model that explains Ca2+-dependent facilitation of TRPV4. In the resting state, an intracellular domain from the channel N terminus forms an autoinhibitory complex with a C-terminal domain that includes a high-affinity CaM binding site. CaM binding, secondary to rises in intracellular Ca2+, displaces the N-terminal domain which may then form a homologous interaction with an identical domain from a second subunit. This represents a novel potentiation mechanism that may also be relevant in other Ca2+-permeable channels.
Collapse
|
524
|
Abstract
The mammalian transient receptor potential (TRP) superfamily consists of six subfamilies that are defined by structural homology: TRPC (conventional or canonical), TRPV (vanilloid), TRPM (melastatin), TRPA (ankyrin), TRPP (polycystin), and TRPML (mucoliptin). This review focuses on channels belonging to the vanilloid (V) and melastatin (M) TRP subfamilies. The TRPV subfamily consists of six members (TRPV1-6) and the TRPM subfamily has eight (TRPM1-8). The basic biophysical properties of these channels are briefly described. All of these channels except TRPV5, TRPV6, and TRPM1 are reportedly present in arterial smooth muscle from various segments of the vasculature. Studies demonstrating involvement of TRPV1, TRPV2, TRPV4, TRPM4, TRPM7, and TRPM8 in regulation of arterial smooth muscle function are reviewed. The functions of TRPV3, TRPM2, TRPM3, and TRPM6 channels in arterial myocytes have not been reported.
Collapse
Affiliation(s)
- Scott Earley
- Vascular Physiology Research Group, Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
525
|
Shirakawa H, Nakagawa T, Kaneko S. [Pathophysiological roles of transient receptor potential channels in glial cells]. YAKUGAKU ZASSHI 2010; 130:281-7. [PMID: 20190511 DOI: 10.1248/yakushi.130.281] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glial cells are abundant in the CNS and play diverse roles in the regulation of neuronal activity, vascular function, and gliotransmitter release, whereas pathologically activated glial cells have been reported to disturb brain function in conjunction with Ca(2+) signaling, however there is no enough explanation for a unique Ca(2+) entry. Transient receptor potential (TRP) superfamily comprises a group of non-selective cation channels that open in response to divergent stimuli in their environment. Although TRP channels are widely distributed in the mammalian brain, their roles remain to be elucidated. Here we provide an overview of the roles of TRP channels in pathophysiological processes, especially focusing on TRPC3 and TRPV4 channels in glial cells. Using rat cortical astrocytes, we found that TRPC3 was upregulated by thrombin via Ca(2+) signaling through TRPC3 itself. Thrombin also upregulated S100B, a marker of reactive astrocytes, and increased cell proliferation, both of which were inhibited by Ca(2+) signaling blockers and specific knockdown of TRPC3 using siRNA, suggesting that TRPC3 contributes to the pathological activation of astrocytes in part through a feed-forward upregulation of its own expression. Moreover, we found that TRPV4 stimulation by its agonist 4alpha-PDD suppressed LPS-induced microglial activation while TRPV4 mRNA was downregulated in LPS-treated cultured rat microglia. These results suggest that TRP channels play pivotal roles in the process of astrocytic and microglial activation.
Collapse
Affiliation(s)
- Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | |
Collapse
|
526
|
Loukin S, Su Z, Zhou X, Kung C. Forward genetic analysis reveals multiple gating mechanisms of TRPV4. J Biol Chem 2010; 285:19884-90. [PMID: 20424166 DOI: 10.1074/jbc.m110.113936] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
TRPV4 is a polymodal cation channel gain-of-function (GOF) allele which causes skeletal dysplasia in humans. To better understand its gating, we screened for additional GOF alleles based on their ability to block yeast proliferation. Repeatedly, only a limited number of such growth-blocking mutations were isolated. Expressed in oocytes, wild-type channels can be strongly activated by either hypotonicity or exposure to the potent agonist 4alphaPDD, although the GOF channels behaved as if they were fully prestimulated as well as lacking a previously uncharacterized voltage-dependent inactivation. Five of six mutations occurred at or near the inner ends of the predicted core helices, giving further direct evidence that this region indeed forms the main intracellular gate in TRP channels. Surprisingly, both wild-type channels as well as these GOF channels maintain strong steady-state outward rectification that is not due to a Ca(2+) block, as has been proposed elsewhere. We conclude that TRPV4 contains an additional voltage-dependent gating mechanism in series with the main intracellular gate.
Collapse
Affiliation(s)
- Stephen Loukin
- Laboratory of Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA.
| | | | | | | |
Collapse
|
527
|
Patwardhan AM, Akopian AN, Ruparel NB, Diogenes A, Weintraub ST, Uhlson C, Murphy RC, Hargreaves KM. Heat generates oxidized linoleic acid metabolites that activate TRPV1 and produce pain in rodents. J Clin Invest 2010; 120:1617-26. [PMID: 20424317 DOI: 10.1172/jci41678] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 03/17/2010] [Indexed: 12/28/2022] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1) channel is the principal detector of noxious heat in the peripheral nervous system. TRPV1 is expressed in many nociceptors and is involved in heat-induced hyperalgesia and thermoregulation. The precise mechanism or mechanisms mediating the thermal sensitivity of TRPV1 are unknown. Here, we have shown that the oxidized linoleic acid metabolites 9- and 13-hydroxyoctadecadienoic acid (9- and 13-HODE) are formed in mouse and rat skin biopsies by exposure to noxious heat. 9- and 13-HODE and their metabolites, 9- and 13-oxoODE, activated TRPV1 and therefore constitute a family of endogenous TRPV1 agonists. Moreover, blocking these substances substantially decreased the heat sensitivity of TRPV1 in rats and mice and reduced nociception. Collectively, our results indicate that HODEs contribute to the heat sensitivity of TRPV1 in rodents. Because oxidized linoleic acid metabolites are released during cell injury, these findings suggest a mechanism for integrating the hyperalgesic and proinflammatory roles of TRPV1 and linoleic acid metabolites and may provide the foundation for investigating new classes of analgesic drugs.
Collapse
Affiliation(s)
- Amol M Patwardhan
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78227, USA
| | | | | | | | | | | | | | | |
Collapse
|
528
|
Sokabe T, Fukumi-Tominaga T, Yonemura S, Mizuno A, Tominaga M. The TRPV4 channel contributes to intercellular junction formation in keratinocytes. J Biol Chem 2010; 285:18749-58. [PMID: 20413591 DOI: 10.1074/jbc.m110.103606] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channel is a physiological sensor for hypo-osmolarity, mechanical deformation, and warm temperature. The channel activation leads to various cellular effects involving Ca(2+) dynamics. We found that TRPV4 interacts with beta-catenin, a crucial component linking adherens junctions and the actin cytoskeleton, thereby enhancing cell-cell junction development and formation of the tight barrier between skin keratinocytes. TRPV4-deficient mice displayed impairment of the intercellular junction-dependent barrier function in the skin. In TRPV4-deficient keratinocytes, extracellular Ca(2+)-induced actin rearrangement and stratification were delayed following significant reduction in cytosolic Ca(2+) increase and small GTPase Rho activation. TRPV4 protein located where the cell-cell junctions are formed, and the channel deficiency caused abnormal cell-cell junction structures, resulting in higher intercellular permeability in vitro. Our results suggest a novel role for TRPV4 in the development and maturation of cell-cell junctions in epithelia of the skin.
Collapse
Affiliation(s)
- Takaaki Sokabe
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki 444-8787, Japan.
| | | | | | | | | |
Collapse
|
529
|
Edwards G, Félétou M, Weston AH. Endothelium-derived hyperpolarising factors and associated pathways: a synopsis. Pflugers Arch 2010; 459:863-79. [PMID: 20383718 DOI: 10.1007/s00424-010-0817-1] [Citation(s) in RCA: 285] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 02/22/2010] [Accepted: 02/24/2010] [Indexed: 12/29/2022]
Abstract
The term endothelium-derived hyperpolarising factor (EDHF) was introduced in 1987 to describe the hypothetical factor responsible for myocyte hyperpolarisations not associated with nitric oxide (EDRF) or prostacyclin. Two broad categories of EDHF response exist. The classical EDHF pathway is blocked by apamin plus TRAM-34 but not by apamin plus iberiotoxin and is associated with endothelial cell hyperpolarisation. This follows an increase in intracellular [Ca(2+)] and the opening of endothelial SK(Ca) and IK(Ca) channels preferentially located in caveolae and in endothelial cell projections through the internal elastic lamina, respectively. In some vessels, endothelial hyperpolarisations are transmitted to myocytes through myoendothelial gap junctions without involving any EDHF. In others, the K(+) that effluxes through SK(Ca) activates myocytic and endothelial Ba(2+)-sensitive K(IR) channels leading to myocyte hyperpolarisation. K(+) effluxing through IK(Ca) activates ouabain-sensitive Na(+)/K(+)-ATPases generating further myocyte hyperpolarisation. For the classical pathway, the hyperpolarising "factor" involved is the K(+) that effluxes through endothelial K(Ca) channels. During vessel contraction, K(+) efflux through activated myocyte BK(Ca) channels generates intravascular K(+) clouds. These compromise activation of Na(+)/K(+)-ATPases and K(IR) channels by endothelium-derived K(+) and increase the importance of gap junctional electrical coupling in myocyte hyperpolarisations. The second category of EDHF pathway does not require endothelial hyperpolarisation. It involves the endothelial release of factors that include NO, HNO, H(2)O(2) and vasoactive peptides as well as prostacyclin and epoxyeicosatrienoic acids. These hyperpolarise myocytes by opening various populations of myocyte potassium channels, but predominantly BK(Ca) and/or K(ATP), which are sensitive to blockade by iberiotoxin or glibenclamide, respectively.
Collapse
Affiliation(s)
- Gillian Edwards
- Faculty of Life Sciences, University of Manchester, CTF Building, 46 Grafton St, Manchester, M13 9NT, UK
| | | | | |
Collapse
|
530
|
Abstract
TRP (transient receptor potential) channels play important roles in the regulation of normal and pathological cellular function. In the vasculature, TRP channels are present both in ECs (endothelial cells) and vascular SMCs (smooth muscle cells) and contribute to vasomotor control mechanisms in most vascular beds. Vascular TRP channels are activated by various stimuli, such as mechanical perturbation, receptor activation and dietary molecules. Some of the specific roles of these channels in normal and impaired vascular function have emerged in recent years and include participation in vascular signalling processes, such as neurotransmission, hormonal signalling, NO production, myogenic tone and autoregulation of blood flow, thermoregulation, responses to oxidative stress and cellular proliferative activity. Current research is aimed at understanding the interactions of TRP channels with other vascular proteins and signalling mechanisms. These studies should reveal new targets for pharmacological therapy of vascular diseases, such as hypertension, ischaemia and vasospasm, and vascular proliferative states.
Collapse
|
531
|
Iliff JJ, Jia J, Nelson J, Goyagi T, Klaus J, Alkayed NJ. Epoxyeicosanoid signaling in CNS function and disease. Prostaglandins Other Lipid Mediat 2010; 91:68-84. [PMID: 19545642 PMCID: PMC2844927 DOI: 10.1016/j.prostaglandins.2009.06.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 06/09/2009] [Accepted: 06/10/2009] [Indexed: 10/20/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) are arachidonic acid metabolites of cytochrome P450 epoxygenase enzymes recognized as key players in vascular function and disease, primarily attributed to their potent vasodilator, anti-inflammatory and pro-angiogenic effects. Although EETs' actions in the central nervous system (CNS) appear to parallel those in peripheral tissue, accumulating evidence suggests that epoxyeicosanoid signaling plays different roles in neural tissue compared to peripheral tissue; roles that reflect distinct CNS functions, cellular makeup and intercellular relationships. This is exhibited at many levels including the expression of EETs-synthetic and -metabolic enzymes in central neurons and glial cells, EETs' role in neuro-glio-vascular coupling during cortical functional activation, the capacity for interaction between epoxyeicosanoid and neuroactive endocannabinoid signaling pathways, and the regulation of neurohormone and neuropeptide release by endogenous EETs. The ability of several CNS cell types to produce and respond to EETs suggests that epoxyeicosanoid signaling is a key integrator of cell-cell communication in the CNS, coordinating cellular responses across different cell types. Under pathophysiological conditions, such as cerebral ischemia, EETs protect neurons, astroglia and vascular endothelium, thus preserving the integrity of cellular networks unique to and essential for proper CNS function. Recognition of EETs' intimate involvement in CNS function in addition to their multi-cellular protective profile has inspired the development of therapeutic strategies against CNS diseases such as cerebral ischemia, tumors, and neural pain and inflammation that are based on targeting the cellular actions of EETs or their biosynthetic and metabolizing enzymes. Based upon the emerging importance of epoxyeicosanoids in cellular function and disease unique to neural systems, we propose that the actions of "neuroactive EETs" are best considered separately, and not in aggregate with all other peripheral EETs functions.
Collapse
Affiliation(s)
- Jeffrey J Iliff
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | | | |
Collapse
|
532
|
Giuffrida A, McMahon LR. In vivo pharmacology of endocannabinoids and their metabolic inhibitors: therapeutic implications in Parkinson's disease and abuse liability. Prostaglandins Other Lipid Mediat 2010; 91:90-103. [PMID: 19523530 PMCID: PMC2844932 DOI: 10.1016/j.prostaglandins.2009.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 05/19/2009] [Accepted: 05/26/2009] [Indexed: 01/03/2023]
Abstract
This review focuses on the behavioral pharmacology of endogenous cannabinoids (endocannabinoids) and indirect-acting cannabinoid agonists that elevate endocannabinoid tone by inhibiting the activity of metabolic enzymes. Similarities and differences between prototype cannabinoid agonists, endocannabinoids and inhibitors of endocannabinoid metabolism are discussed in the context of endocannabinoid pharmacokinetics in vivo. The distribution and function of cannabinoid and non-CB(1)/CB(2) receptors are also covered, with emphasis on their role in disorders characterized by dopamine dysfunction, such as drug abuse and Parkinson's disease. Finally, evidence is presented to suggest that FAAH inhibitors lack the abuse liability associated with CB(1) agonists, although they may modify the addictive properties of other drugs, such as alcohol.
Collapse
Affiliation(s)
- Andrea Giuffrida
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | | |
Collapse
|
533
|
Gao F, Wang DH. Impairment in function and expression of transient receptor potential vanilloid type 4 in Dahl salt-sensitive rats: significance and mechanism. Hypertension 2010; 55:1018-25. [PMID: 20194297 PMCID: PMC2862636 DOI: 10.1161/hypertensionaha.109.147710] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 02/02/2010] [Indexed: 01/18/2023]
Abstract
To examine the role of transient receptor potential vanilloid type 4 (TRPV4) channels in the development of salt-sensitive hypertension, male Dahl salt-sensitive (DS) and -resistant (DR) rats were fed a low-salt (LS) or high-salt (HS) diet for 3 weeks. DS-HS but not DR-HS rats developed hypertension. 4alpha-Phorbol-12,13-didecanoate (a selective TRPV4 activator; 2.5 mg/kg IV) decreased mean arterial pressure in all of the groups with the greatest effects in DR-HS and the least in DS-HS rats (P<0.05). Depressor effects of 4alpha-phorbol-12,13-didecanoate but not dihydrocapsaicin (a selective TRPV1 agonist; 30 microg/kg IV) were abolished by ruthenium red (a TRPV4 antagonist; 3 mg/kg IV) in all of the groups. Blockade of TRPV4 with ruthenium red increased mean arterial pressure in DR-HS rats only (P<0.05). TRPV4 protein contents were decreased in the renal cortex, medulla, and dorsal root ganglia in DS-HS compared with DS-LS rats but increased in dorsal root ganglia and mesenteric arteries in DR-HS compared with DR-LS rats (P<0.05). Mean arterial pressure responses to blockade of small- and large-/intermediate-conductance Ca(2+)-activated K(+) channels (Maxikappa channels) with apamin and charybdotoxin, respectively, were examined. Apamin (100 microg/kg) plus charybdotoxin (100 microg/kg) abolished 4alpha-phorbol-12,13-didecanoate-induced hypotension in DR-LS, DR-HS, and DS-LS rats only. Thus, HS-induced enhancement of TRPV4 function and expression in sensory neurons and resistant vessels in DR rats may prevent salt-induced hypertension possibly via activation of Maxikappa channels given that blockade of TRPV4 elevates mean arterial pressure. In contrast, HS-induced suppression of TRPV4 function and expression in sensory neurons and kidneys in DS rats may contribute to increased salt sensitivity.
Collapse
Affiliation(s)
- Feng Gao
- Department of Medicine, Michigan State University, Michigan
| | - Donna H. Wang
- Department of Medicine, Michigan State University, Michigan
- Neuroscience Program, Michigan State University, Michigan
- Cell and Molecular Biology Program, Michigan State University, Michigan
| |
Collapse
|
534
|
Weick JP, Austin Johnson M, Zhang SC. Developmental regulation of human embryonic stem cell-derived neurons by calcium entry via transient receptor potential channels. Stem Cells 2010; 27:2906-16. [PMID: 19725137 DOI: 10.1002/stem.212] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Spontaneous calcium (Ca(2+)) transients in the developing nervous system can affect proliferation, migration, neuronal subtype specification, and neurite outgrowth. Here, we show that telencephalic human neuroepithelia (hNE) and postmitotic neurons (PMNs) generated from embryonic stem cells display robust Ca(2+) transients. Unlike previous reports in animal models, transients occurred by a Gd(3+)/La(3+)-sensitive, but thapsigargin- and Cd(2+)-insensitive, mechanism, strongly suggestive of a role for transient receptor potential (Trp) channels. Furthermore, Ca(2+) transients in PMNs exhibited an additional sensitivity to the canonical Trp (TrpC) antagonist SKF96365 and shRNA-mediated knockdown of the TrpC1 subunit. Functionally, inhibition of Ca(2+) transients in dividing hNE cells led to a significant reduction in proliferation, whereas either pharmacological inhibition or shRNA-mediated knockdown of the TrpC1 and TrpC4 subunits significantly reduced neurite extension in PMNs. Primary neurons cultured from fetal human cortex displayed nearly identical Ca(2+) transients and pharmacological sensitivities to Trp channel antagonists. Together these data suggest that Trp channels present a novel mechanism for controlling Ca(2+) transients in human neurons and may offer a target for regulating proliferation and neurite outgrowth when engineering cells for therapeutic transplantation.
Collapse
Affiliation(s)
- Jason P Weick
- Waisman Center, University of Wisconsin, Madison, Wisconsin 53705, USA
| | | | | |
Collapse
|
535
|
Campbell WB, Fleming I. Epoxyeicosatrienoic acids and endothelium-dependent responses. Pflugers Arch 2010; 459:881-95. [PMID: 20224870 DOI: 10.1007/s00424-010-0804-6] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Revised: 02/08/2010] [Accepted: 02/12/2010] [Indexed: 12/28/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) are cytochrome P450 metabolites of arachidonic acid that are produced by the vascular endothelium in response to agonists such as bradykinin and acetylcholine or physical stimuli such as shear stress or cyclic stretch. In the vasculature, the EETs have biological actions that are involved in the regulation of vascular tone, hemostasis, and inflammation. In preconstricted arteries in vitro, EETs activate calcium-activated potassium channels on vascular smooth muscle and the endothelium causing membrane hyperpolarization and relaxation. These effects are observed in a variety of arteries from experimental animals and humans; however, this is not a universal finding in all arteries. The mechanism of EET action may vary. In some arteries, EETs are released from the endothelium and are transferred to the smooth muscle where they cause potassium channel activation, hyperpolarization, and relaxation through a guanine nucleotide binding protein-coupled mechanism or transient receptor potential (TRP) channel activation. In other arteries, EETs activate TRP channels on the endothelium to cause endothelial hyperpolarization that is transferred to the smooth muscle by gap junctions or potassium ion. Some arteries use a combination of mechanisms. Acetylcholine and bradykinin increase blood flow in dogs and humans that is inhibited by potassium channel blockers and cytochrome P450 inhibitors. Thus, the EETs are endothelium-derived hyperpolarizing factors mediating a portion of the relaxations to acetylcholine, bradykinin, shear stress, and cyclic stretch and regulate vascular tone in vitro and in vivo.
Collapse
Affiliation(s)
- William B Campbell
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | | |
Collapse
|
536
|
Hypotension induced by activation of the transient receptor potential vanilloid 4 channels: role of Ca2+-activated K+ channels and sensory nerves. J Hypertens 2010; 28:102-10. [PMID: 19996988 DOI: 10.1097/hjh.0b013e328332b865] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To examine the mechanisms involved in hypotension induced by transient receptor potential vanilloid 4 (TRPV4) activation. METHODS Wistar rats were given 50 mg/kg capsaicin subcutaneously 1-2 days postnatally to cause degeneration of capsaicin-sensitive sensory nerves. Vehicle was given to the corresponding newborn rats that formed the control group. After being weaned, male rats were picked for further investigation. At the age of 8 weeks, mean arterial pressure and its response to 4alpha-phorbol 12,13-didecanoate [4alpha-PDD, a selective TRPV4 activator, 2.5 mg/kg, intravenous(ly) or i.v.] with or without CGRP8-37 (1 mg/kg per min, i.v.), an antagonist of calcitonin gene-related peptide (CGRP, a potent vasodilator released from sensory nerves), in vehicle or capsaicin-pretreated rats anesthetized with sodium pentobarbital [50 mg/kg, intraperitoneal(ly)] were monitored to observe the contributions of neuropeptides released from sensory nerves to the 4alpha-PDD-induced hypotension. To detect the roles of various vasodilating factors released by vascular endothelium in the hypotensive effect induced by TRPV4 activation, the corresponding inhibitors/blockers, including indomethacin (a cyclooxygenase inhibitor, 10 mg/kg, i.v.), Nomega-nitro-L-arginine (L-NA, a nitric oxide synthase inhibitor, 20 mg/kg, i.v.), apamin [a blocker of small conductance Ca2+-activated K+ (MaxiK) channels, 50 microg/kg, i.v.] combined with charybdotoxin (a blocker of intermediate and large conductance MaxiK channels, 50 microg/kg, i.v.), were used at various time before 4alpha-PDD injection. Plasma CGRP and substance P levels of rats before or after administration were measured using the corresponding radioimmunoassays. At last, immunohistochemistry stainings were performed to observe expression of TRPV4/CGRP/MaxiK in mesenteric resistance arteries and sensory neurons/nerve fibers. RESULTS Intravenous administration of 4alpha-PDD produced remarkable hypotension in vehicle-pretreated rats. The depressor effect was attenuated by degeneration of capsaicin-sensitive sensory nerves (P < 0.05) or administration of CGRP8-37 (P < 0.05). In both vehicle and capsaicin-pretreated rats, the combined administration of apamin and charybdotoxin markedly reduced the 4alpha-PDD-induced hypotensive effect (P < 0.05), but i.v. administration of indomethacin and Nomega-nitro-L-arginine did not produce the similar effect. Intravenous administration of 4alpha PDD increased plasma CGRP but not substance P levels in vehicle-pretreated rats only (P < 0.05), which was not affected by indomethacin, Nomega-nitro-L-arginine, or apamin and charybdotoxin. Immunohistochemistry staining showed that TRPV4 colocalized with MaxiK channels in endothelium of mesenteric resistance arteries and with CGRP in sensory neurons/nerve fibers. CONCLUSION Our data show that the hypotensive effect induced by TRPV4 activation attributes to, at least in part, activation of MaxiK channels and CGRP receptors upon CGRP release from sensory nerves.
Collapse
|
537
|
Snider NT, Walker VJ, Hollenberg PF. Oxidation of the endogenous cannabinoid arachidonoyl ethanolamide by the cytochrome P450 monooxygenases: physiological and pharmacological implications. Pharmacol Rev 2010; 62:136-54. [PMID: 20133390 PMCID: PMC2835397 DOI: 10.1124/pr.109.001081] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Arachidonoyl ethanolamide (anandamide) is an endogenous amide of arachidonic acid and an important signaling mediator of the endocannabinoid system. Given its numerous roles in maintaining normal physiological function and modulating pathophysiological responses throughout the body, the endocannabinoid system is an important pharmacological target amenable to manipulation directly by cannabinoid receptor ligands or indirectly by drugs that alter endocannabinoid synthesis and inactivation. The latter approach has the possible advantage of more selectivity, thus there is the potential for fewer untoward effects like those that are traditionally associated with cannabinoid receptor ligands. In that regard, inhibitors of the principal inactivating enzyme for anandamide, fatty acid amide hydrolase (FAAH), are currently in development for the treatment of pain and inflammation. However, several pathways involved in anandamide synthesis, metabolism, and inactivation all need to be taken into account when evaluating the effects of FAAH inhibitors and similar agents in preclinical models and assessing their clinical potential. Anandamide undergoes oxidation by several human cytochrome P450 (P450) enzymes, including CYP3A4, CYP4F2, CYP4X1, and the highly polymorphic CYP2D6, forming numerous structurally diverse lipids, which are likely to have important physiological roles, as evidenced by the demonstration that a P450-derived epoxide of anandamide is a potent agonist for the cannabinoid receptor 2. The focus of this review is to emphasize the need for a better understanding of the P450-mediated pathways of the metabolism of anandamide, because these are likely to be important in mediating endocannabinoid signaling as well as the pharmacological responses to endocannabinoid-targeting drugs.
Collapse
Affiliation(s)
- Natasha T Snider
- Department of Molecular & Integrative Physiology, University of Michigan School of Medicine, 7720 Medical Science II, 1301 E. Catherine Street, Ann Arbor, MI 48109-5622, USA.
| | | | | |
Collapse
|
538
|
Townsley MI, Morisseau C, Hammock B, King JA. Impact of epoxyeicosatrienoic acids in lung ischemia-reperfusion injury. Microcirculation 2010; 17:137-46. [PMID: 20163540 PMCID: PMC2923642 DOI: 10.1111/j.1549-8719.2009.00013.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Epoxyeicosatrienoic acids (EETs) are protective in both myocardial and brain ischemia, variously attributed to activation of K(ATP) channels or blockade of adhesion molecule upregulation. In this study, we tested whether EETs would be protective in lung ischemia-reperfusion injury. METHODS The filtration coefficient (K(f)), a measure of endothelial permeability, and expression of the adhesion molecules vascular cell adhesion molecule (VCAM) and intercellular adhesion molecule (ICAM) were measured after 45 minutes ischemia and 30 minutes reperfusion in isolated rat lungs. RESULTS K(f) increased significantly after ischemia-reperfusion alone vs time controls, an effect dependent upon extracellular Ca(2+) although not on the EET-regulated channel TRPV4. Inhibition of endogenous EET degradation or administration of exogenous 11,12- or 14,-15-EET at reperfusion significantly limited the permeability response to ischemia-reperfusion. The beneficial effect of 11,12-EET was not prevented by blockade of K(ATP) channels nor by blockade of TRPV4. Finally, 11,12-EET-dependent alteration in adhesion molecules expression is unlikely to explain its beneficial effect, since the expression of the adhesion molecules VCAM and ICAM in lung after ischemia-reperfusion was similar to that in controls. CONCLUSION EETs are beneficial in the setting of lung ischemia-reperfusion, when administered at reperfusion. However, further study will be needed to elucidate the mechanism of action.
Collapse
Affiliation(s)
- Mary I Townsley
- Department of Physiology, Center for Lung Biology, University of South Alabama, Mobile, AL 36688, USA.
| | | | | | | |
Collapse
|
539
|
Abstract
Cutaneous thermosensation plays an important role in thermal regulation and detection of potentially harmful thermal stimuli. Multiple classes of primary afferents are responsive to thermal stimuli. Afferent nerve fibers mediating the sensation of non-painful warmth or cold seem adapted to convey thermal information over a particular temperature range. In contrast, nociceptive afferents are often activated by both, painful cold and heat stimuli. The transduction mechanisms engaged by thermal stimuli have only recently been discovered. Transient receptor potential (TRP) ion channels that can be activated by temperatures over specific ranges potentially provide the molecular basis for thermosensation. However, non-TRP mechanisms are also likely to contribute to the transduction of thermal stimuli. This review summarizes findings regarding the transduction proteins and the primary afferents activated by innocuous and noxious cold and heat.
Collapse
Affiliation(s)
- Raf J Schepers
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | | |
Collapse
|
540
|
Introduction to TRP channels: structure, function, and regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 661:99-108. [PMID: 20204725 DOI: 10.1007/978-1-60761-500-2_6] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Transient receptor potential or TRP families of ion channels demonstrate great diversity in activation and inhibition, and they are diverse in selectivity of ion conductance. TRP ion channels function as signal integrators through their ion conductance properties, and in some cases kinase activity. They mediate processes such as vision, taste, olfaction, hearing, touch, and thermo- and osmosensation. TRP cation channels function by mediating the flux of Na(+) and Ca(2+) across the plasma membrane and into the cytoplasm. The influx of cations into the cytoplasm depolarizes cells and is necessary for action potentials in excitable cells such as neurons. In non-excitable cells, membrane depolarization by TRP ) and-channels stimulates voltage- dependent channels (Ca(2+), K(+), Cl(-) influences many cellular events, such as transcription, translation, contraction, and migration. TRP channels are important in human physiology, and mutations in TRP genes are associated with at least four diseases. Furthermore, altered expression, function, and/or regulation of TRP channels have been implicated in diseases such as pulmonary hypertension.
Collapse
|
541
|
Phospholipase A2-derived lysophosphatidylcholine triggers Ca2+ entry in dystrophic skeletal muscle fibers. Biochem Biophys Res Commun 2010; 391:401-6. [DOI: 10.1016/j.bbrc.2009.11.070] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 11/10/2009] [Indexed: 11/24/2022]
|
542
|
Shigematsu H, Sokabe T, Danev R, Tominaga M, Nagayama K. A 3.5-nm structure of rat TRPV4 cation channel revealed by Zernike phase-contrast cryoelectron microscopy. J Biol Chem 2009; 285:11210-8. [PMID: 20044482 DOI: 10.1074/jbc.m109.090712] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) is a non-selective cation channel responsive to various stimuli including cell swelling, warm temperatures (27-35 degrees C), and chemical compounds such as phorbol ester derivatives. Here we report the three-dimensional structure of full-length rat TRPV4 purified from baculovirus-infected Sf9 cells. Hexahistidine-tagged rat TRPV4 (His-rTRPV4) was solubilized with detergent and purified through affinity chromatography and size-exclusion chromatography. Chemical cross-linking analysis revealed that detergent-solubilized His-rTRPV4 was a tetramer. The 3.5-nm structure of rat TRPV4 was determined by cryoelectron microscopy using single-particle reconstruction from Zernike phase-contrast images. The overall structure comprises two distinct regions; a larger dense component, likely corresponding to the cytoplasmic N- and C-terminal regions, and a smaller component corresponding to the transmembrane region.
Collapse
Affiliation(s)
- Hideki Shigematsu
- Division of Nano-Structure Physiology, Okazaki Institute for Integrative Bioscience, Higashiyama, Myodaiji, Okazaki 444-8787 Japan
| | | | | | | | | |
Collapse
|
543
|
Mutations in TRPV4 cause Charcot-Marie-Tooth disease type 2C. Nat Genet 2009; 42:170-4. [PMID: 20037586 DOI: 10.1038/ng.512] [Citation(s) in RCA: 232] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 12/03/2009] [Indexed: 01/07/2023]
Abstract
Charcot-Marie-Tooth disease type 2C (CMT2C) is an autosomal dominant neuropathy characterized by limb, diaphragm and laryngeal muscle weakness. Two unrelated families with CMT2C showed significant linkage to chromosome 12q24.11. We sequenced all genes in this region and identified two heterozygous missense mutations in the TRPV4 gene, C805T and G806A, resulting in the amino acid substitutions R269C and R269H. TRPV4 is a well-known member of the TRP superfamily of cation channels. In TRPV4-transfected cells, the CMT2C mutations caused marked cellular toxicity and increased constitutive and activated channel currents. Mutations in TRPV4 were previously associated with skeletal dysplasias. Our findings indicate that TRPV4 mutations can also cause a degenerative disorder of the peripheral nerves. The CMT2C-associated mutations lie in a distinct region of the TRPV4 ankyrin repeats, suggesting that this phenotypic variability may be due to differential effects on regulatory protein-protein interactions.
Collapse
|
544
|
Benfenati V, Ferroni S. Water transport between CNS compartments: functional and molecular interactions between aquaporins and ion channels. Neuroscience 2009; 168:926-40. [PMID: 20026249 DOI: 10.1016/j.neuroscience.2009.12.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Revised: 12/04/2009] [Accepted: 12/05/2009] [Indexed: 12/16/2022]
Abstract
The physiological ability of the mammalian CNS to integrate peripheral stimuli and to convey information to the body is tightly regulated by its capacity to preserve the ion composition and volume of the perineuronal milieu. It is well known that astroglial syncytium plays a crucial role in such process by controlling the homeostasis of ions and water through the selective transmembrane movement of inorganic and organic molecules and the equilibration of osmotic gradients. Astrocytes, in fact, by contacting neurons and cells lining the fluid-filled compartments, are in a strategic position to fulfill this role. They are endowed with ion and water channel proteins that are localized in specific plasma membrane domains facing diverse liquid spaces. Recent data in rodents have demonstrated that the precise dynamics of the astroglia-mediated homeostatic regulation of the CNS is dependent on the interactions between water channels and ion channels, and their anchoring with proteins that allow the formation of macromolecular complexes in specific cellular domains. Interplay can occur with or without direct molecular interactions suggesting the existence of different regulatory mechanisms. The importance of molecular and functional interactions is pinpointed by the numerous observations that as consequence of pathological insults leading to the derangement of ion and volume homeostasis the cell surface expression and/or polarized localization of these proteins is perturbed. Here, we critically discuss the experimental evidence concerning: (1) molecular and functional interplay of aquaporin 4, the major aquaporin protein in astroglial cells, with potassium and gap-junctional channels that are involved in extracellular potassium buffering. (2) the interactions of aquaporin 4 with chloride and calcium channels regulating cell volume homeostasis. The relevance of the crosstalk between water channels and ion channels in the pathogenesis of astroglia-related acute and chronic diseases of the CNS is also briefly discussed.
Collapse
Affiliation(s)
- V Benfenati
- Istituto per lo Studio dei Materiali Nanostrutturati, ISMN, National Research Council, Via Gobetti 101, 40129 Bologna, Italy
| | | |
Collapse
|
545
|
Everaerts W, Vriens J, Owsianik G, Appendino G, Voets T, De Ridder D, Nilius B. Functional characterization of transient receptor potential channels in mouse urothelial cells. Am J Physiol Renal Physiol 2009; 298:F692-701. [PMID: 20015940 DOI: 10.1152/ajprenal.00599.2009] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The bladder urothelium is currently believed to be a sensory structure, contributing to mechano- and chemosensation in the bladder. Transient receptor potential (TRP) cation channels act as polymodal sensors and may underlie some of the receptive properties of urothelial cells. However, the exact TRP channel expression profile of urothelial cells is unclear. In this study, we have performed a systematic analysis of the molecular and functional expression of various TRP channels in mouse urothelium. Urothelial cells from control and trpv4-/- mice were isolated, cultured (12-48 h), and used for quantitative real-time PCR, immunocytochemistry, calcium imaging, and whole cell patch-clamp experiments. At the mRNA level, TRPV4, TRPV2, and TRPM7 were the most abundantly expressed TRP genes. Immunohistochemistry showed a clear expression of TRPV4 in the plasma membrane, whereas TRPV2 was more prominent in the cytoplasm. TRPM7 was detected in the plasma membrane as well as cytoplasmic vesicles. Calcium imaging and patch-clamp experiments using TRP channel agonists and antagonists provided evidence for the functional expression of TRPV4, TRPV2, and TRPM7 but not of TRPA1, TRPV1, and TRPM8. In conclusion, we have demonstrated functional expression of TRPV4, TRPV2, and TRPM7 in mouse urothelial cells. These channels may contribute to the (mechano)sensory function of the urothelial layer and represent potential targets for the treatment of bladder dysfunction.
Collapse
Affiliation(s)
- Wouter Everaerts
- Department of Molecular Cell Biology, Laboratory Ion Channel Research, Campus Gasthuisberg, Katholieke Universiteit Leuven, Leuven, Belgium.
| | | | | | | | | | | | | |
Collapse
|
546
|
Earley S, Brayden J, Reading S. Functional Significance of Transient Receptor Potential Channels in Vascular Function. ACTA ACUST UNITED AC 2009. [DOI: 10.1201/9781420005844.ch26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
547
|
|
548
|
Mendoza SA, Fang J, Gutterman DD, Wilcox DA, Bubolz AH, Li R, Suzuki M, Zhang DX. TRPV4-mediated endothelial Ca2+ influx and vasodilation in response to shear stress. Am J Physiol Heart Circ Physiol 2009; 298:H466-76. [PMID: 19966050 DOI: 10.1152/ajpheart.00854.2009] [Citation(s) in RCA: 272] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transient receptor potential vallinoid type 4 (TRPV4) channel has been implicated in the endothelial shear response and flow-mediated dilation, although the precise functions of this channel remain poorly understood. In the present study, we investigated the role of TRPV4 in shear stress-induced endothelial Ca(2+) entry and the potential link between this signaling response and relaxation of small resistance arteries. Using immunohistochemical analysis and RT-PCR, we detected strong expression of TRPV4 protein and mRNA in the endothelium in situ and endothelial cells freshly isolated from mouse small mesenteric arteries. The selective TRPV4 agonist GSK1016790A increased endothelial Ca(2+) and induced potent relaxation of small mesenteric arteries from wild-type (WT) but not TRPV4(-/-) mice. Luminal flow elicited endothelium-dependent relaxations that involved both nitric oxide and EDHFs. Both nitric oxide and EDHF components of flow-mediated relaxation were markedly reduced in TRPV4(-/-) mice compared with WT controls. Using a fura-2/Mn(2+) quenching assay, shear was observed to produce rapid Ca(2+) influx in endothelial cells, which was markedly inhibited by the TRPV4 channel blocker ruthenium red and TRPV4-specific short interfering RNA. Flow elicited a similar TRPV4-mediated Ca(2+) entry in HEK-293 cells transfected with TRPV4 channels but not in nontransfected cells. Collectively, these data indicate that TRPV4 may be a potential candidate of mechanosensitive channels in endothelial cells through which the shear stimulus is transduced into Ca(2+) signaling, leading to the release of endothelial relaxing factors and flow-mediated dilation of small resistance arteries.
Collapse
Affiliation(s)
- Suelhem A Mendoza
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | |
Collapse
|
549
|
Ding XL, Wang YH, Ning LP, Zhang Y, Ge HY, Jiang H, Wang R, Yue SW. Involvement of TRPV4-NO-cGMP-PKG pathways in the development of thermal hyperalgesia following chronic compression of the dorsal root ganglion in rats. Behav Brain Res 2009; 208:194-201. [PMID: 19948193 DOI: 10.1016/j.bbr.2009.11.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2009] [Revised: 11/18/2009] [Accepted: 11/20/2009] [Indexed: 12/18/2022]
Abstract
The aim of the present study was to test the hypothesis that the TRPV4-NO-cGMP-PKG cascade is involved in the maintenance of thermal hyperalgesia following chronic compression of the dorsal root ganglion (DRG) (the procedure hereafter termed CCD) in rats. CCD rats showed thermal hyperalgesia and increased nitrite production. Intrathecal administration of ruthenium red (TRPV4 antagonist, 0.1-1 nmol), TRPV4 antisense ODN (TRPV4 AS, 40 microg, daily for 7 days), N(G)-L-nitro-arginine methyl ester (l-NAME, inhibitor of NO synthase, 30-300 nmol), 1H-[1,2,4]-oxadiazolo [4,3-a] quinoxalin-1-one (ODQ, a soluble guanylate cyclase inhibitor, 50-100 nmol) or 8-(4-Chlorophenylthio) guanosine 3',5'-cyclic Monophosphothioate, Rp-Isomer sodium salt (Rp-8-pCPT-cGMPS, a PKG inhibitor, 25-50 nmol) induced a significant (P<0.001) and dose-dependent increase in the paw withdrawal latency (PWL) compared with control rats, respectively. Ruthenium red (1 nmol), TRPV4 AS (40 microg, daily for 7 days) or L-NAME (300 nmol) decreased nitrite (an index of nitric oxide formation) in the DRG of CCD rats. In addition, the phorbol ester 4alpha-phorbol 12,13-didecanoate (4alpha-PDD, TRPV4 synthetic activator, 1 nmol), co-administered with L-NAME (300 nmol), attenuated the suppressive effect of L-NAME on CCD-induced thermal hyperalgesia and nitrite production. Our data suggested that the TRPV4-NO-cGMP-PKG pathway could be involved in CCD-induced thermal hyperalgesia.
Collapse
Affiliation(s)
- Xin-Li Ding
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Medical School of Shandong University, Jinan 250012, China
| | | | | | | | | | | | | | | |
Collapse
|
550
|
Phan MN, Leddy HA, Votta BJ, Kumar S, Levy DS, Lipshutz DB, Lee SH, Liedtke W, Guilak F. Functional characterization of TRPV4 as an osmotically sensitive ion channel in porcine articular chondrocytes. ACTA ACUST UNITED AC 2009; 60:3028-37. [PMID: 19790068 DOI: 10.1002/art.24799] [Citation(s) in RCA: 232] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Transient receptor potential vanilloid 4 (TRPV4) is a Ca(2+)-permeable channel that can be gated by tonicity (osmolarity) and mechanical stimuli. Chondrocytes, the cells in cartilage, respond to their osmotic and mechanical environments; however, the molecular basis of this signal transduction is not fully understood. This study was undertaken to demonstrate the presence and functionality of TRPV4 in chondrocytes. METHODS TRPV4 protein expression was measured by immunolabeling and Western blotting. In response to TRPV4 agonist/antagonists, osmotic stress, and interleukin-1 (IL-1), changes in Ca(2+) signaling, cell volume, and prostaglandin E(2) (PGE(2)) production were measured in porcine chondrocytes using fluorescence microscopy, light microscopy, or immunoassay, respectively. RESULTS TRPV4 was expressed abundantly at the RNA and protein levels. Exposure to 4alpha-phorbol 12,13-didecanoate (4alphaPDD), a TRPV4 activator, caused Ca(2+) signaling in chondrocytes, which was blocked by the selective TRPV4 antagonist, GSK205. Blocking TRPV4 diminished the chondrocytes' response to hypo-osmotic stress, reducing the fraction of Ca(2+) responsive cells, the regulatory volume decrease, and PGE(2) production. Ca(2+) signaling was inhibited by removal of extracellular Ca(2+) or depletion of intracellular stores. Specific activation of TRPV4 restored the defective regulatory volume decrease caused by IL-1. Chemical disruption of the primary cilium eliminated Ca(2+) signaling in response to either 4alphaPDD or hypo-osmotic stress. CONCLUSION Our findings indicate that TRPV4 is present in articular chondrocytes, and chondrocyte response to hypo-osmotic stress is mediated by this channel, which involves both an extracellular Ca(2+) and intracellular Ca(2+) release. TRPV4 may also be involved in modulating the production or influence of proinflammatory molecules in response to osmotic stress.
Collapse
Affiliation(s)
- Mimi N Phan
- Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|