201
|
Abstract
TRPC6 is a non-selective cation channel 6 times more permeable to Ca(2+) than to Na(+). Channel homotetramers heterologously expressed have a characteristic doubly rectifying current-voltage relationship and are directly activated by the second messenger diacylglycerol (DAG). TRPC6 proteins are also regulated by specific tyrosine or serine phosphorylation and phosphoinositides. Given its specific expression pattern, TRPC6 is likely to play a number of physiological roles which are confirmed by the analysis of a Trpc6 (-/-) mouse model. In smooth muscle Na(+) influx through TRPC6 channels and activation of voltage-gated Ca(2+) channels by membrane depolarisation is the driving force for contraction. Permeability of pulmonary endothelial cells depends on TRPC6 and induces ischaemia-reperfusion oedema formation in the lungs. TRPC6 was also identified as an essential component of the slit diaphragm architecture of kidney podocytes and plays an important role in the protection of neurons after cerebral ischaemia. Other functions especially in immune and blood cells remain elusive. Recently identified TRPC6 blockers may be helpful for therapeutic approaches in diseases with highly activated TRPC6 channel activity.
Collapse
Affiliation(s)
- Alexander Dietrich
- Walther-Straub-Institute for Pharmacology and Toxicology, School of Medicine, LM-University of Munich, 80336, Munich, Germany,
| | | |
Collapse
|
202
|
Abstract
The family of transient receptor potential cation channels has received in the last 10 years a tremendous interest because members of this family are involved in a plethora of cell functions and have been identified as causal for many hereditary and acquired diseases. We shortly introduce these channels, summarize nomenclature and chromosomal location of the 28 mammalian Trp genes, and list the available Trp-deficient mouse lines.
Collapse
|
203
|
Malczyk M, Veith C, Fuchs B, Hofmann K, Storch U, Schermuly RT, Witzenrath M, Ahlbrecht K, Fecher-Trost C, Flockerzi V, Ghofrani HA, Grimminger F, Seeger W, Gudermann T, Dietrich A, Weissmann N. Classical Transient Receptor Potential Channel 1 in Hypoxia-induced Pulmonary Hypertension. Am J Respir Crit Care Med 2013; 188:1451-9. [DOI: 10.1164/rccm.201307-1252oc] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
204
|
Gonzales AL, Earley S. Regulation of cerebral artery smooth muscle membrane potential by Ca²⁺-activated cation channels. Microcirculation 2013; 20:337-47. [PMID: 23116477 DOI: 10.1111/micc.12023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 10/26/2012] [Indexed: 12/24/2022]
Abstract
Arterial tone is dependent on the depolarizing and hyperpolarizing currents regulating membrane potential and governing the influx of Ca²⁺ needed for smooth muscle contraction. Several ion channels have been proposed to contribute to membrane depolarization, but the underlying molecular mechanisms are not fully understood. In this review, we will discuss the historical and physiological significance of the Ca²⁺-activated cation channel, TRPM4, in regulating membrane potential of cerebral artery smooth muscle cells. As a member of the recently described transient receptor potential super family of ion channels, TRPM4 possesses the biophysical properties and upstream cellular signaling and regulatory pathways that establish it as a major physiological player in smooth muscle membrane depolarization.
Collapse
Affiliation(s)
- Albert L Gonzales
- Vascular Physiology Research Group, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | | |
Collapse
|
205
|
Kistler AD, Singh G, Altintas MM, Yu H, Fernandez IC, Gu C, Wilson C, Srivastava SK, Dietrich A, Walz K, Kerjaschki D, Ruiz P, Dryer S, Sever S, Dinda AK, Faul C, Reiser J. Transient receptor potential channel 6 (TRPC6) protects podocytes during complement-mediated glomerular disease. J Biol Chem 2013; 288:36598-609. [PMID: 24194522 DOI: 10.1074/jbc.m113.488122] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gain-of-function mutations in the calcium channel TRPC6 lead to autosomal dominant focal segmental glomerulosclerosis and podocyte expression of TRPC6 is increased in some acquired human glomerular diseases, particularly in membranous nephropathy. These observations led to the hypothesis that TRPC6 overactivation is deleterious to podocytes through pathological calcium signaling, both in genetic and acquired diseases. Here, we show that the effects of TRPC6 on podocyte function are context-dependent. Overexpression of TRPC6 alone did not directly affect podocyte morphology and cytoskeletal structure. Unexpectedly, however, overexpression of TRPC6 protected podocytes from complement-mediated injury, whereas genetic or pharmacological TRPC6 inactivation increased podocyte susceptibility to complement. Mechanistically, this effect was mediated by Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activation. Podocyte-specific TRPC6 transgenic mice showed stronger CaMKII activation, reduced podocyte foot process effacement and reduced levels of proteinuria during nephrotoxic serum nephritis, whereas TRPC6 null mice exhibited reduced CaMKII activation and higher levels of proteinuria compared with wild type littermates. Human membranous nephropathy biopsy samples showed podocyte staining for active CaMKII, which correlated with the degree of TRPC6 expression. Together, these data suggest a dual and context dependent role of TRPC6 in podocytes where acute activation protects from complement-mediated damage, but chronic overactivation leads to focal segmental glomerulosclerosis.
Collapse
Affiliation(s)
- Andreas D Kistler
- From the Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Abstract
TRP channels constitute a large superfamily of cation channel forming proteins, all related to the gene product of the transient receptor potential (trp) locus in Drosophila. In mammals, 28 different TRP channel genes have been identified, which exhibit a large variety of functional properties and play diverse cellular and physiological roles. In this article, we provide a brief and systematic summary of expression, function, and (patho)physiological role of the mammalian TRP channels.
Collapse
Affiliation(s)
- Maarten Gees
- Laboratory Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Campus Gasthuisberg, Leuven, Belgium
| | | | | | | |
Collapse
|
207
|
Age-related autoregulatory dysfunction and cerebromicrovascular injury in mice with angiotensin II-induced hypertension. J Cereb Blood Flow Metab 2013; 33:1732-42. [PMID: 23942363 PMCID: PMC3824186 DOI: 10.1038/jcbfm.2013.143] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/25/2013] [Accepted: 07/16/2013] [Indexed: 11/09/2022]
Abstract
Hypertension in the elderly substantially contributes to cerebromicrovascular damage and promotes the development of vascular cognitive impairment. Despite the importance of the myogenic mechanism in cerebromicrovascular protection, it is not well understood how aging affects the functional adaptation of cerebral arteries to high blood pressure. Hypertension was induced in young (3 months) and aged (24 months) C57/BL6 mice by chronic infusion of angiotensin II (AngII). In young hypertensive mice, the range of cerebral blood flow autoregulation was extended to higher pressure values, and the pressure-induced tone of middle cerebral artery (MCA) was increased. In aged hypertensive mice, autoregulation was markedly disrupted, and MCAs did not show adaptive increases in myogenic tone. In young mice, the mechanism of adaptation to hypertension involved upregulation of the 20-HETE (20-hydroxy-5,8,11,14-eicosatetraenoic acid)/transient receptor potential cation channel, subfamily C (TRPC6) pathway and this mechanism was impaired in aged hypertensive mice. Downstream consequences of cerebrovascular autoregulatory dysfunction in aged AngII-induced hypertensive mice included exacerbated disruption of the blood-brain barrier and neuroinflammation (microglia activation and upregulation of proinflammatory cytokines and chemokines), which were associated with impaired hippocampal dependent cognitive function. Collectively, aging impairs autoregulatory protection in the brain of mice with AngII-induced hypertension, potentially exacerbating cerebromicrovascular injury and neuroinflammation.
Collapse
|
208
|
Animal models of nephrotic syndrome. Pediatr Nephrol 2013; 28:2079-88. [PMID: 23250714 DOI: 10.1007/s00467-012-2376-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/25/2012] [Accepted: 10/25/2012] [Indexed: 12/28/2022]
Abstract
Animal models of proteinuria and nephrotic syndrome are essential tools for studying the mechanisms of action of abnormalities in individual components of the podocyte and glomerular basement membrane. In recent years a variety of in vivo models have been developed to elucidate the function of specific podocyte proteins and their role in the pathogenesis of proteinuria and glomerulosclerosis. In this overview of the animal models currently available we discuss their contribution to our mechanistic understanding and their potential use in screening for novel targeted therapies of steroid-resistant nephrotic syndrome.
Collapse
|
209
|
Xia Y, Yang XR, Fu Z, Paudel O, Abramowitz J, Birnbaumer L, Sham JSK. Classical transient receptor potential 1 and 6 contribute to hypoxic pulmonary hypertension through differential regulation of pulmonary vascular functions. Hypertension 2013; 63:173-80. [PMID: 24144647 DOI: 10.1161/hypertensionaha.113.01902] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hypoxic pulmonary hypertension is characterized by increased vascular tone, altered vasoreactivity, and vascular remodeling, which are associated with alterations in Ca(2+) homeostasis in pulmonary arterial smooth muscle cells. We have previously shown that classical transient receptor potential 1 and 6 (TRPC1 and TRPC6) are upregulated in pulmonary arteries (PAs) of chronic hypoxic rats, but it is unclear whether these channels are essential for the development of pulmonary hypertension. Here we found that pulmonary hypertension was suppressed in TRPC1 and TRPC6 knockout (Trpc1(-/-) and Trpc6(-/-)) mice compared with wild-type after exposure to 10% O(2) for 1 and 3 weeks. Muscularization of pulmonary microvessels was inhibited, but rarefaction was unaltered in hypoxic Trpc1(-/-) and Trpc6(-/-) mice. Small PAs of normoxic wild-type mice exhibited vasomotor tone, which was significantly enhanced by chronic hypoxia. Similar vasomotor tone was found in normoxic Trpc1(-/-) PAs, but the hypoxia-induced enhancement was blunted. In contrast, there was minimal vascular tone in normoxic Trpc6(-/-) PAs, but the hypoxia-enhanced tone was preserved. Chronic hypoxia caused significant increase in serotonin-induced vasoconstriction; the augmented vasoreactivity was attenuated in Trpc1(-/-) and eliminated in Trpc6(-/-) PAs. Moreover, the effects of 3-week hypoxia on pulmonary arterial pressure, right ventricular hypertrophy, and muscularization of microvessels were further suppressed in TRPC1-TRPC6 double-knockout mice. Our results, therefore, provide clear evidence that TRPC1 and TRPC6 participate differentially in various pathophysiological processes, and that the presence of TRPC1 and TRPC6 is essential for the full development of hypoxic pulmonary hypertension in the mouse model.
Collapse
Affiliation(s)
- Yang Xia
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224.
| | | | | | | | | | | | | |
Collapse
|
210
|
Senadheera S, Bertrand PP, Grayson TH, Leader L, Tare M, Murphy TV, Sandow SL. Enhanced contractility in pregnancy is associated with augmented TRPC3, L-type, and T-type voltage-dependent calcium channel function in rat uterine radial artery. Am J Physiol Regul Integr Comp Physiol 2013; 305:R917-26. [DOI: 10.1152/ajpregu.00225.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In pregnancy, α-adrenoceptor-mediated vasoconstriction is augmented in uterine radial arteries and is accompanied by underlying changes in smooth muscle (SM) Ca2+ activity. This study aims to determine the Ca2+ entry channels associated with altered vasoconstriction in pregnancy, with the hypothesis that augmented vasoconstriction involves transient receptor potential canonical type-3 (TRPC3) and L- and T-type voltage-dependent Ca2+ channels. Immunohistochemistry showed TRPC3, L-type Cav1.2 (as the α1C subunit), T-type Cav3.1 (α1G), and Cav3.2 (α1H) localization to the uterine radial artery SM. Fluorescence intensity of TRPC3, Cav1.2, and Cav3.2 was increased, and Cav3.1 decreased in radial artery SM from pregnant rats. Western blot analysis confirmed increased TRPC3 protein expression in the radial artery from pregnant rats. Pressure myography incorporating pharmacological intervention to examine the role of these channels in uterine radial arteries showed an attenuation of phenylephrine (PE)-induced constriction with Pyr3 {1-[4-[(2,3,3-trichloro-1-oxo-2-propen-1-yl)amino]phenyl]-5-(trifluoromethyl)-1 H-pyrazole-4-carboxylic acid}-mediated TRPC3 inhibition or with nifedipine-mediated L-type channel block alone in vessels from pregnant rats; both effects of which were diminished in radial arteries from nonpregnant rats. Combined TRPC3 and L-type inhibition attenuated PE-induced constriction in radial arteries, and the residual vasoconstriction was reduced and abolished with T-type channel block with NNC 55-0396 in arteries from nonpregnant and pregnant rats, respectively. With SM Ca2+ stores depleted and in the presence of PE, nifedipine, and NNC 55-0396, blockade of TRPC3 reversed PE-induced constriction. These data suggest that TRPC3 channels act synergistically with L- and T-type channels to modulate radial artery vasoconstriction, with the mechanism being augmented in pregnancy.
Collapse
Affiliation(s)
- Sevvandi Senadheera
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Paul P. Bertrand
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - T. Hilton Grayson
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Leo Leader
- Leo Leader, School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Marianne Tare
- Department of Physiology, Monash University, Melbourne, Australia; and
| | - Timothy V. Murphy
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Shaun L. Sandow
- Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydoore, Australia
| |
Collapse
|
211
|
Gerka-Stuyt J, Au A, Peachey NS, Alagramam KN. Transient receptor potential melastatin 1: a hair cell transduction channel candidate. PLoS One 2013; 8:e77213. [PMID: 24146970 PMCID: PMC3795643 DOI: 10.1371/journal.pone.0077213] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/03/2013] [Indexed: 11/29/2022] Open
Abstract
Sound and head movements are perceived through sensory hair cells in the inner ear. Mounting evidence indicates that this process is initiated by the opening of mechanically sensitive calcium-permeable channels, also referred to as the mechanoelectrical transducer (MET) channels, reported to be around the tips of all but the tallest stereocilia. However, the identity of MET channel remains elusive. Literature suggests that the MET channel is a non-selective cation channel with a high Ca2+ permeability and ∼100 picosiemens conductance. These characteristics make members of the transient receptor potential (TRP) superfamily likely candidates for this role. One of these candidates is the transient receptor potential melastatin 1 protein (TRPM1), which is expressed in various cells types within the cochlea of the mouse including the hair cells. Recent studies demonstrate that mutations in the TRPM1 gene underlie the inherited retinal disease complete congenital stationary night blindness in humans and depolarizing bipolar cell dysfunction in the mouse retina, but auditory function was not assessed. Here we investigate the role of Trpm1 in hearing and as a possible hair cell MET channel using mice homozygous for the null allele of Trpm1 (Trpm1−/−) or a missense mutation in the pore domain of TRPM1 (Trpm1tvrm27/tvrm27). Hearing thresholds were evaluated in adult (4–5 months old) mice with auditory-evoked brain stem responses. Our data shows no statistically significant difference in hearing thresholds in Trpm1−/− or Trpm1tvrm27/tvrm27 mutants compared to littermate controls. Further, none of the mutant mice showed any sign of balance disorder, such as head bobbing or circling. These data suggest that TRPM1 is not essential for development of hearing or balance and it is unlikely that TRPM1 is a component of the hair cell MET channel.
Collapse
Affiliation(s)
- John Gerka-Stuyt
- Otolaryngology Head and Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Adrian Au
- Otolaryngology Head and Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Neal S. Peachey
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States of America
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Kumar N. Alagramam
- Otolaryngology Head and Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
212
|
Blodow S, Schneider H, Storch U, Wizemann R, Forst AL, Gudermann T, Mederos y Schnitzler M. Novel role of mechanosensitive AT1B receptors in myogenic vasoconstriction. Pflugers Arch 2013; 466:1343-53. [PMID: 24101294 DOI: 10.1007/s00424-013-1372-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 01/15/2023]
Abstract
Myogenic vasoconstriction is an inherent property of vascular smooth muscle cells (VSMCs) of resistance arteries harboring ill-defined mechanosensing and mechanotransducing elements. G protein-coupled receptors (GPCRs) are discussed as mechanosensors in VSMCs. In this study, we sought to identify and characterize the role and impact of GPCRs on myogenic vasoconstriction. Thus, we analyzed mRNA expression levels of GPCRs in resistance versus preceding conduit arteries revealing a significant enrichment of several GPCRs in resistance vessels. Selective pharmacological blockade of the highly expressed GPCRs in isolated murine mesenteric arteries ex vivo decreased myogenic vasoconstriction. In particular, candesartan and losartan most prominently suppressed myogenic tone, suggesting that AT1 receptors play a central role in myogenic vasoconstriction. Analyzing angiotensinogen(-/-) mice, a relevant contribution of locally produced angiotensin II to myogenic tone could be excluded. Investigation of AT1A (-/-) and AT1B (-/-) murine mesenteric arteries revealed that AT1B, but not AT1A, receptors are key components of myogenic regulation. This notion was supported by examining fura-2-loaded isolated AT1A (-/-) and AT1B (-/-) VSMCs. Our results indicate that in VSMCs, AT1B receptors are more mechanosensitive than AT1A receptors even at comparable receptor expression levels. Furthermore, we demonstrate that the mechanosensitivity of GPCRs is agonist-independent and positively correlates with receptor expression levels.
Collapse
Affiliation(s)
- Stephanie Blodow
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig Maximilians University of Munich, Goethestr. 33, 80336, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
213
|
Chen W, Oberwinkler H, Werner F, Gaßner B, Nakagawa H, Feil R, Hofmann F, Schlossmann J, Dietrich A, Gudermann T, Nishida M, Del Galdo S, Wieland T, Kuhn M. Atrial Natriuretic Peptide–Mediated Inhibition of Microcirculatory Endothelial Ca
2+
and Permeability Response to Histamine Involves cGMP-Dependent Protein Kinase I and TRPC6 Channels. Arterioscler Thromb Vasc Biol 2013; 33:2121-9. [DOI: 10.1161/atvbaha.113.001974] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Wen Chen
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Heike Oberwinkler
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Franziska Werner
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Birgit Gaßner
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Hitoshi Nakagawa
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Robert Feil
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Franz Hofmann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Jens Schlossmann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Alexander Dietrich
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Thomas Gudermann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Motohiro Nishida
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Sabrina Del Galdo
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Thomas Wieland
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Michaela Kuhn
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| |
Collapse
|
214
|
Büch TRH, Schäfer EAM, Demmel MT, Boekhoff I, Thiermann H, Gudermann T, Steinritz D, Schmidt A. Functional expression of the transient receptor potential channel TRPA1, a sensor for toxic lung inhalants, in pulmonary epithelial cells. Chem Biol Interact 2013; 206:462-71. [PMID: 23994502 DOI: 10.1016/j.cbi.2013.08.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/19/2013] [Accepted: 08/22/2013] [Indexed: 12/11/2022]
Abstract
The cation channel TRPA1 functions as a chemosensory protein and is directly activated by a number of noxious inhalants. A pulmonary expression of TRPA1 has been described in sensory nerve endings and its stimulation leads to the acceleration of inflammatory responses in the lung. Whereas the function of TRPA1 in neuronal cells is well defined, only few reports exist suggesting a role in epithelial cells. The aim of the present study was therefore (1) to evaluate the expression of TRPA1 in pulmonary epithelial cell lines, (2) to characterize TRPA1-promoted signaling in these cells, and (3) to study the extra-neuronal expression of this channel in lung tissue sections. Our results revealed that the widely used alveolar type II cell line A549 expresses TRPA1 at the mRNA and protein level. Furthermore, stimulating A549 cells with known TRPA1 activators (i.e., allyl isothiocyanate) led to an increase in intracellular calcium levels, which was sensitive to the TRPA1 blocker ruthenium red. Investigating TRPA1 coupled downstream signaling cascades it was found that TRPA1 activation elicited a stimulation of ERK1/2 whereas other MAP kinases were not affected. Finally, using epithelial as well as neuronal markers in immunohistochemical approaches, a non-neuronal TRPA1 protein expression was detected in distal parts of the porcine lung epithelium, which was also found examining human lung sections. TRPA1-positive staining co-localized with both epithelial and neuronal markers underlining the observed epithelial expression pattern. Our findings of a functional expression of TRPA1 in pulmonary epithelial cells provide causal evidence for a non-neuronal TRPA1-mediated control of inflammatory responses elicited upon TRPA1-mediated registration of toxic inhalants in vivo.
Collapse
Affiliation(s)
- Thomas Robert Heinrich Büch
- Rudolf-Boehm Institute of Pharmacology and Toxicology, Clinical Pharmacology, University Leipzig, Härtelstraße 16-18, 04107 Leipzig, Germany
| | | | | | | | | | | | | | | |
Collapse
|
215
|
Quick K, Zhao J, Eijkelkamp N, Linley JE, Rugiero F, Cox JJ, Raouf R, Gringhuis M, Sexton JE, Abramowitz J, Taylor R, Forge A, Ashmore J, Kirkwood N, Kros CJ, Richardson GP, Freichel M, Flockerzi V, Birnbaumer L, Wood JN. TRPC3 and TRPC6 are essential for normal mechanotransduction in subsets of sensory neurons and cochlear hair cells. Open Biol 2013; 2:120068. [PMID: 22724068 PMCID: PMC3376737 DOI: 10.1098/rsob.120068] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 04/16/2012] [Indexed: 12/21/2022] Open
Abstract
Transient receptor potential (TRP) channels TRPC3 and TRPC6 are expressed in both sensory neurons and cochlear hair cells. Deletion of TRPC3 or TRPC6 in mice caused no behavioural phenotype, although loss of TRPC3 caused a shift of rapidly adapting (RA) mechanosensitive currents to intermediate-adapting currents in dorsal root ganglion sensory neurons. Deletion of both TRPC3 and TRPC6 caused deficits in light touch and silenced half of small-diameter sensory neurons expressing mechanically activated RA currents. Double TRPC3/TRPC6 knock-out mice also showed hearing impairment, vestibular deficits and defective auditory brain stem responses to high-frequency sounds. Basal, but not apical, cochlear outer hair cells lost more than 75 per cent of their responses to mechanical stimulation. FM1-43-sensitive mechanically gated currents were induced when TRPC3 and TRPC6 were co-expressed in sensory neuron cell lines. TRPC3 and TRPC6 are thus required for the normal function of cells involved in touch and hearing, and are potential components of mechanotransducing complexes.
Collapse
Affiliation(s)
- Kathryn Quick
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Loga F, Domes K, Freichel M, Flockerzi V, Dietrich A, Birnbaumer L, Hofmann F, Wegener JW. The role of cGMP/cGKI signalling and Trpc channels in regulation of vascular tone. Cardiovasc Res 2013; 100:280-7. [PMID: 23832809 DOI: 10.1093/cvr/cvt176] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Signalling via cGMP-dependent protein kinase I (cGKI) is the major pathway in vascular smooth muscle (SM), by which endothelial NO regulates vascular tone. Recent evidence suggests that canonical transient receptor potential (Trpc) channels are targets of cGKI in SM and mediate the relaxant effects of cGMP signalling. We tested this concept by investigating the role of cGMP/cGKI signalling on vascular tone and peripheral resistance using Trpc6(-/-), Trpc3(-/-), Trpc3(-/-)/6(-/-), Trpc1(-/-)/3(-/-)/6(-/-), and SM-specific cGKI(-/-) (sm-cGKI(-/-)) mice. METHODS AND RESULTS α-Adrenergic stimulation induced similar contractions in L-NG-nitroarginine methyl ester (l-NAME)-treated aorta and comparably increased peripheral pressure in hind limbs from all mouse lines investigated. After α-adrenergic stimulation, 8-Br-cGMP diminished similarly aortic tone and peripheral pressure in control, Trpc6(-/-), Trpc3(-/-), Trpc3(-/-)/6(-/-), and Trpc1(-/-)/3(-/-)/6(-/-) mice but not in sm-cGKI(-/-) mice. In untreated aorta, α-adrenergic stimulation induced similar contractions in the aorta from control and Trpc3(-/-) mice but larger contractions in sm-cGKI(-/-), Trpc6(-/-), Trpc3(-/-)/6(-/-), and Trpc1(-/-)/3(-/-)/6(-/-) mice, indicating a functional link between cGKI and Trpc6 channels. Trpc3 channels were detected by immunocytochemistry in both isolated aortic smooth muscle cells (SMCs) and aortic endothelial cells (ECs), whereas Trpc6 channels were detected only in ECs. Phenylephrine-stimulated Ca(2+) levels were similar in SMCs from control (Ctr) and Trpc6(-/-) mice. Carbachol-stimulated Ca(2+) levels were reduced in ECs from Trpc6(-/-) mice. Stimulated Ca(2+) levels were lowered by 8-Br-cGMP in Ctr but not in Trpc6(-/-) ECs. CONCLUSIONS The results suggest that cGKI and Trpc1,3,6 channels are not functionally coupled in vascular SM. Deletion of Trpc6 channels impaired endothelial cGKI signalling and vasodilator tone in the aorta.
Collapse
Affiliation(s)
- Florian Loga
- FOR 923, Institut für Pharmakologie und Toxikologie, Technische Universität München, Biedersteiner Str. 29., 80802 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
217
|
Washburn DG, Holt DA, Dodson J, McAtee JJ, Terrell LR, Barton L, Manns S, Waszkiewicz A, Pritchard C, Gillie DJ, Morrow DM, Davenport EA, Lozinskaya IM, Guss J, Basilla JB, Negron LK, Klein M, Willette RN, Fries RE, Jensen TC, Xu X, Schnackenberg CG, Marino JP. The discovery of potent blockers of the canonical transient receptor channels, TRPC3 and TRPC6, based on an anilino-thiazole pharmacophore. Bioorg Med Chem Lett 2013; 23:4979-84. [PMID: 23886683 DOI: 10.1016/j.bmcl.2013.06.047] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/12/2013] [Accepted: 06/17/2013] [Indexed: 11/30/2022]
Abstract
Lead optimization of piperidine amide HTS hits, based on an anilino-thiazole core, led to the identification of analogs which displayed low nanomolar blocking activity at the canonical transient receptor channels 3 and 6 (TRPC3 & 6) based on FLIPR (carbachol stimulated) and electrophysiology (OAG stimulated) assays. In addition, the anilino-thiazole amides displayed good selectivity over other TRP channels (TRPA1, TRPV1, and TRPV4), as well as against cardiac ion channels (CaV1.2, hERG, and NaV1.5). The high oxidation potential of the aliphatic piperidine and aniline groups, as well as the lability of the thiazole amide group contributed to the high clearance observed for this class of compounds. Conversion of an isoquinoline amide to a naphthyridine amide markedly reduced clearance for the bicyclic piperidines, and improved oral bioavailability for this compound series, however TRPC3 and TRPC6 blocking activity was reduced substantially. Although the most potent anilino-thiazole amides ultimately lacked oral exposure in rodents and were not suitable for chronic dosing, analogs such as 14-19, 22, and 23 are potentially valuable in vitro tool compounds for investigating the role of TRPC3 and TRPC6 in cardiovascular disease.
Collapse
Affiliation(s)
- David G Washburn
- Department of Chemistry, Heart Failure Disease Performance Unit, Metabolic Pathways and Cardiovascular Therapeutic Area Unit, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Harper MT, Londono JEC, Quick K, Londono JC, Flockerzi V, Philipp SE, Birnbaumer L, Freichel M, Poole AW. Transient Receptor Potential Channels Function as a Coincidence Signal Detector Mediating Phosphatidylserine Exposure. Sci Signal 2013; 6:ra50. [DOI: 10.1126/scisignal.2003701] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
219
|
Cardioprotection by Klotho through downregulation of TRPC6 channels in the mouse heart. Nat Commun 2013; 3:1238. [PMID: 23212367 PMCID: PMC3526952 DOI: 10.1038/ncomms2240] [Citation(s) in RCA: 250] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 10/31/2012] [Indexed: 12/20/2022] Open
Abstract
Klotho is a membrane protein predominantly produced in the kidney that exerts some anti-ageing effects. Ageing is associated with an increased risk of heart failure; whether Klotho is cardioprotective is unknown. Here we show that Klotho-deficient mice have no baseline cardiac abnormalities but develop exaggerated pathological cardiac hypertrophy and remodeling in response to stress. Cardioprotection by Klotho in normal mice is mediated by downregulation of TRPC6 channels in the heart. We demonstrate that deletion of Trpc6 prevents stress-induced exaggerated cardiac remodeling in Klotho-deficient mice. Furthermore, mice with heart-specific overexpression of TRPC6 develop spontaneous cardiac hypertrophy and remodeling. Klotho overexpression ameliorates cardiac pathologies in these mice and improves their long-term survival. Soluble Klotho present in the systemic circulation inhibits TRPC6 currents in cardiomyocytes by blocking phosphoinositide-3-kinase-dependent exocytosis of TRPC6 channels. These results provide a new perspective on the pathogenesis of cardiomyopathies and open new avenues for treatment of the disease.
Collapse
|
220
|
Chiluiza D, Krishna S, Schumacher VA, Schlöndorff J. Gain-of-function mutations in transient receptor potential C6 (TRPC6) activate extracellular signal-regulated kinases 1/2 (ERK1/2). J Biol Chem 2013; 288:18407-20. [PMID: 23645677 DOI: 10.1074/jbc.m113.463059] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Gain-of-function mutations in the canonical transient receptor potential 6 (TRPC6) gene are a cause of autosomal dominant focal segmental glomerulosclerosis (FSGS). The mechanisms whereby abnormal TRPC6 activity results in proteinuria remain unknown. The ERK1/2 MAPKs are activated in glomeruli and podocytes in several proteinuric disease models. We therefore examined whether FSGS-associated mutations in TRPC6 result in activation of these kinases. In 293T cells and cultured podocytes, overexpression of gain-of-function TRPC6 mutants resulted in increased ERK1/2 phosphorylation, an effect dependent upon channel function. Pharmacologic inhibitor studies implicated several signaling mediators, including calmodulin and calcineurin, supporting the importance of TRPC6-mediated calcium influx in this process. Through medium transfer experiments, we uncovered two distinct mechanisms for ERK activation by mutant TRPC6, a cell-autonomous, EGF receptor-independent mechanism and a non-cell-autonomous mechanism involving metalloprotease-mediated release of a presumed EGF receptor ligand. The inhibitors KN-92 and H89 were able to block both pathways in mutant TRPC6 expressing cells as well as the prolonged elevation of intracellular calcium levels upon carbachol stimulation seen in these cells. However, these effects appear to be independent of their effects on calcium/calmodulin-dependent protein kinase II and PKA, respectively. Phosphorylation of Thr-70, Ser-282, and Tyr-31/285 were not necessary for ERK activation by mutant TRPC6, although a phosphomimetic TRPC6 S282E mutant was capable of ERK activation. Taken together, these results identify two pathways downstream of mutant TRPC6 leading to ERK activation that may play a role in the development of FSGS.
Collapse
Affiliation(s)
- David Chiluiza
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
221
|
Lindemann O, Umlauf D, Frank S, Schimmelpfennig S, Bertrand J, Pap T, Hanley PJ, Fabian A, Dietrich A, Schwab A. TRPC6 regulates CXCR2-mediated chemotaxis of murine neutrophils. THE JOURNAL OF IMMUNOLOGY 2013; 190:5496-505. [PMID: 23636057 DOI: 10.4049/jimmunol.1201502] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Unraveling the mechanisms involved in chemotactic navigation of immune cells is of particular interest for the development of new immunoregulatory therapies. It is generally agreed upon that members of the classical transient receptor potential channel family (TRPC) are involved in chemotaxis. However, the regulatory role of TRPC channels in chemoattractant receptor-mediated signaling has not yet been clarified in detail. In this study, we demonstrate that the TRPC6 channels play a pronounced role in CXCR2-mediated intermediary chemotaxis, whereas N-formyl-methionine-leucine-phenylalanine receptor-mediated end-target chemotaxis is TRPC6 independent. The knockout of TRPC6 channels in murine neutrophils led to a strongly impaired intermediary chemotaxis after CXCR2 activation which is not further reinforced by CXCR2, PI3K, or p38 MAPK inhibition. Furthermore, CXCR2-mediated Ca(2+) influx but not Ca(2+) store release was attenuated in TRPC6(-/-) neutrophils. We demonstrate that the TRPC6 deficiency affected phosphorylation of AKT and MAPK downstream of CXCR2 receptor activation and led to altered remodeling of actin. The relevance of this TRPC6-depending defect in neutrophil chemotaxis is underscored by our in vivo findings. A nonseptic peritoneal inflammation revealed an attenuated recruitment of neutrophils in the peritoneal cavity of TRPC6(-/-) mice. In summary, this paper defines a specific role of TRPC6 channels in CXCR2-induced intermediary chemotaxis. In particular, TRPC6-mediated supply of calcium appears to be critical for activation of downstream signaling components.
Collapse
Affiliation(s)
- Otto Lindemann
- Institute of Physiology II, Westfälische Wilhelms-University, 48149 Münster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Stiber JA, Tang Y, Li T, Rosenberg PB. Cytoskeletal regulation of TRPC channels in the cardiorenal system. Curr Hypertens Rep 2013; 14:492-7. [PMID: 23054893 DOI: 10.1007/s11906-012-0313-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transient receptor potential canonical (TRPC) channels have been implicated in several aspects of cardiorenal physiology including regulation of blood pressure, vasoreactivity, vascular remodeling, and glomerular filtration. Gain and loss of function studies also support the role of TRPC channels in adverse remodeling associated with cardiac hypertrophy and heart failure. This review discusses TRP channels in the cardiovascular and glomerular filtration systems and their role in disease pathogenesis. We describe the regulation of gating of TRPC channels in the cardiorenal system as well as the influence on activation of these channels by the underlying cytoskeleton and scaffolding proteins. We then focus on the role of TRP channels in the pathogenesis of adverse cardiac remodeling and as potential therapeutic targets in the treatment of heart failure.
Collapse
Affiliation(s)
- Jonathan A Stiber
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
223
|
Yang H, Zhao B, Liao C, Zhang R, Meng K, Xu J, Jiao J. High glucose-induced apoptosis in cultured podocytes involves TRPC6-dependent calcium entry via the RhoA/ROCK pathway. Biochem Biophys Res Commun 2013; 434:394-400. [PMID: 23570668 DOI: 10.1016/j.bbrc.2013.03.087] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 03/29/2013] [Indexed: 10/27/2022]
Abstract
Increasing evidence indicates that podocyte apoptosis is a key event in the development of diabetic nephrology. However, the underlying mechanism of this apoptosis remains poorly understood. In this study, we report that high levels of glucose enhanced the expression of TRPC6 and TRPC6-dependent Ca(2+) influx, but glucose levels did not affect TRPC1 and TRPC5 expression. TRPC6 knockdown by siRNA interference attenuated the observed increase in glucose-induced podocyte apoptosis. High glucose levels also increased the generation of ROS; inhibition of ROS activity by N-acetyl-l-cysteine attenuated the high glucose-induced increase in TRPC6 expression and Ca(2+) influx. Exogenous treatment with H2O2 mimicked the high glucose response, resulting in an increase in TRPC6 expression and Ca(2+) influx. Taken together, these data suggest that high glucose levels induce ROS, thereby mediating TRPC6 expression and Ca(2+) influx. Because RhoA activity is increased following TRPC6 activation, we investigated whether TRPC6 is involved in high glucose-induced apoptosis via the RhoA/ROCK pathway. We report that high glucose levels produced an increase in RhoA activity, and this effect was abolished by the knockdown of TRPC6. Moreover, inhibition of the RhoA/ROCK pathway by a ROCK inhibitor, Y27632, also attenuated high glucose-induced apoptosis. We conclude that TRPC6 is involved in high glucose-induced podocyte apoptosis through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- He Yang
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, PR China
| | | | | | | | | | | | | |
Collapse
|
224
|
Abstract
Ca(2+)-mediated remodeling of the actin cytoskeleton is a dynamic process that regulates cell motility through the modulation of rho guanosine triphosphatase (GTPase) signaling. Kidney podocytes are unique, pericyte-like cells with a complex cellular organization consisting of a cell body, major processes, and foot processes (FPs). The FPs form a characteristic interdigitating pattern with FPs of neighboring podocytes, leaving in between filtration slits that are covered by the slit diaphragm (SD). The actin-based FP and the SD form the final barrier to proteinuria. Mutations affecting several podocyte proteins cause disruption of the filtration barrier and rearrangement of the highly dynamic podocyte actin cytoskeleton. Proteins regulating the plasticity of the podocyte actin cytoskeleton are therefore of critical importance for sustained kidney barrier function. Dynamic regulation of the actin-based contractile apparatus in podocyte FPs is essential for sustained kidney filter function. Thus, the podocyte represents an excellent model system to study calcium signaling and actin dynamics in a physiologic context. Here, we discuss the regulation of podocyte actin dynamics by angiotensin or bradykinin-mediated calcium influx and downstream Rho GTPase signaling pathways and how these pathways are operative in other cells including fibroblasts and cancer cells.
Collapse
Affiliation(s)
- Anna Greka
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA.
| | | |
Collapse
|
225
|
Liu BC, Song X, Lu XY, Li DT, Eaton DC, Shen BZ, Li XQ, Ma HP. High glucose induces podocyte apoptosis by stimulating TRPC6 via elevation of reactive oxygen species. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1434-42. [PMID: 23499875 DOI: 10.1016/j.bbamcr.2013.02.031] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/25/2013] [Accepted: 02/26/2013] [Indexed: 12/20/2022]
Abstract
Podocyte number is significantly reduced in diabetic patients and animal models, but the mechanism remains unclear. In the present study, we found that high glucose induced apoptosis in control podocytes which express transient receptor potential canonical 6 (TRPC6) channels, but not in TRPC6 knockdown podocytes in which TRPC6 was knocked down by TRPC6 silencing short hairpin RNA (shRNA). This effect was reproduced by treatment of podocytes with the reactive oxygen species (ROS), hydrogen peroxide (H2O2). Single-channel data from cell-attached, patch-clamp experiments showed that both high glucose and H2O2 activated the TRPC6 channel in control podocytes, but not in TRPC6 knockdown podocytes. Confocal microscopy showed that high glucose elevated ROS in podocytes and that H2O2 reduced the membrane potential of podocytes and elevated intracellular Ca(2+) via activation of TRPC6. Since intracellular Ca(2+) overload induces apoptosis, H2O2-induced apoptosis may result from TRPC6-mediated elevation of intracellular Ca(2+). These data together suggest that high glucose induces apoptosis in podocytes by stimulating TRPC6 via elevation of ROS.
Collapse
Affiliation(s)
- Bing-Chen Liu
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
226
|
Medic N, Desai A, Olivera A, Abramowitz J, Birnbaumer L, Beaven MA, Gilfillan AM, Metcalfe DD. Knockout of the Trpc1 gene reveals that TRPC1 can promote recovery from anaphylaxis by negatively regulating mast cell TNF-α production. Cell Calcium 2013; 53:315-26. [PMID: 23489970 DOI: 10.1016/j.ceca.2013.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 01/10/2013] [Accepted: 02/07/2013] [Indexed: 11/28/2022]
Abstract
Antigen-mediated mast cell (MC) degranulation is the critical early event in the induction of allergic reactions. Transient receptor potential channels (TRPC), particularly TRPC1, are thought to contribute to such MC activation. To explore the contribution of TRPC1 in MC-driven allergic reactions, we examined antigen-mediated anaphylaxis in Trpc1⁻/⁻ and WT mice, and TRPC1 involvement in the activation of MCs derived from the bone marrow (BMMCs) of these mice. In vivo, we observed a similar induction of passive systemic anaphylaxis in the Trpc1⁻/⁻ mice compared to WT controls. Nevertheless, there was delayed recovery from this response in Trpc1⁻/⁻ mice. Furthermore, contrary to expectations, Trpc1⁻/⁻ BMMCs responded to antigen with enhanced calcium signaling but with little defect in degranulation or associated signaling. In contrast, antigen-mediated production of TNF-α, and other cytokines, was enhanced in the Trpc1⁻/⁻ BMMCs, as were calcium-dependent events required for these responses. Additionally, circulating levels of TNF-α in response to antigen were preferentially elevated in the Trpc1⁻/⁻ mice, and administration of an anti-TNF-α antibody blocked the delay in recovery from anaphylaxis in these mice. These data thus provide evidence that, in this model, TRPC1 promotes recovery from the anaphylactic response by repressing antigen-mediated TNF-α release from MCs.
Collapse
Affiliation(s)
- Nevenka Medic
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive MSC 1881, Bethesda, MD 20892-1881, USA
| | | | | | | | | | | | | | | |
Collapse
|
227
|
Zhang Y, Lu W, Yang K, Xu L, Lai N, Tian L, Jiang Q, Duan X, Chen M, Wang J. Bone morphogenetic protein 2 decreases TRPC expression, store-operated Ca(2+) entry, and basal [Ca(2+)]i in rat distal pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 2013; 304:C833-43. [PMID: 23447035 DOI: 10.1152/ajpcell.00036.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent studies indicate that multiple bone morphogenetic protein (BMP) family ligands and receptors are involved in the development of pulmonary arterial hypertension, yet the underlying mechanisms are incompletely understood. Although BMP2 and BMP4 share high homology in amino acid sequence, they appear to exert divergent effects on chronic hypoxic pulmonary hypertension (CHPH). While BMP4 promotes vascular remodeling, BMP2 prevents CHPH. We previously demonstrated that BMP4 upregulates the expression of canonical transient receptor potential channel (TRPC) proteins and, thereby, enhances store-operated Ca(2+) entry (SOCE) and elevates intracellular Ca(2+) concentration ([Ca(2+)]i) in pulmonary arterial smooth muscle cells (PASMCs). In this study, we investigated the effects of BMP2 on these variables in rat distal PASMCs. We found that treatment with BMP2 (50 ng/ml, 60 h) inhibited TRPC1, TRPC4, and TRPC6 mRNA and protein expression. Moreover, BMP2 treatment led to reduced SOCE and decreased basal [Ca(2+)]i in PASMCs. These alterations were associated with decreased PASMC proliferation and migration. Conversely, knockdown of BMP2 with specific small interference RNA resulted in increased cellular levels of TRPC1, TRPC4, and TRPC6 mRNA and protein, enhanced SOCE, elevated basal [Ca(2+)]i, and increased proliferation and migration of PASMCs. Together, these results indicate that BMP2 participates in regulating Ca(2+) signaling in PASMCs by inhibiting TRPC1, TRPC4, and TRPC6 expression, thus leading to reduced SOCE and basal [Ca(2+)]i and inhibition of cell proliferation and migration.
Collapse
Affiliation(s)
- Yi Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Abstract
Genetically modified mouse models have unparalleled power to determine the mechanisms behind different processes involved in the molecular and physiologic etiology of various classes of human pulmonary hypertension (PH). Processes known to be involved in PH for which there are extensive mouse models available include the following: (1) Regulation of vascular tone through secreted vasoactive factors; (2) regulation of vascular tone through potassium and calcium channels; (3) regulation of vascular remodeling through alteration in metabolic processes, either through alteration in substrate usage or through circulating factors; (4) spontaneous vascular remodeling either before or after development of elevated pulmonary pressures; and (5) models in which changes in tone and remodeling are primarily driven by inflammation. PH development in mice is of necessity faster and with different physiologic ramifications than found in human disease, and so mice make poor models of natural history of PH. However, transgenic mouse models are a perfect tool for studying the processes involved in pulmonary vascular function and disease, and can effectively be used to test interventions designed against particular molecular pathways and processes involved in disease.
Collapse
Affiliation(s)
- Mita Das
- Department of Internal Medicine, University of Arkansas Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | |
Collapse
|
229
|
Hofstra JM, Lainez S, van Kuijk WHM, Schoots J, Baltissen MPA, Hoefsloot LH, Knoers NVAM, Berden JHM, Bindels RJM, van der Vlag J, Hoenderop JGJ, Wetzels JFM, Nijenhuis T. New TRPC6 gain-of-function mutation in a non-consanguineous Dutch family with late-onset focal segmental glomerulosclerosis. Nephrol Dial Transplant 2013; 28:1830-8. [PMID: 23291369 DOI: 10.1093/ndt/gfs572] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Focal segmental glomerulosclerosis (FSGS) is a leading cause of steroid-resistant nephrotic syndrome. Hereditary FSGS is frequently caused by mutations in important structural podocyte proteins, including the slit diaphragm-associated transient receptor potential channel C6 (TRPC6). METHODS In five patients with biopsy-proven autosomal-dominant FSGS from five different Dutch families, all 13 exons of TRPC6 were sequenced. Upon identification of a novel TRPC6 sequence variant, the resultant amino acid change was introduced in the wild-type TRPC6 protein and functionally tested using patch-clamp analyses and cell-surface biotinylation experiments. RESULTS None of the previously described TRPC6 mutations were found in our cohort. In one family, we identified a novel c.524G>A sequence variant resulting in a p.Arg175Gln (R175Q) substitution in the TRPC6 protein. This sequence variant was absent in 449 control subjects and from public SNP databases. The mutation was located in the third ankyrin repeat domain (ANK3) in the cytoplasmic N-tail of TRPC6, important for protein-protein interaction and regulation of ion channel activity. Patch-clamp analyses of the mutant channel indeed showed an increased TRPC6 channel-mediated current. However, cell-surface expression of the mutant channel was not increased. CONCLUSIONS We identified a novel TRPC6 p.Arg175Gln gain-of-function mutation that shows increased TRPC6-mediated current, which is not due to altered cell-surface expression. This is the first mutation identified in ANK3 of the TRPC6 N-tail and is most likely responsible for the late-onset autosomal dominant FSGS in this family.
Collapse
Affiliation(s)
- Julia M Hofstra
- Department of Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Suresh Babu S, Wojtowicz A, Freichel M, Birnbaumer L, Hecker M, Cattaruzza M. Mechanism of stretch-induced activation of the mechanotransducer zyxin in vascular cells. Sci Signal 2012; 5:ra91. [PMID: 23233529 DOI: 10.1126/scisignal.2003173] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vascular cells respond to supraphysiological amounts of stretch with a characteristic phenotypic change that results in dysfunctional remodeling of the affected arteries. Although the pathophysiological consequences of stretch-induced signaling are well characterized, the mechanism of mechanotransduction is unclear. We focused on the mechanotransducer zyxin, which translocates to the nucleus to drive gene expression in response to stretch. In cultured human endothelial cells and perfused femoral arteries isolated from wild-type and several knockout mouse strains, we characterized a multistep signaling pathway whereby stretch led to a transient receptor potential channel 3-mediated release of the endothelial vasoconstrictor peptide endothelin-1 (ET-1). ET-1, through autocrine activation of its B-type receptor, elicited the release of pro-atrial natriuretic peptide (ANP), which caused the autocrine activation of the ANP receptor guanylyl cyclase A (GC-A). Activation of GC-A, in turn, led to protein kinase G-mediated phosphorylation of zyxin at serine 142, thereby triggering the translocation of zyxin to the nucleus, where it was required for stretch-induced gene expression. Thus, we have identified a stretch-induced signaling pathway in vascular cells that leads to the activation of zyxin, a cytoskeletal protein specifically involved in transducing mechanical stimuli.
Collapse
Affiliation(s)
- Sahana Suresh Babu
- Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
231
|
Nagy GA, Botond G, Borhegyi Z, Plummer NW, Freund TF, Hájos N. DAG-sensitive and Ca(2+) permeable TRPC6 channels are expressed in dentate granule cells and interneurons in the hippocampal formation. Hippocampus 2012. [PMID: 23193081 DOI: 10.1002/hipo.22081] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Members of the transient receptor potential (TRP) cation channel family play important roles in several neuronal functions. To understand the precise role of these channels in information processing, their presence on neuronal elements must be revealed. In this study, we investigated the localization of TRPC6 channels in the adult hippocampal formation. Immunostainings with a specific antibody, which was validated in Trpc6 knockout mice, showed that in the dentate gyrus, TRPC6 channels are strongly expressed in granule cells. Immunogold staining revealing the subcellular localization of TRPC6 channels clarified that these proteins were predominantly present on the membrane surface of the dendritic shafts of dentate granule cells, and also in their axons, often associated with intracellular membrane cisternae. In addition, TRPC6 channels could be observed in the dendrites of some interneurons. Double immunofluorescent staining showed that TRPC6 channels were present in the dendrites of hilar interneurons and hippocampal interneurons with horizontal dendrites in the stratum oriens expressing mGlu1a receptors, whereas parvalbumin immunoreactivity was revealed in TRPC6-expressing dendrites with radial appearance in the stratum radiatum. Electron microscopy showed that the immunogold particles depicting TRPC6 channels were located on the surface membranes of the interneuron dendrites. Our results suggest that TRPC6 channels are in a key position to alter the information entry into the trisynaptic loop of the hippocampal formation from the entorhinal cortex, and to control the function of both feed-forward and feed-back inhibitory circuits in this brain region. © 2012 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gergő A Nagy
- Institute of Experimental Medicine, Hungarian Academy of Sciences, H-1450 Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
232
|
Kochukov MY, Balasubramanian A, Noel RC, Marrelli SP. Role of TRPC1 and TRPC3 channels in contraction and relaxation of mouse thoracic aorta. J Vasc Res 2012; 50:11-20. [PMID: 23095462 DOI: 10.1159/000342461] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 07/04/2012] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND/AIMS Canonical transient receptor potential (TRPC) channels modulate membrane potential and intracellular Ca(2+). We examined the role of TRPC1 and TRPC3 channels in vasocontraction and relaxation in mouse aorta. METHODS Vasocontraction and relaxation of aorta from wild-type (WT), TRPC1 KO and TRPC3 knockout (KO) mice were measured for phenylephrine (Phe) and carbachol (CCh). Intracellular Ca(2+) was measured in primary aorta endothelial cells (EC) and whole cell K(+) current in freshly isolated smooth muscle cells (SMC). RESULTS AND CONCLUSION TRPC1 KO aorta showed increased vasocontraction to Phe compared to WT and TRPC3 KO aorta due to diminished role of BK(Ca) channels. BK(Ca) mRNA (aorta) and whole cell current (SMC) were reduced versus WT. Contraction in WT aorta was increased to TRPC1 KO level by BK(Ca) channel inhibition. Relaxation to CCh was reduced in TRPC1 KO and TRPC3 KO aortas with concomitant reduction in EC Ca(2+) response. Pyr3 (TRPC3 blocker) reduced the Ca(2+) response to CCh in EC from WT, but not TRPC3 KO mice. In summary, TRPC1 attenuates receptor-mediated contraction through activation and/or expression of SMC BK(Ca) channels while TRPC3 does not contribute to receptor-mediated constriction. Both TRPC1 and TRPC3 participate in EC Ca(2+) influx and vasorelaxation of aorta.
Collapse
Affiliation(s)
- M Y Kochukov
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
233
|
Chubanov V, Mederos y Schnitzler M, Meißner M, Schäfer S, Abstiens K, Hofmann T, Gudermann T. Natural and synthetic modulators of SK (K(ca)2) potassium channels inhibit magnesium-dependent activity of the kinase-coupled cation channel TRPM7. Br J Pharmacol 2012; 166:1357-76. [PMID: 22242975 DOI: 10.1111/j.1476-5381.2012.01855.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Transient receptor potential cation channel subfamily M member 7 (TRPM7) is a bifunctional protein comprising a TRP ion channel segment linked to an α-type protein kinase domain. TRPM7 is essential for proliferation and cell growth. Up-regulation of TRPM7 function is involved in anoxic neuronal death, cardiac fibrosis and tumour cell proliferation. The goal of this work was to identify non-toxic inhibitors of the TRPM7 channel and to assess the effect of blocking endogenous TRPM7 currents on the phenotype of living cells. EXPERIMENTAL APPROACH We developed an aequorin bioluminescence-based assay of TRPM7 channel activity and performed a hypothesis-driven screen for inhibitors of the channel. The candidates identified were further assessed electrophysiologically and in cell biological experiments. KEY RESULTS TRPM7 currents were inhibited by modulators of small conductance Ca²⁺ -activated K⁺ channels (K(Ca)2.1-2.3; SK) channels, including the antimalarial plant alkaloid quinine, CyPPA, dequalinium, NS8593, SKA31 and UCL 1684. The most potent compound NS8593 (IC₅₀ 1.6 µM) specifically targeted TRPM7 as compared with other TRP channels, interfered with Mg²⁺ -dependent regulation of TRPM7 channel and inhibited the motility of cultured cells. NS8593 exhibited full and reversible block of native TRPM7-like currents in HEK 293 cells, freshly isolated smooth muscle cells, primary podocytes and ventricular myocytes. CONCLUSIONS AND IMPLICATIONS This study reveals a tight overlap in the pharmacological profiles of TRPM7 and K(Ca)2.1-2.3 channels. NS8593 acts as a negative gating modulator of TRPM7 and is well-suited to study functional features and cellular roles of endogenous TRPM7.
Collapse
Affiliation(s)
- V Chubanov
- Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
234
|
Tauseef M, Knezevic N, Chava KR, Smith M, Sukriti S, Gianaris N, Obukhov AG, Vogel SM, Schraufnagel DE, Dietrich A, Birnbaumer L, Malik AB, Mehta D. TLR4 activation of TRPC6-dependent calcium signaling mediates endotoxin-induced lung vascular permeability and inflammation. ACTA ACUST UNITED AC 2012; 209:1953-68. [PMID: 23045603 PMCID: PMC3478927 DOI: 10.1084/jem.20111355] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lung vascular endothelial barrier disruption and the accompanying inflammation are primary pathogenic features of acute lung injury (ALI); however, the basis for the development of both remains unclear. Studies have shown that activation of transient receptor potential canonical (TRPC) channels induces Ca(2+) entry, which is essential for increased endothelial permeability. Here, we addressed the role of Toll-like receptor 4 (TLR4) intersection with TRPC6-dependent Ca(2+) signaling in endothelial cells (ECs) in mediating lung vascular leakage and inflammation. We find that the endotoxin (lipopolysaccharide; LPS) induces Ca(2+) entry in ECs in a TLR4-dependent manner. Moreover, deletion of TRPC6 renders mice resistant to endotoxin-induced barrier dysfunction and inflammation, and protects against sepsis-induced lethality. TRPC6 induces Ca(2+) entry in ECs, which is secondary to the generation of diacylglycerol (DAG) induced by LPS. Ca(2+) entry mediated by TRPC6, in turn, activates the nonmuscle myosin light chain kinase (MYLK), which not only increases lung vascular permeability but also serves as a scaffold to promote the interaction of myeloid differentiation factor 88 and IL-1R-associated kinase 4, which are required for NF-κB activation and lung inflammation. Our findings suggest that TRPC6-dependent Ca(2+) entry into ECs, secondary to TLR4-induced DAG generation, participates in mediating both lung vascular barrier disruption and inflammation induced by endotoxin.
Collapse
Affiliation(s)
- Mohammad Tauseef
- Department of Pharmacology, 2 Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 61605, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Pathogenic role of store-operated and receptor-operated ca(2+) channels in pulmonary arterial hypertension. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:951497. [PMID: 23056939 PMCID: PMC3465915 DOI: 10.1155/2012/951497] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/12/2012] [Accepted: 07/16/2012] [Indexed: 12/31/2022]
Abstract
Pulmonary circulation is an important circulatory system in which the body brings in oxygen. Pulmonary arterial hypertension (PAH) is a progressive and fatal disease that predominantly affects women. Sustained pulmonary vasoconstriction, excessive pulmonary vascular remodeling, in situ thrombosis, and increased pulmonary vascular stiffness are the major causes for the elevated pulmonary vascular resistance (PVR) in patients with PAH. The elevated PVR causes an increase in afterload in the right ventricle, leading to right ventricular hypertrophy, right heart failure, and eventually death. Understanding the pathogenic mechanisms of PAH is important for developing more effective therapeutic approach for the disease. An increase in cytosolic free Ca2+ concentration ([Ca2+]cyt) in pulmonary arterial smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction and an important stimulus for PASMC migration and proliferation which lead to pulmonary vascular wall thickening and remodeling. It is thus pertinent to define the pathogenic role of Ca2+ signaling in pulmonary vasoconstriction and PASMC proliferation to develop new therapies for PAH. [Ca2+]cyt in PASMC is increased by Ca2+ influx through Ca2+ channels in the plasma membrane and by Ca2+ release or mobilization from the intracellular stores, such as sarcoplasmic reticulum (SR) or endoplasmic reticulum (ER). There are two Ca2+ entry pathways, voltage-dependent Ca2+ influx through voltage-dependent Ca2+ channels (VDCC) and voltage-independent Ca2+ influx through store-operated Ca2+ channels (SOC) and receptor-operated Ca2+ channels (ROC). This paper will focus on the potential role of VDCC, SOC, and ROC in the development and progression of sustained pulmonary vasoconstriction and excessive pulmonary vascular remodeling in PAH.
Collapse
|
236
|
Davis J, Burr AR, Davis GF, Birnbaumer L, Molkentin JD. A TRPC6-dependent pathway for myofibroblast transdifferentiation and wound healing in vivo. Dev Cell 2012; 23:705-15. [PMID: 23022034 DOI: 10.1016/j.devcel.2012.08.017] [Citation(s) in RCA: 280] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 06/05/2012] [Accepted: 08/30/2012] [Indexed: 02/06/2023]
Abstract
After injury or cytokine stimulation, fibroblasts transdifferentiate into myofibroblasts, contractile cells that secrete extracellular matrix for wound healing and tissue remodeling. Here, a genome-wide screen identified TRPC6, a Ca(2+) channel necessary and sufficient for myofibroblast transformation. TRPC6 overexpression fully activated myofibroblast transformation, while fibroblasts lacking Trpc6 were refractory to transforming growth factor β (TGF-β) and angiotensin II-induced transdifferentiation. Trpc6 gene-deleted mice showed impaired dermal and cardiac wound healing after injury. The profibrotic ligands TGF-β and angiotensin II induced TRPC6 expression through p38 mitogen-activated protein kinase (MAPK) serum response factor (SRF) signaling via the TRPC6 promoter. Once induced, TRPC6 activates the Ca(2+)-responsive protein phosphatase calcineurin, which itself induced myofibroblast transdifferentiation. Moreover, inhibition of calcineurin prevented TRPC6-dependent transdifferentiation and dermal wound healing. These results demonstrate an obligate function for TRPC6 and calcineurin in promoting myofibroblast differentiation, suggesting a comprehensive pathway for myofibroblast formation in conjunction with TGF-β, p38 MAPK, and SRF.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
237
|
Fransen P, Van Hove CE, van Langen J, Schrijvers DM, Martinet W, De Meyer GRY, Bult H. Contribution of transient and sustained calcium influx, and sensitization to depolarization-induced contractions of the intact mouse aorta. BMC PHYSIOLOGY 2012; 12:9. [PMID: 22943445 PMCID: PMC3499395 DOI: 10.1186/1472-6793-12-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 08/28/2012] [Indexed: 01/31/2023]
Abstract
Background Electrophysiological studies of L-type Ca2+ channels in isolated vascular smooth muscle cells revealed that depolarization of these cells evoked a transient and a time-independent Ca2+ current. The sustained, non-inactivating current occurred at voltages where voltage-dependent activation and inactivation overlapped (voltage window) and its contribution to basal tone or active tension in larger multicellular blood vessel preparations is unknown at present. This study investigated whether window Ca2+ influx affects isometric contraction of multicellular C57Bl6 mouse aortic segments. Results Intracellular Ca2+ (Cai2+, Fura-2), membrane potential and isometric force were measured in aortic segments, which were clamped at fixed membrane potentials by increasing extracellular K+ concentrations. K+ above 20 mM evoked biphasic contractions, which were not affected by inhibition of IP3- or Ca2+ induced Ca2+ release with 2-aminoethoxydiphenyl borate or ryanodine, respectively, ruling out the contribution of intracellular Ca2+ release. The fast force component paralleled Cai2+ increase, but the slow contraction coincided with Cai2+ decrease. In the absence of extracellular Ca2+, basal tension and Cai2+ declined, and depolarization failed to evoke Cai2+ signals or contraction. Subsequent re-introduction of external Ca2+ elicited only slow contractions, which were now matched by Cai2+ increase. After Cai2+ attained steady-state, isometric force kept increasing due to Ca2+- sensitization of the contractile elements. The slow force responses displayed a bell-shaped voltage-dependence, were suppressed by hyperpolarization with levcromakalim, and enhanced by an agonist of L-type Ca2+ channels (BAY K8644). Conclusion The isometric response of mouse aortic segments to depolarization consists of a fast, transient contraction paralleled by a transient Ca2+ influx via Ca2+ channels which completely inactivate. Ca2+ channels, which did not completely inactivate during the depolarization, initiated a second, sustained phase of contraction, which was matched by a sustained non-inactivating window Ca2+ influx. Together with sensitization, this window L-type Ca2+ influx is a major determinant of basal and active tension of mouse aortic smooth muscle.
Collapse
Affiliation(s)
- Paul Fransen
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1 Building T, 2.18, Wilrijk B-2610, Belgium.
| | | | | | | | | | | | | |
Collapse
|
238
|
Smedlund K, Bah M, Vazquez G. On the role of endothelial TRPC3 channels in endothelial dysfunction and cardiovascular disease. Cardiovasc Hematol Agents Med Chem 2012; 10:265-74. [PMID: 22827251 PMCID: PMC3465809 DOI: 10.2174/187152512802651051] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 04/25/2012] [Accepted: 04/27/2012] [Indexed: 05/05/2023]
Abstract
In endothelium, calcium (Ca(2+)) influx through plasma membrane Ca(2+)-permeable channels plays a fundamental role in several physiological functions and in the pathogenesis of cardiovascular disease. Current knowledge on the influence of Ca(2+) influx in signaling events associated to endothelial dysfunction has grown significantly over recent years, particularly after identification of members of the Transient Receptor Potential Canonical (TRPC) family of channel forming proteins as prominent mediators of Ca(2+) entry in endothelial cells. Among TRPC members TRPC3 has been at the center of many of these physiopathological processes. Progress in elucidating the mechanism/s underlying regulation of endothelial TRPC3 and characterization of signaling events downstream TRPC3 activation are of most importance to fully appreciate the role of this peculiar cation channel in cardiovascular disease and its potential use as a therapeutic target. In this updated review we focus on TRPC3 channels, revising and discussing current knowledge on channel expression and regulation in endothelium and the roles of TRPC3 in cardiovascular disease in relation to endothelial dysfunction.
Collapse
Affiliation(s)
| | | | - G. Vazquez
- Correspondence to: Guillermo Vazquez, PhD, Department of Physiology and Pharmacology, UTHSC Mailstop 1800, Toledo OH 43614 USA. FAX: 419 383 2871;
| |
Collapse
|
239
|
Martinsen A, Baeyens N, Yerna X, Morel N. Rho kinase regulation of vasopressin-induced calcium entry in vascular smooth muscle cell: comparison between rat isolated aorta and cultured aortic cells. Cell Calcium 2012; 52:413-21. [PMID: 22883550 DOI: 10.1016/j.ceca.2012.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 07/03/2012] [Accepted: 07/18/2012] [Indexed: 01/05/2023]
Abstract
In addition to its role in artery contraction, Rho kinase (ROCK) is reported to be involved in the Ca(2+) response to vasoconstrictor agonist in rat aorta. However the signaling pathway mediated by ROCK had not been investigated so far and it was not known whether ROCK also contributed to Ca(2+) signaling in cultured vascular smooth muscle cells (VSMC), which undergo profound phenotypic changes. Our results showed that in VSMC, ROCK inhibition by Y-27632 or H-1152 had no effect on the Ca(2+) response to vasopressin, while in aorta the vasopressin-induced Ca(2+) entry was significantly decreased. The inhibition of myosin light chain kinase (MLCK) by ML-7 depressed the vasopressin-induced Ca(2+) signal in aorta but not in VSMC. The difference in ROCK sensitivity of vasopressin-induced Ca(2+) entry between aorta and VSMC was not related to an alteration of the RhoA/ROCK pathway. However, MLCK expression and activity were depressed in cultured cells compared to aorta. We concluded that the regulation of vasopressin-induced Ca(2+) entry by ROCK in aorta could involve the myosin cytoskeleton and could be prevented by the downregulation of MLCK in VSMC. These results underline the important differences in Ca(2+) regulation between whole tissue and cultured cells.
Collapse
|
240
|
Affiliation(s)
- Ivana Y Kuo
- Departments of †Pharmacology and ‡Cellular and Molecular Physiology School of Medicine, Yale University , 333 Cedar Street, New Haven, Connecticut 06520
| | | |
Collapse
|
241
|
Yildirim E, Carey MA, Card JW, Dietrich A, Flake GP, Zhang Y, Bradbury JA, Rebolloso Y, Germolec DR, Morgan DL, Zeldin DC, Birnbaumer L. Severely blunted allergen-induced pulmonary Th2 cell response and lung hyperresponsiveness in type 1 transient receptor potential channel-deficient mice. Am J Physiol Lung Cell Mol Physiol 2012; 303:L539-49. [PMID: 22797250 DOI: 10.1152/ajplung.00389.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Transient receptor potential channels (TRPCs) are widely expressed and regulate Ca²⁺ entry in the cells that participate in the pathophysiology of airway hyperreactivity, inflammation, and remodeling. In vitro studies point to a role for TRPC1-mediated Ca²⁺ signaling in several of these cell types; however, physiological evidence is lacking. Here we identify TRPC1 signaling as proinflammatory and a regulator of lung hyperresponsiveness during allergen-induced pulmonary response. TRPC1-deficient (Trpc1(-/-)) mice are hyposensitive to methacholine challenge and have significantly reduced allergen-induced pulmonary leukocyte infiltration coupled with an attenuated T helper type 2 (Th2) cell response. Upon in vitro allergen exposure, Trpc1(-/-) splenocytes show impaired proliferation and T cell receptor-induced IL-2 production. A high number of germinal centers in spleens of Trpc1(-/-) mice and elevated levels of immunoglobulins in their serum are indicative of dysregulated B cell function and homeostasis. Thus we propose that TRPC1 signaling is necessary in lymphocyte biology and in regulation of allergen-induced lung hyperresponsiveness, making TRPC1 a potential target for treatment of immune diseases and asthma.
Collapse
Affiliation(s)
- Eda Yildirim
- Intramural Research Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Is the antiproteinuric effect of cyclosporine a independent of its immunosuppressive function in T cells? Int J Nephrol 2012; 2012:809456. [PMID: 22778954 PMCID: PMC3384901 DOI: 10.1155/2012/809456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 04/20/2012] [Accepted: 05/03/2012] [Indexed: 12/21/2022] Open
Abstract
The antiproteinuric effect of cyclosporine A(CsA) has been believed to result from its immunosuppressive effect on the transcription factor NFAT in T cells. However, current evidences supporting this hypothesis are missing. A recent study showed that CsA has a direct antiproteinuric effect on podocytes, suggesting a novel non-immunosuppressive mechanism for CsA's antiproteinuric effect. Conditional NFATc1 activation in podoyctes per se is sufficient to induce proteinuria in mice, indicating that NFAT activation in podocytes is a critical pathogenic molecular event leading to podocyte injury and proteinuria. Meanwhile, evidence showed that TRPC6-mediated Ca(2+) influx stimulates NFAT-dependent TRPC6 expression. Altogether, these advances in podocyte research indicate that calcineurin-NFAT signal or calcineurin-synaptopodin axis has a direct proteinuric effect on podocytes which raises the possibility of developing specific antiproteinuric drugs that lack the unwanted effects of calcineurin or NFAT inhibition.
Collapse
|
243
|
Freichel M, Almering J, Tsvilovskyy V. The Role of TRP Proteins in Mast Cells. Front Immunol 2012; 3:150. [PMID: 22701456 PMCID: PMC3372879 DOI: 10.3389/fimmu.2012.00150] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 05/22/2012] [Indexed: 01/16/2023] Open
Abstract
Transient receptor potential (TRP) proteins form cation channels that are regulated through strikingly diverse mechanisms including multiple cell surface receptors, changes in temperature, in pH and osmolarity, in cytosolic free Ca(2+) concentration ([Ca(2+)](i)), and by phosphoinositides which makes them polymodal sensors for fine tuning of many cellular and systemic processes in the body. The 28 TRP proteins identified in mammals are classified into six subfamilies: TRPC, TRPV, TRPM, TRPA, TRPML, and TRPP. When activated, they contribute to cell depolarization and Ca(2+) entry. In mast cells, the increase of [Ca(2+)](i) is fundamental for their biological activity, and several entry pathways for Ca(2+) and other cations were described including Ca(2+) release activated Ca(2+) (CRAC) channels. Like in other non-excitable cells, TRP channels could directly contribute to Ca(2+) influx via the plasma membrane as constituents of Ca(2+) conducting channel complexes or indirectly by shifting the membrane potential and regulation of the driving force for Ca(2+) entry through independent Ca(2+) entry channels. Here, we summarize the current knowledge about the expression of individual Trp genes with the majority of the 28 members being yet identified in different mast cell models, and we highlight mechanisms how they can regulate mast cell functions. Since specific agonists or blockers are still lacking for most members of the TRP family, studies to unravel their function and activation mode still rely on experiments using genetic approaches and transgenic animals. RNAi approaches suggest a functional role for TRPC1, TRPC5, and TRPM7 in mast cell derived cell lines or primary mast cells, and studies using Trp gene knock-out mice reveal a critical role for TRPM4 in mast cell activation and for mast cell mediated cutaneous anaphylaxis, whereas a direct role of cold- and menthol-activated TRPM8 channels seems to be unlikely for the development of cold urticaria at least in mice.
Collapse
Affiliation(s)
- Marc Freichel
- Pharmakologisches Institut, Universität HeidelbergHeidelberg, Germany
| | - Julia Almering
- Pharmakologisches Institut, Universität HeidelbergHeidelberg, Germany
| | | |
Collapse
|
244
|
Wenzel D, Koch M, Matthey M, Heinemann JC, Fleischmann BK. Identification of a Novel Vasoconstrictor Peptide Specific for the Systemic Circulation. Hypertension 2012; 59:1256-62. [DOI: 10.1161/hypertensionaha.111.188367] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Daniela Wenzel
- From the Institute of Physiology I (D.W., M.M., J.C.H., B.K.F.), University of Bonn, Bonn, Germany; Institute for Oral and Musculo-skeletal Biology (M.K.), Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Manuel Koch
- From the Institute of Physiology I (D.W., M.M., J.C.H., B.K.F.), University of Bonn, Bonn, Germany; Institute for Oral and Musculo-skeletal Biology (M.K.), Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Michaela Matthey
- From the Institute of Physiology I (D.W., M.M., J.C.H., B.K.F.), University of Bonn, Bonn, Germany; Institute for Oral and Musculo-skeletal Biology (M.K.), Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Jan C. Heinemann
- From the Institute of Physiology I (D.W., M.M., J.C.H., B.K.F.), University of Bonn, Bonn, Germany; Institute for Oral and Musculo-skeletal Biology (M.K.), Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Bernd K. Fleischmann
- From the Institute of Physiology I (D.W., M.M., J.C.H., B.K.F.), University of Bonn, Bonn, Germany; Institute for Oral and Musculo-skeletal Biology (M.K.), Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
245
|
Kauffenstein G, Laher I, Matrougui K, Guérineau NC, Henrion D. Emerging role of G protein-coupled receptors in microvascular myogenic tone. Cardiovasc Res 2012; 95:223-32. [PMID: 22637750 DOI: 10.1093/cvr/cvs152] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Blood flow autoregulation results from the ability of resistance arteries to reduce or increase their diameters in response to changes in intravascular pressure. The mechanism by which arteries maintain a constant blood flow to organs over a range of pressures relies on this myogenic response, which defines the intrinsic property of the smooth muscle to contract in response to stretch. The resistance to flow created by myogenic tone (MT) prevents tissue damage and allows the maintenance of a constant perfusion, despite fluctuations in arterial pressure. Interventions targeting MT may provide a more rational therapeutic approach in vascular disorders, such as hypertension, vasospasm, chronic heart failure, or diabetes. Despite its early description by Bayliss in 1902, the cellular and molecular mechanisms underlying MT remain poorly understood. We now appreciate that MT requires a complex mechanotransduction converting a physical stimulus (pressure) into a biological response (change in vessel diameter). Although smooth muscle cell depolarization and a rise in intracellular calcium concentration are recognized as cornerstones of the myogenic response, the role of wall strain-induced formation of vasoactive mediators is less well established. The vascular system expresses a large variety of Class 1 G protein-coupled receptors (GPCR) activated by an eclectic range of chemical entities, including peptides, lipids, nucleotides, and amines. These messengers can function in blood vessels as vasoconstrictors. This review focuses on locally generated GPCR agonists and their proposed contributions to MT. Their interplay with pivotal G(q-11) and G(12-13) protein signalling is also discussed.
Collapse
Affiliation(s)
- Gilles Kauffenstein
- Biologie Neurovasculaire et Mitochondriale Intégrée, UMR CNRS 6214 INSERM 1083, Université d'Angers, France
| | | | | | | | | |
Collapse
|
246
|
Miehe S, Crause P, Schmidt T, Löhn M, Kleemann HW, Licher T, Dittrich W, Rütten H, Strübing C. Inhibition of diacylglycerol-sensitive TRPC channels by synthetic and natural steroids. PLoS One 2012; 7:e35393. [PMID: 22530015 PMCID: PMC3328449 DOI: 10.1371/journal.pone.0035393] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 03/16/2012] [Indexed: 12/19/2022] Open
Abstract
TRPC channels are a family of nonselective cation channels that regulate ion homeostasis and intracellular Ca2+ signaling in numerous cell types. Important physiological functions such as vasoregulation, neuronal growth, and pheromone recognition have been assigned to this class of ion channels. Despite their physiological relevance, few selective pharmacological tools are available to study TRPC channel function. We, therefore, screened a selection of pharmacologically active compounds for TRPC modulating activity. We found that the synthetic gestagen norgestimate inhibited diacylglycerol-sensitive TRPC3 and TRPC6 with IC50s of 3–5 µM, while half-maximal inhibition of TRPC5 required significantly higher compound concentrations (>10 µM). Norgestimate blocked TRPC-mediated vasopressin-induced cation currents in A7r5 smooth muscle cells and caused vasorelaxation of isolated rat aorta, indicating that norgestimate could be an interesting tool for the investigation of TRP channel function in native cells and tissues. The steroid hormone progesterone, which is structurally related to norgestimate, also inhibited TRPC channel activity with IC50s ranging from 6 to 18 µM but showed little subtype selectivity. Thus, TRPC channel inhibition by high gestational levels of progesterone may contribute to the physiological decrease of uterine contractility and immunosuppression during pregnancy.
Collapse
Affiliation(s)
- Susanne Miehe
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
| | - Peter Crause
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
| | - Thorsten Schmidt
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
| | - Matthias Löhn
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
| | - Heinz-Werner Kleemann
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
| | - Thomas Licher
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
| | - Werner Dittrich
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
| | - Hartmut Rütten
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
| | - Carsten Strübing
- Sanofi-Aventis Deutschland GmbH, Research and Development, Frankfurt am Main, Germany
- * E-mail:
| |
Collapse
|
247
|
Amelioration of glucolipotoxicity-induced endoplasmic reticulum stress by a "chemical chaperone" in human THP-1 monocytes. EXPERIMENTAL DIABETES RESEARCH 2012; 2012:356487. [PMID: 22550476 PMCID: PMC3328920 DOI: 10.1155/2012/356487] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Accepted: 02/13/2012] [Indexed: 12/29/2022]
Abstract
Chronic ER stress is emerging as a trigger that imbalances a number of systemic and arterial-wall factors and promote atherosclerosis. Macrophage apoptosis within advanced atherosclerotic lesions is also known to increase the risk of atherothrombotic disease. We hypothesize that glucolipotoxicity might mediate monocyte activation and apoptosis through ER stress. Therefore, the aims of this study are (a) to investigate whether glucolipotoxicity could impose ER stress and apoptosis in THP-1 human monocytes and (b) to investigate whether 4-Phenyl butyric acid (PBA), a chemical chaperone could resist the glucolipotoxicity-induced ER stress and apoptosis. Cells subjected to either glucolipotoxicity or tunicamycin exhibited increased ROS generation, gene and protein (PERK, GRP-78, IRE1α, and CHOP) expression of ER stress markers. In addition, these cells showed increased TRPC-6 channel expression and apoptosis as revealed by DNA damage and increased caspase-3 activity. While glucolipotoxicity/tunicamycin increased oxidative stress, ER stress, mRNA expression of TRPC-6, and programmed the THP-1 monocytes towards apoptosis, all these molecular perturbations were resisted by PBA. Since ER stress is one of the underlying causes of monocyte dysfunction in diabetes and atherosclerosis, our study emphasize that chemical chaperones such as PBA could alleviate ER stress and have potential to become novel therapeutics.
Collapse
|
248
|
Kusudo T, Wang Z, Mizuno A, Suzuki M, Yamashita H. TRPV4 deficiency increases skeletal muscle metabolic capacity and resistance against diet-induced obesity. J Appl Physiol (1985) 2012; 112:1223-32. [DOI: 10.1152/japplphysiol.01070.2011] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Transient receptor potential channel V4 (TRPV4) functions as a nonselective cation channel in various cells and plays physiological roles in osmotic and thermal sensation. However, the function of TRPV4 in energy metabolism is unknown. Here, we report that TRPV4 deficiency results in increased muscle oxidative capacity and resistance to diet-induced obesity in mice. Although no difference in body weight was observed between wild-type and Trpv4−/− mice when fed a standard chow diet, obesity phenotypes induced by a high-fat diet were significantly improved in Trpv4−/− mice, without any change in food intake. Quantitative analysis of mRNA revealed the constitutive upregulation of many genes, including those for transcription factors such as peroxisome proliferator-activated receptor α and for metabolic enzymes such as phosphoenolpyruvate carboxykinase. These upregulated genes were especially prominent in oxidative skeletal muscle, in which the activity of Ca2+-dependent phosphatase calcineurin was elevated, suggesting that other Ca2+ channels function in the skeletal muscle of Trpv4−/− mice. Indeed, gene expressions for TRPC3 and TRPC6 increased in the muscles of Trpv4−/− mice compared with those of wild-type mice. The number of oxidative type I fiber also increased in the mutant muscles following myogenin gene induction. These results strongly suggested that inactivation of Trpv4 induces compensatory increases in TRPC3 and TRPC6 production, and elevation of calcineurin activity, affecting energy metabolism through increased expression of genes involved in fuel oxidation in skeletal muscle and thereby contributing to increased energy expenditure and protection from diet-induced obesity in mice.
Collapse
Affiliation(s)
- Tatsuya Kusudo
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai; and
| | - Zhonghua Wang
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai; and
| | - Atsuko Mizuno
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Makoto Suzuki
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Hitoshi Yamashita
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai; and
| |
Collapse
|
249
|
Berwick ZC, Dick GM, Tune JD. Heart of the matter: coronary dysfunction in metabolic syndrome. J Mol Cell Cardiol 2012; 52:848-56. [PMID: 21767548 PMCID: PMC3206994 DOI: 10.1016/j.yjmcc.2011.06.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 06/16/2011] [Accepted: 06/30/2011] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MetS) is a collection of risk factors including obesity, dyslipidemia, insulin resistance/impaired glucose tolerance, and/or hypertension. The incidence of obesity has reached pandemic levels, as ~20-30% of adults in most developed countries can be classified as having MetS. This increased prevalence of MetS is critical as it is associated with a two-fold elevated risk for cardiovascular disease. Although the pathophysiology underlying this increase in disease has not been clearly defined, recent evidence indicates that alterations in the control of coronary blood flow could play an important role. The purpose of this review is to highlight current understanding of the effects of MetS on regulation of coronary blood flow and to outline the potential mechanisms involved. In particular, the role of neurohumoral modulation via sympathetic α-adrenoceptors and the renin-angiotensin-aldosterone system (RAAS) are explored. Alterations in the contribution of end-effector K(+), Ca(2+), and transient receptor potential (TRP) channels are also addressed. Finally, future perspectives and potential therapeutic targeting of the microcirculation in MetS are discussed. This article is part of a Special Issue entitled "Coronary Blood Flow".
Collapse
Affiliation(s)
- Zachary C. Berwick
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Gregory M. Dick
- Department of Exercise Physiology Center for Cardiovascular and Respiratory Sciences West Virginia University School of Medicine
| | - Johnathan D. Tune
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
250
|
Zhao Z, Ni Y, Chen J, Zhong J, Yu H, Xu X, He H, Yan Z, Scholze A, Liu D, Zhu Z, Tepel M. Increased migration of monocytes in essential hypertension is associated with increased transient receptor potential channel canonical type 3 channels. PLoS One 2012; 7:e32628. [PMID: 22438881 PMCID: PMC3306381 DOI: 10.1371/journal.pone.0032628] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 02/01/2012] [Indexed: 02/06/2023] Open
Abstract
Increased transient receptor potential canonical type 3 (TRPC3) channels have been observed in patients with essential hypertension. In the present study we tested the hypothesis that increased monocyte migration is associated with increased TRPC3 expression. Monocyte migration assay was performed in a microchemotaxis chamber using chemoattractants formylated peptide Met-Leu-Phe (fMLP) and tumor necrosis factor-α (TNF-α). Proteins were identified by immunoblotting and quantitative in-cell Western assay. The effects of TRP channel-inhibitor 2–aminoethoxydiphenylborane (2-APB) and small interfering RNA knockdown of TRPC3 were investigated. We observed an increased fMLP-induced migration of monocytes from hypertensive patients compared with normotensive control subjects (246±14% vs 151±10%). The TNF-α-induced migration of monocytes in patients with essential hypertension was also significantly increased compared to normotensive control subjects (221±20% vs 138±18%). In the presence of 2-APB or after siRNA knockdown of TRPC3 the fMLP-induced monocyte migration was significantly blocked. The fMLP-induced changes of cytosolic calcium were significantly increased in monocytes from hypertensive patients compared to normotensive control subjects. The fMLP-induced monocyte migration was significantly reduced in the presence of inhibitors of tyrosine kinase and phosphoinositide 3-kinase. We conclude that increased monocyte migration in patients with essential hypertension is associated with increased TRPC3 channels.
Collapse
Affiliation(s)
- Zhigang Zhao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Yinxing Ni
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Jing Chen
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Jian Zhong
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Hao Yu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Xingsen Xu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Hongbo He
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Alexandra Scholze
- Department of Nephrology, Charité, Berlin, Germany; and University of Southern Denmark, Institute for Molecular Medicine, Odense, Denmark
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
- * E-mail: (DL); (Z. Zhu)
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
- * E-mail: (DL); (Z. Zhu)
| | - Martin Tepel
- Department of Nephrology, Charité, Berlin, Germany; and University of Southern Denmark, Institute for Molecular Medicine, Odense, Denmark
| |
Collapse
|