Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
EMBO J ; 27(23): 3092-103, 2008 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-18987636

RESUMEN

Despite the central physiological function of the myogenic response, the underlying signalling pathways and the identity of mechanosensors in vascular smooth muscle (VSM) are still elusive. In contrast to present thinking, we show that membrane stretch does not primarily gate mechanosensitive transient receptor potential (TRP) ion channels, but leads to agonist-independent activation of G(q/11)-coupled receptors, which subsequently signal to TRPC channels in a G protein- and phospholipase C-dependent manner. Mechanically activated receptors adopt an active conformation, allowing for productive G protein coupling and recruitment of beta-arrestin. Agonist-independent receptor activation by mechanical stimuli is blocked by specific antagonists and inverse agonists. Increasing the AT(1) angiotensin II receptor density in mechanically unresponsive rat aortic A7r5 cells resulted in mechanosensitivity. Myogenic tone of cerebral and renal arteries is profoundly diminished by the inverse angiotensin II AT(1) receptor agonist losartan independently of angiotensin II (AII) secretion. This inhibitory effect is enhanced in blood vessels of mice deficient in the regulator of G-protein signalling-2. These findings suggest that G(q/11)-coupled receptors function as sensors of membrane stretch in VSM cells.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Mecanorreceptores/fisiología , Músculo Liso Vascular/fisiología , Receptores Acoplados a Proteínas G/fisiología , Vasoconstricción , Angiotensina II/metabolismo , Animales , Arrestinas/metabolismo , Línea Celular , Humanos , Ratas , Ratas Sprague-Dawley , Receptores de Angiotensina/fisiología , Canales de Potencial de Receptor Transitorio/metabolismo , Fosfolipasas de Tipo C/metabolismo , beta-Arrestinas
2.
Blood ; 113(6): 1326-31, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19074007

RESUMEN

Oxygen-dependent antimicrobial activity of human polymorphonuclear leukocytes (PMNs) relies on the phagocyte nicotinamide adenine dinucleotide phosphate (NADPH) oxidase to generate oxidants. As the oxidase transfers electrons from NADPH the membrane will depolarize and concomitantly terminate oxidase activity, unless there is charge translocation to compensate. Most experimental data implicate proton channels as the effectors of this charge compensation, although large-conductance Ca2+-activated K+ (BK) channels have been suggested to be essential for normal PMN antimicrobial activity. To test this latter notion, we directly assessed the role of BK channels in phagocyte function, including the NADPH oxidase. PMNs genetically lacking BK channels (BK(-/-)) had normal intracellular and extracellular NADPH oxidase activity in response to both receptor-independent and phagocytic challenges. Furthermore, NADPH oxidase activity of human PMNs and macrophages was normal after treatment with BK channel inhibitors. Although BK channel inhibitors suppressed endotoxin-mediated tumor necrosis factor-alpha secretion by bone marrow-derived macrophages (BMDMs), BMDMs of BK(-/-) and wild-type mice responded identically and exhibited the same ERK, PI3K/Akt, and nuclear factor-kappaB activation. Based on these data, we conclude that the BK channel is not required for NADPH oxidase activity in PMNs or macrophages or for endotoxin-triggered tumor necrosis factor-alpha release and signal transduction BMDMs.


Asunto(s)
Inmunidad Innata , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Macrófagos/inmunología , Neutrófilos/inmunología , Animales , Femenino , Citometría de Flujo , Indoles/farmacología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , NADPH Oxidasas/metabolismo , FN-kappa B/metabolismo , Fagocitos/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estallido Respiratorio , Transducción de Señal , Superóxidos/metabolismo , Arterias Tibiales/citología , Arterias Tibiales/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 29(2): 232-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074484

RESUMEN

OBJECTIVE: Vascular smooth muscle cells (VSMCs) and circulating mesenchymal progenitor cells (MSCs) with a VSMC phenotype contribute to neointima formation and lumen loss after angioplasty and during allograft arteriosclerosis. We hypothesized that phosphoinositol-Akt-mammalian target of rapamycin-p70S6 kinase (PI3K/Akt/mTOR/p70S6K) pathway activation regulates VSMC differentiation from MSCs. METHODS AND RESULTS: We studied effects of PI3K/Akt/mTOR signaling on phenotypic modulation of MSC and VSMC marker expression, including L-type Ca(2+) channels. Phosphorylation of Akt and p70S6K featured downregulation of VSMC markers in dedifferentiated MSCs. mTOR inhibition with rapamycin at below pharmacological concentrations blocked p70S6K phosphorylation and induced a differentiated contractile phenotype with smooth muscle (sm)-calponin, sm-alpha-actin, and SM protein 22-alpha (SM22alpha) expression. The PI3K inhibitor Ly294002 abolished Akt and p70S6K phosphorylation and reversed the dedifferentiated phenotype via induction of sm-calponin, sm-alpha-actin, SM22alpha, and myosin light chain kinase. Rapamycin acted antiproliferative without impairing MSC viability. In VSMCs, rapamycin increased a homing chemokine for MSCs, stromal cell-derived factor-1-alpha, at mRNA and protein levels. The CXCR4-mediated MSC migration toward conditioned medium of rapamycin-treated VSMCs was enhanced. CONCLUSIONS: We describe novel pleiotropic effects of rapamycin at very low concentrations that stabilized differentiated contractile VSMCs from MSCs in addition to exerting antiproliferative and enhanced homing effects.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Canales de Calcio Tipo L/metabolismo , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Quimiotaxis , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/enzimología , Morfolinas/farmacología , Proteínas Musculares/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factores de Tiempo
4.
J Biomed Biotechnol ; 2009: 135249, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20029633

RESUMEN

Calcium sparks represent local, rapid, and transient calcium release events from a cluster of ryanodine receptors (RyRs) in the sarcoplasmic reticulum. In arterial smooth muscle cells (SMCs), calcium sparks activate calcium-dependent potassium channels causing decrease in the global intracellular [Ca2+] and oppose vasoconstriction. This is in contrast to cardiac and skeletal muscle, where spatial and temporal summation of calcium sparks leads to global increases in intracellular [Ca2+] and myocyte contraction. We summarize the present data on local RyR calcium signaling in arterial SMCs in comparison to striated muscle and muscle-specific differences in coupling between L-type calcium channels and RyRs. Accordingly, arterial SMC Ca(v)1.2 L-type channels regulate intracellular calcium stores content, which in turn modulates calcium efflux though RyRs. Downregulation of RyR2 up to a certain degree is compensated by increased SR calcium content to normalize calcium sparks. This indirect coupling between Ca(v)1.2 and RyR in arterial SMCs is opposite to striated muscle, where triggering of calcium sparks is controlled by rapid and direct cross-talk between Ca(v)1.1/Ca(v)1.2 L-type channels and RyRs. We discuss the role of RyR isoforms in initiation and formation of calcium sparks in SMCs and their possible molecular binding partners and regulators, which differ compared to striated muscle.


Asunto(s)
Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Estriado/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Arterias/metabolismo , Humanos
5.
FASEB J ; 21(14): 4101-11, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17666455

RESUMEN

Hyperforin, a bicyclic polyprenylated acylphloroglucinol derivative, is the main active principle of St. John's wort extract responsible for its antidepressive profile. Hyperforin inhibits the neuronal serotonin and norepinephrine uptake comparable to synthetic antidepressants. In contrast to synthetic antidepressants directly blocking neuronal amine uptake, hyperforin increases synaptic serotonin and norepinephrine concentrations by an indirect and yet unknown mechanism. Our attempts to identify the molecular target of hyperforin resulted in the identification of TRPC6. Hyperforin induced sodium and calcium entry as well as currents in TRPC6-expressing cells. Sodium currents and the subsequent breakdown of the membrane sodium gradients may be the rationale for the inhibition of neuronal amine uptake. The hyperforin-induced cation entry was highly specific and related to TRPC6 and was suppressed in cells expressing a dominant negative mutant of TRPC6, whereas phylogenetically related channels, i.e., TRPC3 remained unaffected. Furthermore, hyperforin induces neuronal axonal sprouting like nerve growth factor in a TRPC6-dependent manner. These findings support the role of TRPC channels in neurite extension and identify hyperforin as the first selective pharmacological tool to study TRPC6 function. Hyperforin integrates inhibition of neurotransmitter uptake and neurotrophic property by specific activation of TRPC6 and represents an interesting lead-structure for a new class of antidepressants.


Asunto(s)
Hypericum/química , Hypericum/fisiología , Floroglucinol/análogos & derivados , Canales Catiónicos TRPC/metabolismo , Terpenos/farmacología , Animales , Compuestos Bicíclicos con Puentes/antagonistas & inhibidores , Compuestos Bicíclicos con Puentes/farmacología , Calcio/antagonistas & inhibidores , Calcio/metabolismo , Línea Celular , Depresión/tratamiento farmacológico , Depresión/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células PC12 , Floroglucinol/antagonistas & inhibidores , Floroglucinol/farmacología , Extractos Vegetales/uso terapéutico , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sodio/antagonistas & inhibidores , Sodio/metabolismo , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/biosíntesis , Canales Catiónicos TRPC/genética , Terpenos/antagonistas & inhibidores
6.
Mol Cell Biol ; 25(16): 6980-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16055711

RESUMEN

Among the TRPC subfamily of TRP (classical transient receptor potential) channels, TRPC3, -6, and -7 are gated by signal transduction pathways that activate C-type phospholipases as well as by direct exposure to diacylglycerols. Since TRPC6 is highly expressed in pulmonary and vascular smooth muscle cells, it represents a likely molecular candidate for receptor-operated cation entry. To define the physiological role of TRPC6, we have developed a TRPC6-deficient mouse model. These mice showed an elevated blood pressure and enhanced agonist-induced contractility of isolated aortic rings as well as cerebral arteries. Smooth muscle cells of TRPC6-deficient mice have higher basal cation entry, increased TRPC-carried cation currents, and more depolarized membrane potentials. This higher basal cation entry, however, was completely abolished by the expression of a TRPC3-specific small interference RNA in primary TRPC6(-)(/)(-) smooth muscle cells. Along these lines, the expression of TRPC3 in wild-type cells resulted in increased basal activity, while TRPC6 expression in TRPC6(-/-) smooth muscle cells reduced basal cation influx. These findings imply that constitutively active TRPC3-type channels, which are up-regulated in TRPC6-deficient smooth muscle cells, are not able to functionally replace TRPC6. Thus, TRPC6 has distinct nonredundant roles in the control of vascular smooth muscle tone.


Asunto(s)
Canales de Calcio/genética , Canales de Calcio/fisiología , Contracción Muscular , Músculo Liso Vascular/citología , Animales , Aorta/patología , Arterias/citología , Bario/farmacología , Presión Sanguínea , Western Blotting , Cationes , ADN Complementario/metabolismo , Dependovirus/genética , Electrofisiología , Electroporación , Vectores Genéticos , Canales Iónicos/metabolismo , Ratones , Ratones Transgénicos , Modelos Genéticos , Músculos/citología , Miocitos del Músculo Liso/citología , Técnicas de Placa-Clamp , Fenilefrina/farmacología , Presión , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Canales Catiónicos TRPC , Canal Catiónico TRPC6 , Factores de Tiempo
7.
PLoS One ; 11(4): e0153101, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27054717

RESUMEN

INTRODUCTION: Obliterative vasculopathy and fibrosis are hallmarks of systemic sclerosis (SSc), a severe systemic autoimmune disease. Bone marrow-derived mesenchymal stromal cells (MSCs) from SSc patients may harbor disease-specific abnormalities. We hypothesized disturbed vascular smooth muscle cell (VSMC) differentiation with increased propensity towards myofibroblast differentiation in response to SSc-microenvironment defining growth factors and determined responsible mechanisms. METHODS: We studied responses of multipotent MSCs from SSc-patients (SSc-MSCs) and healthy controls (H-MSCs) to long-term exposure to CTGF, b-FGF, PDGF-BB or TGF-ß1. Differentiation towards VSMC and myofibroblast lineages was analyzed on phenotypic, biochemical, and functional levels. Intracellular signaling studies included analysis of TGF-ß receptor regulation, SMAD, AKT, ERK1/2 and autocrine loops. RESULTS: VSMC differentiation towards both, contractile and synthetic VSMC phenotypes in response to CTGF and b-FGF was disturbed in SSc-MSCs. H-MSCs and SSc-MSCs responded equally to PDGF-BB with prototypic fibroblastic differentiation. TGF-ß1 initiated myofibroblast differentiation in both cell types, yet with striking phenotypic and functional differences: In relation to H-MSC-derived myofibroblasts induced by TGF-ß1, those obtained from SSc-MSCs expressed more contractile proteins, migrated towards TGF-ß1, had low proliferative capacity, and secreted higher amounts of collagen paralleled by reduced MMP expression. Higher levels of TGF-ß receptor 1 and enhanced canonical and noncanonical TGF-ß signaling in SSc-MSCs accompanied aberrant differentiation response of SSc-MSCs in comparison to H-MSCs. CONCLUSIONS: Deregulated VSMC differentiation with a shift towards myofibroblast differentiation expands the concept of disturbed endogenous regenerative capacity of MSCs from SSc patients. Disease related intrinsic hyperresponsiveness to TGF-ß1 with increased collagen production may represent one responsible mechanism. Better understanding of repair barriers and harnessing beneficial differentiation processes in MSCs could widen options of autologous MSC application in SSc patients.


Asunto(s)
Células Madre Mesenquimatosas/citología , Músculo Liso Vascular/citología , Miofibroblastos/citología , Esclerodermia Sistémica/patología , Adulto , Anciano , Becaplermina , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Persona de Mediana Edad , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología
8.
Stroke ; 35(7): 1709-14, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15155963

RESUMEN

BACKGROUND AND PURPOSE: Because of their mixed estrogen-agonist and estrogen-antagonist properties, selective estrogen receptor modulators (SERMs) are considered promising substitutes for hormone replacement therapy. Raloxifene and other SERMs confer estrogen-like cardiovascular protective effects but lack the carcinogenic activity of exogenous estrogen. However, little is known about the cerebrovascular action of raloxifene. Therefore, we studied the effects of raloxifene on the mechanisms regulating rat cerebral artery tone. METHODS: Ring segments of the isolated rat posterior communicating cerebral arteries were mounted in a microvessel myograph for measurement of isometric tension. Whole-cell L-type voltage-sensitive Ca2+ currents were recorded using the perforated patch-clamp technique. Raloxifene (0.1 to 10 micromol/L) reduced the contractile responses to U46619, phenylephrine, and endothelin-1 in normal Krebs solution or to CaCl2 in Ca2+-free, high K+-containing solution. Raloxifene-induced relaxation was identical in endothelium-intact and endothelium-denuded rings. ICI 182780 had no effect on raloxifene-induced relaxation. Raloxifene reduced L-type Ca2+ currents with a pD2 of 5.98+/-0.06, close to that (6.44+/-0.09) for raloxifene-induced relaxation of 60 mmol/L K+-contracted rings. CONCLUSIONS: This study demonstrates that raloxifene acutely relaxes rat cerebral arteries largely via an endothelium-independent mechanism, involving inhibition of Ca2+ influx through L-type Ca2+ channels.


Asunto(s)
Canales de Calcio Tipo L/efectos de los fármacos , Arterias Cerebrales/efectos de los fármacos , Clorhidrato de Raloxifeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Vasoconstricción/efectos de los fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Circulación Cerebrovascular/efectos de los fármacos , Endotelio Vascular , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Vasoconstrictores/farmacología
9.
J Hypertens ; 20(5): 885-93, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12011649

RESUMEN

Cilnidipine is a novel dihydropyridine (DHP) antagonist. However, its pharmacological effects on vascular DHP-sensitive L-type channels and protein kinase C (PKC)-mediated arterial contraction is incompletely understood. To address this issue, we studied the effects of cilnidipine on multi-subunit, C-class L-type Ca2+ channels in rat aortic A7r5 cells, as well as on Ca2+ channel (L-type) alpha1C-b and (T-type) alpha1G subunits in the Xenopus oocyte expression system. Cilnidipine dose- and time-dependently inhibited Ba2+ currents in A7r5 cells, with half-maximal inhibitions (IC50) at 10 nmol/l after 10 min. Unlike classical pharmacological Ca2+ channel blockers, cilnidipine's block of Ca2+ currents did not reach steady-state levels within 10 min, indicating steady-state half-maximal inhibition of native, multi-subunit L-type channels at < 10 nmol/l. In contrast, smooth muscle alpha1Cb currents were blocked by cilnidipine at much higher doses (steady-state IC50, 20 micromol/l) whereas alpha1G currents were not inhibited by cilnidipine (30 micromol/l). Cilnidipine dose-dependently inhibited depolarization- and Ca2+-induced contractions of rat aortic rings, with an IC50 of 10 nmol/l at 10 min. However, the onset of the effects was very slow, with approximately 71% inhibition by 3 nmol/l cilnidipine after 90 min exposure to cilnidipine. In contrast, cilnidipine did not inhibit phorbol 12-myristate-13-acetate (100 nmol/l)-mediated contractions. We conclude that cilnidipine represents an extremely slow-acting DHP that targets multi-subunit L-type channels, but not PKC in arterial smooth muscle. Because cilnidipine is less potent in cells expressing the pore-forming alpha1C-b subunit, the data further suggest that this unique slow-acting mechanism of cilnidipine is mediated by a complex interaction of cilnidipine with alpha1C-b and accessory channel subunits.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Dihidropiridinas/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Calcio/farmacología , Canales de Calcio Tipo L/fisiología , Línea Celular , Conductividad Eléctrica , Electrofisiología , Isoformas de Proteínas/fisiología , Proteína Quinasa C/metabolismo , Ratas , Acetato de Tetradecanoilforbol/farmacología , Vasoconstricción/efectos de los fármacos
10.
J Biol Chem ; 283(49): 33942-54, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18818211

RESUMEN

The protective epithelial barrier in our skin undergoes constant regulation, whereby the balance between differentiation and proliferation of keratinocytes plays a major role. Impaired keratinocyte differentiation and proliferation are key elements in the pathophysiology of several important dermatological diseases, including atopic dermatitis and psoriasis. Ca(2+) influx plays an essential role in this process presumably mediated by different transient receptor potential (TRP) channels. However, investigating their individual role was hampered by the lack of specific stimulators or inhibitors. Because we have recently identified hyperforin as a specific TRPC6 activator, we investigated the contribution of TRPC6 to keratinocyte differentiation and proliferation. Like the endogenous differentiation stimulus high extracellular Ca(2+) concentration ([Ca(2+)](o)), hyperforin triggers differentiation in HaCaT cells and in primary cultures of human keratinocytes by inducing Ca(2+) influx via TRPC6 channels and additional inhibition of proliferation. Knocking down TRPC6 channels prevents the induction of Ca(2+)- and hyperforin-induced differentiation. Importantly, TRPC6 activation is sufficient to induce keratinocyte differentiation similar to the physiological stimulus [Ca(2+)](o). Therefore, TRPC6 activation by hyperforin may represent a new innovative therapeutic strategy in skin disorders characterized by altered keratinocyte differentiation.


Asunto(s)
Queratinocitos/citología , Canales Catiónicos TRPC/fisiología , Compuestos Bicíclicos con Puentes/farmacología , Calcio/química , Cationes , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Queratinocitos/metabolismo , Modelos Biológicos , Técnicas de Cultivo de Órganos/métodos , Floroglucinol/análogos & derivados , Floroglucinol/farmacología , Piel/metabolismo , Enfermedades de la Piel/metabolismo , Canales Catiónicos TRPC/química , Canal Catiónico TRPC6 , Terpenos/farmacología , Factores de Tiempo , Transfección
11.
J Physiol ; 584(Pt 1): 205-19, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17673505

RESUMEN

In arterial vascular smooth muscle cells (VSMCs), Ca(2+) sparks stimulate nearby Ca(2+)-activated K(+) (BK) channels that hyperpolarize the membrane and close L-type Ca(2+) channels. We tested the contribution of L-type Ca(v)1.2 channels to Ca(2+) spark regulation in tibial and cerebral artery VSMCs using VSMC-specific Ca(v)1.2 channel gene disruption in (SMAKO) mice and an approach based on Poisson statistical analysis of activation frequency and first latency of elementary events. Ca(v)1.2 channel gene inactivation reduced Ca(2+) spark frequency and amplitude by approximately 50% and approximately 80%, respectively. These effects were associated with lower global cytosolic Ca(2+) levels and reduced sarcoplasmic reticulum (SR) Ca(2+) load. Elevating cytosolic Ca(2+) levels reversed the effects completely. The activation frequency and first latency of elementary events in both wild-type and SMAKO VSMCs weakly reflected the voltage dependency of L-type channels. This study provides evidence that local and tight coupling between the Ca(v)1.2 channels and ryanodine receptors (RyRs) is not required to initiate Ca(2+) sparks. Instead, Ca(v)1.2 channels contribute to global cytosolic [Ca(2+)], which in turn influences luminal SR calcium and thus Ca(2+) sparks.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Calcio/metabolismo , Citosol/metabolismo , Miocitos del Músculo Liso/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Arteria Basilar/metabolismo , Células Cultivadas , Dihidropiridinas , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Arterias Tibiales/metabolismo
12.
Pflugers Arch ; 455(3): 465-77, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17647013

RESUMEN

Among the classical transient receptor potential (TRPC) subfamily, TRPC1 is described as a mechanosensitive and store-operated channel proposed to be activated by hypoosmotic cell swelling and positive pipette pressure as well as regulated by the filling status of intracellular Ca(2+) stores. However, evidence for a physiological role of TRPC1 may most compellingly be obtained by the analysis of a TRPC1-deficient mouse model. Therefore, we have developed and analyzed TRPC1(-/-) mice. Pressure-induced constriction of cerebral arteries was not impaired in TRPC1(-/-) mice. Smooth muscle cells from cerebral arteries activated by hypoosmotic swelling and positive pipette pressure showed no significant differences in cation currents compared to wild-type cells. Moreover, smooth muscle cells of TRPC1(-/-) mice isolated from thoracic aortas and cerebral arteries showed no change in store-operated cation influx induced by thapsigargin, inositol-1,4,5 trisphosphate, and cyclopiazonic acid compared to cells from wild-type mice. In contrast to these results, small interference RNAs decreasing the expression of stromal interaction molecule 1 (STIM1) inhibited thapsigargin-induced store-operated cation influx, demonstrating that STIM1 and TRPC1 are mutually independent. These findings also imply that, as opposed to current concepts, TRPC1 is not an obligatory component of store-operated and stretch-activated ion channel complexes in vascular smooth muscle cells.


Asunto(s)
Músculo Liso Vascular/fisiología , Canales Catiónicos TRPC/fisiología , Secuencia de Aminoácidos , Animales , Aorta Torácica/citología , Secuencia de Bases , Canales de Calcio , Arterias Cerebrales/citología , Indoles/farmacología , Inositol 1,4,5-Trifosfato/farmacología , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/biosíntesis , Ratones , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/deficiencia , Tapsigargina/farmacología
13.
Exp Physiol ; 92(6): 1067-76, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17675416

RESUMEN

17,18-Epoxyeicosatetraenoic acid (17,18-EETeTr) stimulates vascular large-conductance K(+) (BK) channels. BK channels are composed of the pore-forming BK alpha and auxiliary BK beta1 subunits that confer an increased sensitivity for changes in membrane potential and calcium to BK channels. Ryanodine-sensitive calcium-release channels (RyR3) in the sarcoplasmic reticulum (SR) control the process. To elucidate the mechanism of BK channel activation, we performed whole-cell and perforated-patch clamp experiments in freshly isolated cerebral and mesenteric artery vascular smooth muscle cells (VSMC) from Sprague-Dawley rats, BK beta1 gene-deficient (-/-), BK alpha (-/-), RyR3 (-/-) and wild-type mice. The 17,18-EETeTr (100 nm) increased tetraethylammonium (1 mm)-sensitive outward K(+) currents in VSMC from wild-type rats and wild-type mice. The effects were not inhibited by the epoxyeicosatrienoic acid (EET) antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (10 mum). BK channel currents were increased 3.5-fold in VSMC from BK beta1 (-/-) mice, whereas a 2.9-fold stimulation was observed in VSMC from RyR3 (-/-) mice (at membrane voltage 60 mV). The effects were similar compared with those observed in cells from wild-type mice. The BK current increase was neither influenced by strong internal calcium buffering (Ca(2)(+), 100 nm), nor by external calcium influx. The 17,18-EETeTr did not induce outward currents in VSMC BK alpha (-/-) cells. We next tested the vasodilator effects of 17,18-EETeTr on isolated arteries of BK alpha-deficient mice. Vasodilatation was largely inhibited in cerebral and mesenteric arteries isolated from BK alpha (-/-) mice compared with that observed in wild-type and BK beta1 (-/-) arteries. We conclude that 17,18-EETeTr represents an endogenous BK channel agonist and vasodilator. Since 17,18-EETeTr is active in small arteries lacking BK beta1, the data further suggest that BK alpha represents the molecular target for the principal action of 17,18-EETeTr. Finally, the action of 17,18-EETeTr is not mediated by changes of the internal global calcium concentration or local SR calcium release events.


Asunto(s)
Ácidos Araquidónicos/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Subunidades de Proteína/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Calcio/fisiología , Señalización del Calcio/fisiología , Arterias Cerebrales/citología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Masculino , Potenciales de la Membrana/fisiología , Arterias Mesentéricas/citología , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Ratones , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Técnicas de Placa-Clamp , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Ratas , Ratas Sprague-Dawley , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/fisiología
14.
Am J Physiol Cell Physiol ; 293(1): C45-54, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17329399

RESUMEN

Large-conductance Ca(2+)-activated K(+) (BK) channels are reported to be essential for NADPH oxidase-dependent microbial killing and innate immunity in leukocytes. Using human peripheral blood and mouse bone marrow neutrophils, pharmacological targeting, and BK channel gene-deficient (BK(-/-)) mice, we stimulated NADPH oxidase activity with 12-O-tetradecanoylphorbol-13-acetate (PMA) and performed patch-clamp recordings on isolated neutrophils. Although PMA stimulated NADPH oxidase activity as assessed by O(2)(-) and H(2)O(2) production, our patch-clamp experiments failed to show PMA-activated BK channel currents in neutrophils. In our studies, PMA induced slowly activating currents, which were insensitive to the BK channel inhibitor iberiotoxin. Instead, the currents were blocked by Zn(2+), which indicates activation of proton channel currents. BK channels are gated by elevated intracellular Ca(2+) and membrane depolarization. We did not observe BK channel currents, even during extreme depolarization to +140 mV and after elevation of intracellular Ca(2+) by N-formyl-L-methionyl-L-leucyl-phenylalanine. As a control, we examined BK channel currents in cerebral and tibial artery smooth muscle cells, which showed characteristic BK channel current pharmacology. Iberiotoxin did not block killing of Staphylococcus aureus or Candida albicans. Moreover, we addressed the role of BK channels in a systemic S. aureus and Yersinia enterocolitica mouse infection model. After 3 and 5 days of infection, we found no differences in the number of bacteria in spleen and kidney between BK(-/-) and BK(+/+) mice. In conclusion, our experiments failed to identify functional BK channels in neutrophils. We therefore conclude that BK channels are not essential for innate immunity.


Asunto(s)
Inmunidad Innata , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Infecciones Estafilocócicas/metabolismo , Yersiniosis/metabolismo , Animales , Actividad Bactericida de la Sangre , Calcio/metabolismo , Cloruros/farmacología , Modelos Animales de Enfermedad , Activación Enzimática , Activadores de Enzimas/farmacología , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Gran Conductancia Activados por el Calcio/deficiencia , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Neutrófilos/inmunología , Neutrófilos/microbiología , Técnicas de Placa-Clamp , Péptidos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus , Acetato de Tetradecanoilforbol/farmacología , Yersiniosis/genética , Yersiniosis/inmunología , Yersiniosis/microbiología , Yersinia enterocolitica , Compuestos de Zinc/farmacología
15.
Hypertension ; 44(3): 271-6, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15302842

RESUMEN

Periadventitial adipose tissue produces vasoactive substances that influence vascular contraction. Earlier studies addressed this issue in aorta, a vessel that does not contribute to peripheral vascular resistance. We tested the hypothesis that periadventitial adipose tissue modulates contraction of smaller arteries more relevant to blood pressure regulation. We studied mesenteric artery rings surrounded by periadventitial adipose tissue from adult male Sprague-Dawley rats. The contractile response to serotonin, phenylephrine, and endothelin I was markedly reduced in intact vessels compared with vessels without periadventitial fat. The contractile response to U46619 or depolarizing high K+-containing solutions (60 mmol/L) was similar in vessels with and without periadventitial fat. The K+ channel opener cromakalim induced relaxation of vessels precontracted by serotonin but not by U46619 or high K+-containing solutions (60 mmol/L), suggesting that K+ channels are involved. The intracellular membrane potential of smooth muscle cells was more hyperpolarized in intact vessels than in vessels without periadventitial fat. Both the anticontractile effect and membrane hyperpolarization of periadventitial fat were abolished by inhibition of delayed-rectifier K+ (K(v)) channels with 4-aminopyridine (2 mmol/L) or 3,4-diaminopyridine (1 mmol/L). Blocking other K+ channels with glibenclamide (3 micromol/L), apamin (1 micromol/L), iberiotoxin (100 nmol/L), tetraethylammonium ions (1 mmol/L), tetrapentylammonium ions (10 micromol/L), or Ba2+ (3 micromol/L) had no effect. Longitudinal removal of half the perivascular tissue reduced the anticontractile effect of fat by almost 50%, whereas removal of the endothelium had no effect. We suggest that visceral periadventitial adipose tissue controls mesenteric arterial tone by inducing vasorelaxation via K(v) channel activation in vascular smooth muscle cells.


Asunto(s)
4-Aminopiridina/análogos & derivados , Tejido Adiposo/fisiología , Arteria Mesentérica Superior/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Resistencia Vascular/fisiología , Vasodilatación/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , 4-Aminopiridina/farmacología , Tejido Adiposo/metabolismo , Amifampridina , Animales , Apamina/farmacología , Bario/farmacología , Cromakalim/farmacología , Canales de Potasio de Tipo Rectificador Tardío , Endotelina-1/farmacología , Gliburida/farmacología , Masculino , Arteria Mesentérica Superior/efectos de los fármacos , Péptidos/farmacología , Fenilefrina/farmacología , Potasio/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Compuestos de Amonio Cuaternario/farmacología , Ratas , Ratas Sprague-Dawley , Serotonina/farmacología , Tetraetilamonio/farmacología , Vasodilatación/efectos de los fármacos , Vísceras
16.
Am J Physiol Regul Integr Comp Physiol ; 287(3): R685-95, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15031138

RESUMEN

To study the mechanisms by which missense mutations in alpha-tropomyosin cause familial hypertrophic cardiomyopathy, we generated transgenic rats overexpressing alpha-tropomyosin with one of two disease-causing mutations, Asp(175)Asn or Glu(180)Gly, and analyzed phenotypic changes at molecular, morphological, and physiological levels. The transgenic proteins were stably integrated into the sarcomere, as shown by immunohistochemistry using a human-specific anti-alpha-tropomyosin antibody, ARG1. In transgenic rats with either alpha-tropomyosin mutation, molecular markers of cardiac hypertrophy were induced. Ca(2+) sensitivity of cardiac skinned-fiber preparations from animals with mutation Asp(175)Asn, but not Glu(180)Gly, was decreased. Furthermore, elevated frequency and amplitude of spontaneous Ca(2+) waves were detected only in cardiomyocytes from animals with mutation Asp(175)Asn, suggesting an increase in intracellular Ca(2+) concentration compensating for the reduced Ca(2+) sensitivity of isometric force generation. Accordingly, in Langendorff-perfused heart preparations, myocardial contraction and relaxation were accelerated in animals with mutation Asp(175)Asn. The results allow us to propose a hypothesis of the pathogenetic changes caused by alpha-tropomyosin mutation Asp(175)Asn in familial hypertrophic cardiomyopathy on the basis of changes in Ca(2+) handling as a sensitive mechanism to compensate for alterations in sarcomeric structure.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/fisiopatología , Corazón/fisiopatología , Mutación Missense , Tropomiosina/genética , Animales , Animales Modificados Genéticamente , Asparagina , Ácido Aspártico , Biomarcadores/análisis , Calcio/metabolismo , Calcio/farmacología , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/metabolismo , Expresión Génica , Ácido Glutámico , Glicina , Ventrículos Cardíacos , Humanos , Inmunohistoquímica , Técnicas In Vitro , Fibras Musculares Esqueléticas/efectos de los fármacos , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Ratas , Sarcómeros/metabolismo , Transgenes , Tropomiosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA