Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Haematologica ; 106(8): 2147-2160, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32675225

RESUMEN

Clinical studies suggested that endothelial dysfunction and damage could be involved in the development and severity of acute graft-versus-host disease (aGVHD). Accordingly, we found increased percentage of apoptotic Casp3+ blood vessels in duodenal and colonic mucosa biopsies of patients with severe aGVHD. In murine experimental aGVHD, we detected severe microstructural endothelial damage and reduced endothelial pericyte coverage accompanied by reduced expression of endothelial tight junction proteins leading to increased endothelial leakage in aGVHD target organs. During intestinal aGVHD, colonic vasculature structurally changed, reflected by increased vessel branching and vessel diameter. Because recent data demonstrated an association of endothelium-related factors and steroid refractory aGVHD (SR-aGVHD), we analyzed human biopsies and murine tissues from SR-aGVHD. We found extensive tissue damage but low levels of alloreactive T cell infiltration in target organs, providing the rationale for T-cell independent SR-aGVHD treatment strategies. Consequently, we tested the endothelium-protective PDE5 inhibitor sildenafil, which reduced apoptosis and improved metabolic activity of endothelial cells in vitro. Accordingly, sildenafil treatment improved survival and reduced target organ damage during experimental SR-aGVHD. Our results demonstrate extensive damage, structural changes, and dysfunction of the vasculature during aGVHD. Therapeutic intervention by endothelium-protecting agents is an attractive approach for SR-aGVHD complementing current anti-inflammatory treatment options.


Asunto(s)
Enfermedad Injerto contra Huésped , Animales , Células Endoteliales , Endotelio , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Humanos , Ratones , Esteroides , Linfocitos T
2.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34768842

RESUMEN

Endothelial dysfunction (ED) comes with age, even without overt vessel damage such as that which occurs in atherosclerosis and diabetic vasculopathy. We hypothesized that aging would affect the downstream signalling of the endothelial nitric oxide (NO) system in the vascular smooth muscle (VSM). With this in mind, resistance mesenteric arteries were isolated from 13-week (juvenile) and 40-week-old (aged) mice and tested under isometric conditions using wire myography. Acetylcholine (ACh)-induced relaxation was reduced in aged as compared to juvenile vessels. Pretreatment with L-NAME, which inhibits nitrix oxide synthases (NOS), decreased ACh-mediated vasorelaxation, whereby differences in vasorelaxation between groups disappeared. Endothelium-independent vasorelaxation by the NO donor sodium nitroprusside (SNP) was similar in both groups; however, SNP bolus application (10-6 mol L-1) as well as soluble guanylyl cyclase (sGC) activation by runcaciguat (10-6 mol L-1) caused faster responses in juvenile vessels. This was accompanied by higher cGMP concentrations and a stronger response to the PDE5 inhibitor sildenafil in juvenile vessels. Mesenteric arteries and aortas did not reveal apparent histological differences between groups (van Gieson staining). The mRNA expression of the α1 and α2 subunits of sGC was lower in aged animals, as was PDE5 mRNA expression. In conclusion, vasorelaxation is compromised at an early age in mice even in the absence of histopathological alterations. Vascular smooth muscle sGC is a key element in aged vessel dysfunction.


Asunto(s)
Arterias Mesentéricas/fisiología , Guanilil Ciclasa Soluble/fisiología , Acetilcolina/farmacología , Factores de Edad , Animales , Aorta/metabolismo , GMP Cíclico/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Guanilato Ciclasa/metabolismo , Masculino , Arterias Mesentéricas/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
3.
Development ; 140(8): 1720-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23533173

RESUMEN

Arteriogenesis requires growth of pre-existing arteriolar collateral networks and determines clinical outcome in arterial occlusive diseases. Factors responsible for the development of arteriolar collateral networks are poorly understood. The Notch ligand Delta-like 4 (Dll4) promotes arterial differentiation and restricts vessel branching. We hypothesized that Dll4 may act as a genetic determinant of collateral arterial networks and functional recovery in stroke and hind limb ischemia models in mice. Genetic loss- and gain-of-function approaches in mice showed that Dll4-Notch signaling restricts pial collateral artery formation by modulating arterial branching morphogenesis during embryogenesis. Adult Dll4(+/-) mice showed increased pial collateral numbers, but stroke volume upon middle cerebral artery occlusion was not reduced compared with wild-type littermates. Likewise, Dll4(+/-) mice showed reduced blood flow conductance after femoral artery occlusion, and, despite markedly increased angiogenesis, tissue ischemia was more severe. In peripheral arteries, loss of Dll4 adversely affected excitation-contraction coupling in arterial smooth muscle in response to vasopressor agents and arterial vessel wall adaption in response to increases in blood flow, collectively contributing to reduced flow reserve. We conclude that Dll4-Notch signaling modulates native collateral formation by acting on vascular branching morphogenesis during embryogenesis. Dll4 furthermore affects tissue perfusion by acting on arterial function and structure. Loss of Dll4 stimulates collateral formation and angiogenesis, but in the context of ischemic diseases such beneficial effects are overruled by adverse functional changes, demonstrating that ischemic recovery is not solely determined by collateral number but rather by vessel functionality.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isquemia/fisiopatología , Proteínas de la Membrana/metabolismo , Microvasos/embriología , Morfogénesis/fisiología , Neovascularización Fisiológica/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Análisis de Varianza , Animales , Proteínas de Unión al Calcio , Inmunohistoquímica , Isquemia/metabolismo , Ratones , Microvasos/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Flujo Sanguíneo Regional/fisiología , Microtomografía por Rayos X
4.
Circ Res ; 115(2): 263-72, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24838176

RESUMEN

RATIONALE: Vascular wall stretch is the major stimulus for the myogenic response of small arteries to pressure. The molecular mechanisms are elusive, but recent findings suggest that G protein-coupled receptors can elicit a stretch response. OBJECTIVE: To determine whether angiotensin II type 1 receptors (AT1R) in vascular smooth muscle cells exert mechanosensitivity and identify the downstream ion channel mediators of myogenic vasoconstriction. METHODS AND RESULTS: We used mice deficient in AT1R signaling molecules and putative ion channel targets, namely AT1R, angiotensinogen, transient receptor potential channel 6 (TRPC6) channels, or several subtypes of the voltage-gated K+ (Kv7) gene family (KCNQ3, 4, or 5). We identified a mechanosensing mechanism in isolated mesenteric arteries and in the renal circulation that relies on coupling of the AT1R subtype a to a Gq/11 protein as a critical event to accomplish the myogenic response. Arterial mechanoactivation occurs after pharmacological block of AT1R and in the absence of angiotensinogen or TRPC6 channels. Activation of AT1R subtype a by osmotically induced membrane stretch suppresses an XE991-sensitive Kv channel current in patch-clamped vascular smooth muscle cells, and similar concentrations of XE991 enhance mesenteric and renal myogenic tone. Although XE991-sensitive KCNQ3, 4, and 5 channels are expressed in vascular smooth muscle cells, XE991-sensitive K+ current and myogenic contractions persist in arteries deficient in these channels. CONCLUSIONS: Our results provide definitive evidence that myogenic responses of mouse mesenteric and renal arteries rely on ligand-independent, mechanoactivation of AT1R subtype a. The AT1R subtype a signal relies on an ion channel distinct from TRPC6 or KCNQ3, 4, or 5 to enact vascular smooth muscle cell activation and elevated vascular resistance.


Asunto(s)
Arterias Mesentéricas/fisiología , Miocitos del Músculo Liso/fisiología , Presorreceptores/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Arteria Renal/fisiología , 4-Aminopiridina/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Antracenos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Células HEK293 , Hemorreología , Humanos , Canales de Potasio KCNQ/fisiología , Canal de Potasio KCNQ3/fisiología , Losartán/farmacología , Arterias Mesentéricas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Presión Osmótica , Receptor de Angiotensina Tipo 1/deficiencia , Receptor de Angiotensina Tipo 1/genética , Arteria Renal/citología , Canales Catiónicos TRPC/fisiología , Canal Catiónico TRPC6 , Transcripción Genética , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología
5.
Arterioscler Thromb Vasc Biol ; 34(9): 1827-30, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25012133

RESUMEN

Perivascular adipose tissue has been recognized unequivocally as a major player in the pathology of metabolic and cardiovascular diseases. Through its production of adipokines and the release of other thus far unidentified factors, this recently discovered adipose tissue modulates vascular regulation and the myogenic response. After the discovery of its ability to diminish the vessel's response to vasoconstrictors, a new paradigm established adipose-derived relaxing factor (ADRF) as a paracrine smooth muscle cells' potassium channel opener that could potentially help combat vascular dysfunction. This review will discuss the role of ADRF in vascular dysfunction in obesity and hypertension, the different potassium channels that can be activated by this factor, and describes new pharmacological tools that can mimic the ADRF effect and thus can be beneficial against vascular dysfunction in cardiovascular disease.


Asunto(s)
Tejido Adiposo/fisiopatología , Factores Biológicos/fisiología , Vasos Sanguíneos/patología , Enfermedades Cardiovasculares/fisiopatología , Canales de Potasio KCNQ/metabolismo , Adipoquinas/metabolismo , Tejido Adiposo/patología , Animales , Enfermedades Cardiovasculares/metabolismo , Endotelio Vascular/fisiopatología , Humanos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/antagonistas & inhibidores , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Obesidad/metabolismo , Obesidad/fisiopatología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Vasoconstrictores/farmacología
6.
Hypertension ; 81(3): 561-571, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38354270

RESUMEN

BACKGROUND: Small arteries exhibit resting tone, a partially contracted state that maintains arterial blood pressure. In arterial smooth muscle cells, potassium channels control contraction and relaxation. Perivascular adipose tissue (PVAT) has been shown to exert anticontractile effects on the blood vessels. However, the mechanisms by which PVAT signals small arteries, and their relevance remain largely unknown. We aimed to uncover key molecular components in adipose-vascular coupling. METHODS: A wide spectrum of genetic mouse models targeting Kcnq3, Kcnq4, and Kcnq5 genes (Kcnq3-/-, Kcnq4-/-, Kcnq5-/-, Kcnq5dn/dn, Kcnq4-/-/Kcnq5dn/dn, and Kcnq4-/-/Kcnq5-/-), telemetry blood pressure measurements, targeted lipidomics, RNA-Seq profiling, wire-myography, patch-clamp, and sharp-electrode membrane potential measurements was used. RESULTS: We show that PVAT causes smooth muscle cell KV7.5 family of voltage-gated potassium (K+) channels to hyperpolarize the membrane potential. This effect relaxes small arteries and regulates blood pressure. Oxygenation of polyunsaturated fats generates oxylipins, a superclass of lipid mediators. We identified numerous oxylipins released by PVAT, which potentiate vasodilatory action in small arteries by opening smooth muscle cell KV7.5 family of voltage-gated potassium (K+) channels. CONCLUSIONS: Our results reveal a key molecular function of the KV7.5 family of voltage-gated potassium (K+) channels in the adipose-vascular coupling, translating PVAT signals, particularly oxylipins, to the central physiological function of vasoregulation. This novel pathway opens new therapeutic perspectives.


Asunto(s)
Oxilipinas , Vasodilatación , Animales , Ratones , Tejido Adiposo , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Oxilipinas/metabolismo , Potasio/metabolismo
7.
Sci Rep ; 12(1): 3038, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35194063

RESUMEN

Transient receptor potential channel subfamily C, member 6 (TRPC6), a non-selective cation channel that controls influx of Ca2+ and other monovalent cations into cells, is widely expressed in the kidney. TRPC6 gene variations have been linked to chronic kidney disease but its role in acute kidney injury (AKI) is unknown. Here we aimed to investigate the putative role of TRPC6 channels in AKI. We used Trpc6-/- mice and pharmacological blockade (SH045 and BI-749327), to evaluate short-term AKI outcomes. Here, we demonstrate that neither Trpc6 deficiency nor pharmacological inhibition of TRPC6 influences the short-term outcomes of AKI. Serum markers, renal expression of epithelial damage markers, tubular injury, and renal inflammatory response assessed by the histological analysis were similar in wild-type mice compared to Trpc6-/- mice as well as in vehicle-treated versus SH045- or BI-749327-treated mice. In addition, we also found no effect of TRPC6 modulation on renal arterial myogenic tone by using blockers to perfuse isolated kidneys. Therefore, we conclude that TRPC6 does not play a role in the acute phase of AKI. Our results may have clinical implications for safety and health of humans with TRPC6 gene variations, with respect to mutated TRPC6 channels in the response of the kidney to acute ischemic stimuli.


Asunto(s)
Lesión Renal Aguda/genética , Variación Genética , Isquemia/genética , Riñón/irrigación sanguínea , Resultados Negativos , Canal Catiónico TRPC6/genética , Canal Catiónico TRPC6/fisiología , APACHE , Lesión Renal Aguda/patología , Animales , Calcio/metabolismo , Isquemia/patología , Riñón/metabolismo , Ratones Transgénicos
8.
J Am Heart Assoc ; 11(4): e022070, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35132870

RESUMEN

Background Blood pressure and tissue perfusion are controlled in part by the level of intrinsic (myogenic) arterial tone. However, many of the molecular determinants of this response are unknown. We previously found that mice with targeted disruption of the gene encoding the angiotensin II type 1a receptor (AT1AR) (Agtr1a), the major murine angiotensin II type 1 receptor (AT1R) isoform, showed reduced myogenic tone; however, uncontrolled genetic events (in this case, gene ablation) can lead to phenotypes that are difficult or impossible to interpret. Methods and Results We tested the mechanosensitive function of AT1R using tamoxifen-inducible smooth muscle-specific AT1aR knockout (smooth muscle-Agtr1a-/-) mice and studied downstream signaling cascades mediated by Gq/11 and/or ß-arrestins. FR900359, Sar1Ile4Ile8-angiotensin II (SII), TRV120027 and TRV120055 were used as selective Gq/11 inhibitor and biased agonists to activate noncanonical ß-arrestin and canonical Gq/11 signaling of the AT1R, respectively. Myogenic and Ang II-induced constrictions were diminished in the perfused renal vasculature, mesenteric and cerebral arteries of smooth muscle-Agtr1a-/- mice. Similar effects were observed in arteries of global mutant Agtr1a-/- but not Agtr1b-/- mice. FR900359 decreased myogenic tone and angiotensin II-induced constrictions whereas selective biased targeting of AT1R-ß-arrestin signaling pathways had no effects. Conclusions This study demonstrates that myogenic arterial constriction requires Gq/11-dependent signaling pathways of mechanoactivated AT1R but not G protein-independent, noncanonical pathways in smooth muscle cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Receptor de Angiotensina Tipo 1 , Vasoconstricción , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Angiotensina II/metabolismo , Animales , Arterias Cerebrales/metabolismo , Ratones , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , beta-Arrestinas/metabolismo
9.
Front Med (Lausanne) ; 6: 107, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31245374

RESUMEN

Only a few years ago, alamandine was found to be a member of the protective arm of the renin-angiotensin system. It turned out to be an endogenous ligand of the G protein-coupled receptor MrgD. So far, MrgD had predominantly been studied in a neuronal context. The expression of the receptor in non-neuronal tissue showed hitherto unknown effects mediated by MrgD, most strikingly alamandine-induced vasodilation. Alamandine being a part of the non-classical renin-angiotensin system, a protective role of receptor activation seemed natural. This review summarizes the different effects of MrgD activation by alamandine in vasculature, in the central nervous system, and in organs as kidney and heart. Alamandine and MrgD are promising novel drug targets to protect the kidney and heart through anti-hypertensive actions.

10.
Front Med (Lausanne) ; 6: 128, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31263699

RESUMEN

Hypertension is ranked as the third cause of disability-adjusted life-years. The percentage of the population suffering from hypertension will continue to increase over the next years. Renovascular disease is one of the most common causes of secondary hypertension. Vascular changes seen in hypertension are partially based on dysfunctional calcium signaling. This signaling can be studied using calcium indicators (loading dyes and genetically encoded calcium indicators; GECIs). Most progress in development has been seen in GECIs, which are used in an increasing number of publications concerning calcium signaling in vasculature and the kidney. The use of transgenic mouse models expressing GECIs will facilitate new possibilities to study dysfunctional calcium signaling in a cell type-specific manner, thus helping to identify more specific targets for treatment of (renal) hypertension.

11.
J Am Heart Assoc ; 8(9): e010090, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31030596

RESUMEN

Background Hypertension is the major risk factor for cardiovascular disease, the most common cause of death worldwide. Resistance arteries are capable of adapting their diameter independently in response to pressure and flow-associated shear stress. Ryanodine receptors (RyRs) are major Ca2+-release channels in the sarcoplasmic reticulum membrane of myocytes that contribute to the regulation of contractility. Vascular smooth muscle cells exhibit 3 different RyR isoforms (RyR1, RyR2, and RyR3), but the impact of individual RyR isoforms on adaptive vascular responses is largely unknown. Herein, we generated tamoxifen-inducible smooth muscle cell-specific RyR2-deficient mice and tested the hypothesis that vascular smooth muscle cell RyR2s play a specific role in elementary Ca2+ signaling and adaptive vascular responses to vascular pressure and/or flow. Methods and Results Targeted deletion of the Ryr2 gene resulted in a complete loss of sarcoplasmic reticulum-mediated Ca2+-release events and associated Ca2+-activated, large-conductance K+ channel currents in peripheral arteries, leading to increased myogenic tone and systemic blood pressure. In the absence of RyR2, the pulmonary artery pressure response to sustained hypoxia was enhanced, but flow-dependent effects, including blood flow recovery in ischemic hind limbs, were unaffected. Conclusions Our results establish that RyR2-mediated Ca2+-release events in VSCM s specifically regulate myogenic tone (systemic circulation) and arterial adaptation in response to changes in pressure (hypoxic lung model), but not flow. They further suggest that vascular smooth muscle cell-expressed RyR2 deserves scrutiny as a therapeutic target for the treatment of vascular responses in hypertension and chronic vascular diseases.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo , Animales , Aorta/metabolismo , Aorta/fisiopatología , Arterias/metabolismo , Arterias/fisiopatología , Presión Sanguínea/fisiología , Señalización del Calcio , Miembro Posterior/irrigación sanguínea , Hipoxia/metabolismo , Hipoxia/fisiopatología , Flujometría por Láser-Doppler , Pulmón/irrigación sanguínea , Ratones , Ratones Noqueados , Músculo Liso Vascular/fisiopatología , Miografía , Técnicas de Placa-Clamp , Inhibidores de Fosfodiesterasa/farmacología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Vasoconstricción
12.
Br J Pharmacol ; 174(2): 150-162, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28000293

RESUMEN

BACKGROUND AND PURPOSE: KV 7.1 voltage-gated potassium channels are expressed in vascular smooth muscle cells (VSMC) of diverse arteries, including mesenteric arteries. Based on pharmacological evidence using R-L3 (KV 7.1 channel opener), HMR1556, chromanol 293B (KV 7.1 channel blockers), stimulation of these channels has been suggested to evoke profound relaxation in various vascular beds of rats. However, the specificity of these drugs in vivo is uncertain. EXPERIMENTAL APPROACH: We used Kcnq1-/- mice and pharmacological tools to determine whether KV 7.1 channels play a role in the regulation of arterial tone. KEY RESULTS: R-L3 produced similar concentration-dependent relaxations (EC50  ~ 1.4 µM) of arteries from wild-type (Kcnq1+/+ ) and Kcnq1-/- mice, pre-contracted with either phenylephrine or 60 mM KCl. This relaxation was not affected by 10 µM chromanol 293B, 10 µM HMR1556 or 30 µM XE991 (pan-KV 7 channel blocker). The anti-contractile effects of the perivascular adipose tissue (PVAT) were normal in Kcnq1-/- arteries. Chromanol 293B and HMR1556 did not affect the anti-contractile effects of (PVAT). Isolated VSMCs from Kcnq1-/- mice exhibited normal peak KV currents. The KV 7.2-5 channel opener retigabine caused similar relaxations in Kcnq1-/- and wild-type vessels. CONCLUSION AND IMPLICATIONS: We conclude that KV 7.1 channels were apparently not involved in the control of arterial tone by α1 -adrenoceptor agonists and PVAT. In addition, R-L3 is an inappropriate pharmacological tool for studying the function of native vascular KV 7.1 channels in mice.


Asunto(s)
Aorta/metabolismo , Canal de Potasio KCNQ1/metabolismo , Animales , Aorta/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Canal de Potasio KCNQ1/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Piperidinas/farmacología , Relación Estructura-Actividad , Tiazoles/farmacología , Compuestos de Tosilo/farmacología
13.
J Clin Invest ; 124(2): 675-86, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24401273

RESUMEN

High blood pressure is the leading risk factor for death worldwide. One of the hallmarks is a rise of peripheral vascular resistance, which largely depends on arteriole tone. Ca2+-activated chloride currents (CaCCs) in vascular smooth muscle cells (VSMCs) are candidates for increasing vascular contractility. We analyzed the vascular tree and identified substantial CaCCs in VSMCs of the aorta and carotid arteries. CaCCs were small or absent in VSMCs of medium-sized vessels such as mesenteric arteries and larger retinal arterioles. In small vessels of the retina, brain, and skeletal muscle, where contractile intermediate cells or pericytes gradually replace VSMCs, CaCCs were particularly large. Targeted disruption of the calcium-activated chloride channel TMEM16A, also known as ANO1, in VSMCs, intermediate cells, and pericytes eliminated CaCCs in all vessels studied. Mice lacking vascular TMEM16A had lower systemic blood pressure and a decreased hypertensive response following vasoconstrictor treatment. There was no difference in contractility of medium-sized mesenteric arteries; however, responsiveness of the aorta and small retinal arterioles to the vasoconstriction-inducing drug U46619 was reduced. TMEM16A also was required for peripheral blood vessel contractility, as the response to U46619 was attenuated in isolated perfused hind limbs from mutant mice. Out data suggest that TMEM16A plays a general role in arteriolar and capillary blood flow and is a promising target for the treatment of hypertension.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Canales de Cloruro/metabolismo , Hipertensión/fisiopatología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Anoctamina-1 , Arteriolas/patología , Presión Sanguínea/fisiología , Encéfalo/metabolismo , Clonación Molecular , ADN Complementario/metabolismo , Electrofisiología , Antagonistas de Estrógenos/farmacología , Células HEK293 , Humanos , Hipertensión/tratamiento farmacológico , Potenciales de la Membrana/efectos de los fármacos , Arterias Mesentéricas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Proteínas de Neoplasias/metabolismo , Pericitos/metabolismo , Retina/metabolismo , Tamoxifeno/farmacología , Factores de Tiempo , Resistencia Vascular , Vasoconstrictores/farmacología
14.
Hypertension ; 61(1): 151-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23184384

RESUMEN

KCNQ channels have been identified in arterial smooth muscle. However, their role in vasoregulation and chronic vascular diseases remains elusive. We tested the hypothesis that KCNQ channels contribute to periadventitial vasoregulation in peripheral skeletal muscle arteries by perivascular adipose tissue and that they represent novel targets to rescue periadventitial vascular dysfunction. Two models, spontaneously hypertensive rats and New Zealand obese mice, were studied using quantitative polymerase chain reaction, the patch-clamp technique, membrane potential measurements, myography of isolated vessels, and blood pressure telemetry. In rat Gracilis muscle arteries, anticontractile effects of perivascular fat were inhibited by the KCNQ channel blockers XE991 and linopirdine but not by other selective K(+) channel inhibitors. Accordingly, XE991 and linopirdine blocked noninactivating K(+) currents in freshly isolated Gracilis artery smooth muscle cells. mRNAs of several KCNQ channel subtypes were detected in those arteries, with KCNQ4 channels being dominant. In spontaneously hypertensive rats, the anticontractile effect of perivascular fat in Gracilis muscle arteries was largely reduced compared with Wistar rats. However, the vasodilator effects of KCNQ channel openers and mRNA expression of KCNQ channels were normal. Furthermore, KCNQ channel openers restored the diminished anticontractile effects of perivascular fat in spontaneously hypertensive rats. Moreover, KCNQ channel openers reduced arterial blood pressure in both models of hypertension independent of ganglionic blockade. Thus, our data suggest that KCNQ channels play a pivotal role in periadventitial vasoregulation of peripheral skeletal muscle arteries, and KCNQ channel opening may be an effective mechanism to improve impaired periadventitial vasoregulation and associated hypertension.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Presión Arterial/fisiología , Arterias/metabolismo , Canales de Potasio KCNQ/metabolismo , Músculo Esquelético/irrigación sanguínea , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Antracenos/farmacología , Presión Arterial/efectos de los fármacos , Arterias/efectos de los fármacos , Indoles/farmacología , Contracción Isométrica/efectos de los fármacos , Contracción Isométrica/fisiología , Canales de Potasio KCNQ/genética , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Piridinas/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Wistar
15.
PLoS One ; 7(8): e41951, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22870268

RESUMEN

BACKGROUND: Hydrogen sulfide (H(2)S) is a potent vasodilator. However, the complex mechanisms of vasoregulation by H(2)S are not fully understood. We tested the hypotheses that (1) H(2)S exerts vasodilatory effects by opening KCNQ-type voltage-dependent (K(v)) K(+) channels and (2) that H(2)S-producing cystathionine-γ-lyase (CSE) in perivascular adipose tissue plays a major role in this pathway. METHODOLOGY/PRINCIPAL FINDINGS: Wire myography of rat and mouse aortas was used. NaHS and 5-(4-hydroxyphenyl)-3H-1,2-dithiole-3-thione (ADTOH) were used as H(2)S donors. KCNQ-type K(v) channels were blocked by XE991. 4-Propargylglycine (PPG) and ß-cyano-l-alanine (BCA), or 2-(aminooxy)-acetic acid (AOAA) were used as inhibitors of CSE or cystathionine-ß-synthase (CBS), respectively. NaHS and ADTOH produced strong vasorelaxation in rat and mouse aortas, which were abolished by KCNQ channel inhibition with XE991. Perivascular adipose tissue (PVAT) exerted an anticontractile effect in these arteries. CSE inhibition by PPG and BCA reduced this effect in aortas from rats but not from mice. CBS inhibition with AOAA did not inhibit the anticontractile effects of PVAT. XE991, however, almost completely suppressed the anticontractile effects of PVAT in both species. Exogenous l-cysteine, substrate for the endogenous production of H(2)S, induced vasorelaxation only at concentrations >5 mmol/l, an effect unchanged by CSE inhibition. CONCLUSIONS/SIGNFICANCE: Our results demonstrate potent vasorelaxant effects of H(2)S donors in large arteries of both rats and mice, in which XE991-sensitive KCNQ-type channel opening play a pivotal role. CSE-H(2)S seems to modulate the effect of adipocyte-derived relaxing factor in rat but not in mouse aorta. The present study provides novel insight into the interaction of CSE-H(2)S and perivascular adipose tissue. Furthermore, with additional technical advances, a future clinical approach targeting vascular H(2)S/KCNQ pathways to influence states of vascular dysfunction may be possible.


Asunto(s)
Aorta/enzimología , Cistationina gamma-Liasa/metabolismo , Sulfuro de Hidrógeno/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Contaminantes Atmosféricos/farmacología , Animales , Aorta/citología , Cisteína/farmacología , Activación del Canal Iónico/efectos de los fármacos , Canales de Potasio KCNQ/metabolismo , Masculino , Ratones , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley
16.
J Mol Med (Berl) ; 89(12): 1219-29, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21818582

RESUMEN

Central angiotensin II (AngII) plays an important role in the regulation of the sympathetic nervous system. The underlining molecular mechanisms are largely unknown. Spinophilin (SPL) is a regulator of G protein-coupled receptor signaling. Deletion of SPL induces sympathetically mediated arterial hypertension in mice. We tested the hypothesis that SPL restrains blood pressure (BP) by regulating AngII activity. We equipped SPL(-/-) and SPL(+/+) mice with telemetric devices and applied AngII (1.0 mg kg(-1) day(-1), minipumps) or the AngII subtype 1 receptor (AT1-R) blocker valsartan (50 mg kg(-1) day(-1), gavage). We assessed autonomic nervous system activity through intraperitoneal application of trimethaphan, metoprolol, and atropine. We also tested the effect of intracerebroventricular (icv) AngII on blood pressure in SPL(-/-) and in SPL(+/+) mice. Chronic infusion of AngII upregulates SPL expression in the hypothalamus of SPL(+/+) mice. Compared with SPL(+/+) mice, SPL(-/-) mice showed a greater increase in daytime BP with AngII (19.2 ± 0.8 vs. 13.5 ± 1.6 mmHg, p < 0.05). SPL(-/-) showed a greater depressor response to valsartan. BP and heart rate decreased more with trimethaphan and metoprolol in AngII-treated SPL(-/-) than in AngII-treated SPL(+/+) mice. SPL(-/-) mice responded more to icv AngII. Furthermore, brainstem AT1-R and AngII type 2 receptor (AT2-R) expression was reduced in SPL(-/-) mice. AngII treatment normalized AT1-R and AT2-R expression levels. In summary, our findings suggest that SPL restrains AngII-mediated sympathetic nervous system activation. SPL is a hitherto unrecognized molecule with regard to central blood pressure control and may pave the way to novel strategies for the treatment of hypertension.


Asunto(s)
Angiotensina II/fisiología , Presión Sanguínea/fisiología , Proteínas de Microfilamentos/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas , Encéfalo/metabolismo , Regulación de la Expresión Génica , Frecuencia Cardíaca , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/genética , Receptor de Angiotensina Tipo 2/metabolismo , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo , Vasopresinas/genética , Vasopresinas/metabolismo
17.
J Hypertens ; 28(9): 1875-82, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20577128

RESUMEN

BACKGROUND: Perivascular adipose tissue secretes an adipocyte-derived relaxing factor (ADRF) that opens voltage-dependent K (Kv) channels in peripheral arteries. We studied the role of KCNQ-type Kv channels and tested the hypothesis that hydrogen sulfide (H2S) could be an ADRF. METHODS: We performed isometric contraction studies on systemic arteries of rats and mice. RESULTS: In mesenteric arteries and aortas without perivascular adipose tissue, the KCNQ channel openers retigabine, VRX0530727, VRX0621238, and VRX0621688 produced concentration-dependent vasorelaxation; VRX0621688 was the most potent vasodilator. The KCNQ inhibitor XE991 (30 micromol/l) blocked the effects of both the drugs and ADRF. Inhibitors of cystathionine gamma lyase (CSE) beta-cyano-L-alanine (BCA, 5 mmol/l) and 4-propargyl glycine (PPG, 10 mmol/l) also blocked the relaxations. CSE is expressed in perivascular adipose tissue and endogenously generates H2S. The H2S donor NaHS produced concentration-dependent vasorelaxation, which was also blocked by XE991. The vasodilatory capacities of retigabine, VRX0530727, VRX0621238, and VRX0621688 were preserved following inhibition of H2S generation in perivascular fat. CONCLUSION: We suggest that KCNQ channel opening is a powerful mechanism to produce vasorelaxation of systemic arteries in rats and mice. Furthermore, KCNQ channels play a major role in the paracrine control of vascular tone by perivascular adipose tissue, which is at least in part mediated or modulated by H2S. In conditions of reduced H2S release from perivascular adipose tissue, these paracrine effects can be mimicked by synthetic KCNQ channel openers.


Asunto(s)
Arterias/efectos de los fármacos , Arterias/fisiología , Sulfuro de Hidrógeno/metabolismo , Canales de Potasio KCNQ/agonistas , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Tejido Adiposo/fisiología , Animales , Antracenos/farmacología , Aorta/efectos de los fármacos , Aorta/fisiología , Carbamatos/farmacología , Técnicas In Vitro , Contracción Isométrica/efectos de los fármacos , Contracción Isométrica/fisiología , Canales de Potasio KCNQ/antagonistas & inhibidores , Canales de Potasio KCNQ/fisiología , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , Comunicación Paracrina/efectos de los fármacos , Comunicación Paracrina/fisiología , Fenilendiaminas/farmacología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA