Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 101(4): 1487-1559, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33769101

RESUMEN

Brain function critically depends on a close matching between metabolic demands, appropriate delivery of oxygen and nutrients, and removal of cellular waste. This matching requires continuous regulation of cerebral blood flow (CBF), which can be categorized into four broad topics: 1) autoregulation, which describes the response of the cerebrovasculature to changes in perfusion pressure; 2) vascular reactivity to vasoactive stimuli [including carbon dioxide (CO2)]; 3) neurovascular coupling (NVC), i.e., the CBF response to local changes in neural activity (often standardized cognitive stimuli in humans); and 4) endothelium-dependent responses. This review focuses primarily on autoregulation and its clinical implications. To place autoregulation in a more precise context, and to better understand integrated approaches in the cerebral circulation, we also briefly address reactivity to CO2 and NVC. In addition to our focus on effects of perfusion pressure (or blood pressure), we describe the impact of select stimuli on regulation of CBF (i.e., arterial blood gases, cerebral metabolism, neural mechanisms, and specific vascular cells), the interrelationships between these stimuli, and implications for regulation of CBF at the level of large arteries and the microcirculation. We review clinical implications of autoregulation in aging, hypertension, stroke, mild cognitive impairment, anesthesia, and dementias. Finally, we discuss autoregulation in the context of common daily physiological challenges, including changes in posture (e.g., orthostatic hypotension, syncope) and physical activity.


Asunto(s)
Circulación Cerebrovascular/fisiología , Trastornos Cerebrovasculares/fisiopatología , Homeostasis/fisiología , Animales , Humanos , Enfermedades del Sistema Nervioso/fisiopatología , Acoplamiento Neurovascular
2.
Circ Res ; 134(6): 810-832, 2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38484034

RESUMEN

Hypertension is extremely common, affecting approximately 1 in every 2 adults globally. Chronic hypertension is the leading modifiable risk factor for cardiovascular disease and premature mortality worldwide. Despite considerable efforts to define mechanisms that underlie hypertension, a potentially major component of the disease, the role of circadian biology has been relatively overlooked in both preclinical models and humans. Although the presence of daily and circadian patterns has been observed from the level of the genome to the whole organism, the functional and structural impact of biological rhythms, including mechanisms such as circadian misalignment, remains relatively poorly defined. Here, we review the impact of daily rhythms and circadian systems in regulating blood pressure and the onset, progression, and consequences of hypertension. There is an emphasis on the impact of circadian biology in relation to vascular disease and end-organ effects that, individually or in combination, contribute to complex phenotypes such as cognitive decline and the loss of cardiac and brain health. Despite effective treatment options for some individuals, control of blood pressure remains inadequate in a substantial portion of the hypertensive population. Greater insight into circadian biology may form a foundation for novel and more widely effective molecular therapies or interventions to help in the prevention, treatment, and management of hypertension and its related pathophysiology.


Asunto(s)
Enfermedades Cardiovasculares , Hipertensión , Adulto , Humanos , Presión Sanguínea/fisiología , Ritmo Circadiano , Corazón
3.
Cell ; 139(5): 1012-21, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19945383

RESUMEN

The amygdala processes and directs inputs and outputs that are key to fear behavior. However, whether it directly senses fear-evoking stimuli is unknown. Because the amygdala expresses acid-sensing ion channel-1a (ASIC1a), and ASIC1a is required for normal fear responses, we hypothesized that the amygdala might detect a reduced pH. We found that inhaled CO(2) reduced brain pH and evoked fear behavior in mice. Eliminating or inhibiting ASIC1a markedly impaired this activity, and localized ASIC1a expression in the amygdala rescued the CO(2)-induced fear deficit of ASIC1a null animals. Buffering pH attenuated fear behavior, whereas directly reducing pH with amygdala microinjections reproduced the effect of CO(2). These data identify the amygdala as an important chemosensor that detects hypercarbia and acidosis and initiates behavioral responses. They also give a molecular explanation for how rising CO(2) concentrations elicit intense fear and provide a foundation for dissecting the bases of anxiety and panic disorders.


Asunto(s)
Acidosis/metabolismo , Amígdala del Cerebelo/metabolismo , Trastornos de Ansiedad/metabolismo , Dióxido de Carbono/metabolismo , Canales Iónicos Sensibles al Ácido , Animales , Bicarbonatos/metabolismo , Humanos , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Pletismografía , Canales de Sodio/genética , Canales de Sodio/metabolismo
4.
Annu Rev Physiol ; 82: 275-295, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-31618600

RESUMEN

Cerebral small vessel disease (SVD) is characterized by changes in the pial and parenchymal microcirculations. SVD produces reductions in cerebral blood flow and impaired blood-brain barrier function, which are leading contributors to age-related reductions in brain health. End-organ effects are diverse, resulting in both cognitive and noncognitive deficits. Underlying phenotypes and mechanisms are multifactorial, with no specific treatments at this time. Despite consequences that are already considerable, the impact of SVD is predicted to increase substantially with the growing aging population. In the face of this health challenge, the basic biology, pathogenesis, and determinants of SVD are poorly defined. This review summarizes recent progress and concepts in this area, highlighting key findings and some major unanswered questions. We focus on phenotypes and mechanisms that underlie microvascular aging, the greatest risk factor for cerebrovascular disease and its subsequent effects.


Asunto(s)
Envejecimiento/patología , Enfermedades de los Pequeños Vasos Cerebrales/patología , Animales , Capilares/crecimiento & desarrollo , Capilares/patología , Circulación Cerebrovascular , Humanos
5.
Am J Physiol Heart Circ Physiol ; 323(6): H1108-H1117, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36269650

RESUMEN

Changes in vascular structure contribute to vascular events and loss of brain health. We examined changes in cerebral arterioles at the onset of hypertension and the hypothesis that alterations during hypertension would recover with the return of mean arterial pressure (MAP) to normal. MAP was measured with radiotelemetry in awake male C57BL/6J mice at baseline and during infusion of vehicle or angiotensin II (ANG II, 1.4 mg/kg/day using osmotic pumps) for 28 days, followed by a 28-day recovery. With ANG II treatment, MAP increased through day 28. On day 30, MAP began to recover, reaching levels not different from vehicle on day 37. We measured intravascular pressure, diameter, wall thickness (WT), wall:lumen ratio (W:L), cross-sectional area (CSA), and slope of the tangential elastic modulus (ET) in maximally dilated arterioles. Variables were similar in both groups at day 1, with no significant change with vehicle treatment. With ANG II treatment, CSA, WT, and W:L increased on days 7-28. Internal and external diameter was reduced at 14 and 28 days. ET versus wall stress was reduced on days 7-28. During recovery, the diameter remained at days 14 and 28 values, whereas other variables returned partly or completely to normal. Thus, CSA, WT, W:L, and ET versus wall stress changed rapidly during hypertension and recovered with MAP. In contrast, inward remodeling developed slowly and did not recover. This lack of recovery has mechanistic implications for the long-term impact of hypertension on vascular determinants of brain health.NEW & NOTEWORTHY Changes in vascular structure contribute to vascular events and loss of brain health. We examined the inherent structural plasticity of cerebral arterioles during and after a period of hypertension. Arteriolar wall thickness, diameter, wall-to-lumen ratio, and biological stiffness changed rapidly during hypertension and recovered with blood pressure. In contrast, inward remodeling developed slowly and did not recover. This lack of recovery of arteriolar diameter has implications for the long-term impact of hypertension on vascular determinants of brain health.


Asunto(s)
Presión Arterial , Hipertensión , Animales , Ratones , Masculino , Ratones Endogámicos C57BL , Presión Sanguínea , Arteriolas , Angiotensina II/farmacología
6.
Stroke ; 52(7): 2404-2413, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34107734

RESUMEN

Background and Purpose: Hypertension is a leading risk factor for cerebrovascular disease and loss of brain health. While the brain renin-angiotensin system (RAS) contributes to hypertension, its potential impact on the local vasculature is unclear. We tested the hypothesis that activation of the brain RAS would alter the local vasculature using a modified deoxycorticosterone acetate (DOCA) model. Methods: C57BL/6 mice treated with DOCA (50 mg SQ; or shams) were given tap H2O and H2O with 0.9% NaCl for 1 to 3 weeks. Results: In isolated cerebral arteries and parenchymal arterioles from DOCA-treated male mice, endothelium- and nitric oxide-dependent dilation was progressively impaired, while mesenteric arteries were unaffected. In contrast, cerebral endothelial function was not significantly affected in female mice treated with DOCA. In males, mRNA expression of renal Ren1 was markedly reduced while RAS components (eg, Agt and Ace) were increased in both brain and cerebral arteries with central RAS activation. In NZ44 reporter mice expressing GFP (green fluorescent protein) driven by the angiotensin II type 1A receptor (Agtr1a) promoter, DOCA increased GFP expression ≈3-fold in cerebral arteries. Impaired endothelial responses were restored to normal by losartan, an AT1R (angiotensin II type 1 receptor) antagonist. Last, DOCA treatment produced inward remodeling of parenchymal arterioles. Conclusions: These findings suggest activation of the central and cerebrovascular RAS impairs endothelial (nitric oxide dependent) signaling in brain through expression and activation of AT1R and sex-dependent effects. The central RAS may be a key contributor to vascular dysfunction in brain in a preclinical (low renin) model of hypertension. Because the brain RAS is also activated during aging and other diseases, a common mechanism may promote loss of endothelial and brain health despite diverse cause.


Asunto(s)
Trastornos Cerebrovasculares/metabolismo , Endotelio Vascular/metabolismo , Hipertensión/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Receptor de Angiotensina Tipo 1/biosíntesis , Sistema Renina-Angiotensina/fisiología , Animales , Trastornos Cerebrovasculares/inducido químicamente , Trastornos Cerebrovasculares/genética , Acetato de Desoxicorticosterona/toxicidad , Femenino , Hipertensión/inducido químicamente , Hipertensión/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/genética , Receptor de Angiotensina Tipo 1/genética , Sistema Renina-Angiotensina/efectos de los fármacos
8.
Circ Res ; 125(10): 907-920, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31451088

RESUMEN

RATIONALE: Precise regulation of cerebral blood flow is critical for normal brain function. Insufficient cerebral blood flow contributes to brain dysfunction and neurodegeneration. Carbon dioxide (CO2), via effects on local acidosis, is one of the most potent regulators of cerebral blood flow. Although a role for nitric oxide in intermediate signaling has been implicated, mechanisms that initiate CO2-induced vasodilation remain unclear. OBJECTIVE: Acid-sensing ion channel-1A (ASIC1A) is a proton-gated cation channel that is activated by extracellular acidosis. Based on work that implicated ASIC1A in the amygdala and bed nucleus of the stria terminalis in CO2-evoked and acid-evoked behaviors, we hypothesized that ASIC1A might also mediate microvascular responses to CO2. METHODS AND RESULTS: To test this hypothesis, we genetically and pharmacologically manipulated ASIC1A and assessed effects on CO2-induced dilation of cerebral arterioles in vivo. Effects of inhalation of 5% or 10% CO2 on arteriolar diameter were greatly attenuated in mice with global deficiency in ASIC1A (Asic1a-/-) or by local treatment with the ASIC inhibitor, psalmotoxin. Vasodilator effects of acetylcholine, which acts via endothelial nitric oxide synthase were unaffected, suggesting a nonvascular source of nitric oxide may be key for CO2 responses. Thus, we tested whether neurons may be the cell type through which ASIC1A influences microvessels. Using mice in which Asic1a was specifically disrupted in neurons, we found effects of CO2 on arteriolar diameter were also attenuated. CONCLUSIONS: Together, these data are consistent with a model wherein activation of ASIC1A, particularly in neurons, is critical for CO2-induced nitric oxide production and vasodilation. With these findings, ASIC1A emerges as major regulator of microvascular tone.


Asunto(s)
Canales Iónicos Sensibles al Ácido/deficiencia , Circulación Cerebrovascular/fisiología , Hipercapnia/metabolismo , Vasodilatación/fisiología , Canales Iónicos Sensibles al Ácido/genética , Animales , Dióxido de Carbono/farmacología , Circulación Cerebrovascular/efectos de los fármacos , Hipercapnia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico/metabolismo , Vasodilatación/efectos de los fármacos
10.
Circ Res ; 120(3): 449-471, 2017 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-28154097

RESUMEN

The consequences of cerebrovascular disease are among the leading health issues worldwide. Large and small cerebral vessel disease can trigger stroke and contribute to the vascular component of other forms of neurological dysfunction and degeneration. Both forms of vascular disease are driven by diverse risk factors, with hypertension as the leading contributor. Despite the importance of neurovascular disease and subsequent injury after ischemic events, fundamental knowledge in these areas lag behind our current understanding of neuroprotection and vascular biology in general. The goal of this review is to address select key structural and functional changes in the vasculature that promote hypoperfusion and ischemia, while also affecting the extent of injury and effectiveness of therapy. In addition, as damage to the blood-brain barrier is one of the major consequences of ischemia, we discuss cellular and molecular mechanisms underlying ischemia-induced changes in blood-brain barrier integrity and function, including alterations in endothelial cells and the contribution of pericytes, immune cells, and matrix metalloproteinases. Identification of cell types, pathways, and molecules that control vascular changes before and after ischemia may result in novel approaches to slow the progression of cerebrovascular disease and lessen both the frequency and impact of ischemic events.


Asunto(s)
Isquemia Encefálica/fisiopatología , Trastornos Cerebrovasculares/fisiopatología , Endotelio Vascular/fisiopatología , Accidente Cerebrovascular/fisiopatología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Isquemia Encefálica/metabolismo , Trastornos Cerebrovasculares/metabolismo , Endotelio Vascular/metabolismo , Humanos , Factores de Riesgo , Accidente Cerebrovascular/metabolismo
13.
Physiol Genomics ; 48(2): 124-34, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26534936

RESUMEN

The ligand activated nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) in the endothelium regulates vascular function and blood pressure (BP). We previously reported that transgenic mice (E-V290M) with selectively targeted endothelial-specific expression of dominant negative PPARγ exhibited endothelial dysfunction when treated with a high-fat diet, and exhibited an augmented pressor response to angiotensin II (ANG II). We hypothesize that interference with endothelial PPARγ would exacerbate ANG II-induced endothelial dysfunction. Endothelial function was examined in E-V290M mice infused with a subpressor dose of ANG II (120 ng·kg(-1)·min(-1)) or saline for 2 wk. ANG II infusion significantly impaired the responses to the endothelium-dependent agonist acetylcholine both in basilar and carotid arteries from E-V290M but not NT mice. This impairment was not due to increased BP, which was not significantly different in ANG II-infused E-V290M compared with NT mice. Superoxide levels, and expression of the pro-oxidant Nox2 gene was elevated, whereas expression of the anti-oxidant genes Catalase and SOD3 decreased in carotid arteries from ANG II-infused E-V290M mice. Increased p65 and decreased Iκ-Bα suggesting increased NF-κB activity was also observed in aorta from ANG II-infused E-V290M mice. The responses to acetylcholine were significantly improved both in basilar and carotid arteries after treatment with Tempol (1 mmol/l), a scavenger of superoxide. These findings provide evidence that interference with endothelial PPARγ accelerates ANG II-mediated endothelial dysfunction both in cerebral and conduit arteries through an oxidative stress-dependent mechanism, suggesting a role for endothelial PPARγ in protecting against ANG II-induced endothelial dysfunction.


Asunto(s)
Angiotensina II/metabolismo , Endotelio Vascular/metabolismo , PPAR gamma/metabolismo , Acetilcolina/metabolismo , Animales , Antioxidantes/metabolismo , Presión Sanguínea , Arterias Carótidas/patología , Catalasa/metabolismo , Dieta Alta en Grasa , Endotelio Vascular/patología , Frecuencia Cardíaca , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Estrés Oxidativo , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Fluorescencia , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo
14.
Am J Physiol Heart Circ Physiol ; 311(1): H146-56, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27106041

RESUMEN

Carotid artery disease is a major contributor to stroke and cognitive deficits. Angiotensin II (Ang II) promotes vascular dysfunction and disease through mechanisms that include the IL-6/STAT3 pathway. Here, we investigated the importance of suppressor of cytokine signaling 3 (SOCS3) in models of Ang II-induced vascular dysfunction. We examined direct effects of Ang II on carotid arteries from SOCS3-deficient (SOCS3(+/-)) mice and wild-type (WT) littermates using organ culture and then tested endothelial function with acetylcholine (ACh). A low concentration of Ang II (1 nmol/l) did not affect ACh-induced vasodilation in WT but reduced that of SOCS3(+/-) mice by ∼50% (P < 0.05). In relation to mechanisms, effects of Ang II in SOCS3(+/-) mice were prevented by inhibitors of STAT3, IL-6, NF-κB, or superoxide. Systemic Ang II (1.4 mg/kg per day for 14 days) also reduced vasodilation to ACh in WT. Surprisingly, SOCS3 deficiency prevented most of the endothelial dysfunction. To examine potential underlying mechanisms, we performed bone marrow transplantation. WT mice reconstituted with SOCS3(+/-) bone marrow were protected from Ang II-induced endothelial dysfunction, whereas reconstitution of SOCS3(+/-) mice with WT bone marrow exacerbated Ang II-induced effects. The SOCS3 genotype of bone marrow-derived cells did not influence direct effects of Ang II on vascular function. These data provide new mechanistic insight into the influence of SOCS3 on the vasculature, including divergent effects depending on the source of Ang II. Bone marrow-derived cells deficient in SOCS3 protect against systemic Ang II-induced vascular dysfunction.


Asunto(s)
Angiotensina II , Aorta/metabolismo , Arteria Basilar/metabolismo , Células de la Médula Ósea/metabolismo , Arterias Carótidas/metabolismo , Hipertensión/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Vasodilatación , Animales , Aorta/efectos de los fármacos , Aorta/fisiopatología , Arteria Basilar/efectos de los fármacos , Arteria Basilar/fisiopatología , Trasplante de Médula Ósea , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Genotipo , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Hipertensión/prevención & control , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Técnicas de Cultivo de Órganos , Fenotipo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Superóxidos/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/deficiencia , Proteína 3 Supresora de la Señalización de Citocinas/genética , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
15.
Cell Mol Neurobiol ; 36(2): 241-58, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26988697

RESUMEN

The impact of vascular risk factors on cognitive function has garnered much interest in recent years. The appropriate distribution of oxygen, glucose, and other nutrients by the cerebral vasculature is critical for proper cognitive performance. The cerebral microvasculature is a key site of vascular resistance and a preferential target for small vessel disease. While deleterious effects of vascular risk factors on microvascular function are known, the contribution of this dysfunction to cognitive deficits is less clear. In this review, we summarize current evidence for microvascular dysfunction in brain. We highlight effects of select vascular risk factors (hypertension, diabetes, and hyperhomocysteinemia) on the pial and parenchymal circulation. Lastly, we discuss potential links between microvascular disease and cognitive function, highlighting current gaps in our understanding.


Asunto(s)
Trastornos del Conocimiento/fisiopatología , Microvasos/fisiopatología , Animales , Enfermedades Cardiovasculares/patología , Humanos , Microcirculación , Factores de Riesgo , Resistencia Vascular
18.
Blood ; 119(13): 3176-83, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22186991

RESUMEN

Hyperhomocysteinemia confers a high risk for thrombotic vascular events, but homocysteine-lowering therapies have been ineffective in reducing the incidence of secondary vascular outcomes, raising questions regarding the role of homocysteine as a mediator of cardiovascular disease. Therefore, to determine the contribution of elevated homocysteine to thrombosis susceptibility, we studied Cbs(-/-) mice conditionally expressing a zinc-inducible mutated human CBS (I278T) transgene. Tg-I278T Cbs(-/-) mice exhibited severe hyperhomocysteinemia and endothelial dysfunction in cerebral arterioles. Surprisingly, however, these mice did not display increased susceptibility to arterial or venous thrombosis as measured by photochemical injury in the carotid artery, chemical injury in the carotid artery or mesenteric arterioles, or ligation of the inferior vena cava. A survey of hemostatic and hemodynamic parameters revealed no detectible differences between control and Tg-I278T Cbs(-/-) mice. Our data demonstrate that severe elevation in homocysteine leads to the development of vascular endothelial dysfunction but is not sufficient to promote thrombosis. These findings may provide insights into the failure of homocysteine-lowering trials in secondary prevention from thrombotic vascular events.


Asunto(s)
Modelos Animales de Enfermedad , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/patología , Ratones , Trombosis/etiología , Animales , Cistationina betasintasa/genética , Femenino , Pruebas Hematológicas , Hemodinámica/genética , Hemodinámica/fisiología , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Factores de Riesgo , Índice de Severidad de la Enfermedad
20.
Circ Res ; 111(11): 1446-58, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22962432

RESUMEN

RATIONALE: Activation of peroxisome proliferator-activated receptor-γ (PPARγ) by thiazolidinediones lowers blood pressure, whereas PPARγ mutations cause hypertension. Previous studies suggest these effects may be mediated through the vasculature, but the underlying mechanisms remain unclear. OBJECTIVE: To identify PPARγ mechanisms and transcriptional targets in vascular smooth muscle and their role in regulating resistance artery tone. METHODS AND RESULTS: We studied mesenteric artery (MA) from transgenic mice expressing dominant-negative (DN) mutant PPARγ driven by a smooth muscle cell-specific promoter. MA from transgenic mice exhibited a robust increase in myogenic tone. Patch clamp analysis revealed a reduced large conductance Ca(2+)-activated K(+) (BKCa) current in freshly dissociated smooth muscle cell from transgenic MA. Inhibition of protein kinase C corrected both enhanced myogenic constriction and impaired the large conductance Ca(2+)-activated K(+) channel function. Gene expression profiling revealed a marked loss of the regulator of G protein signaling 5 (RGS5) mRNA in transgenic MA, which was accompanied by a substantial increase in angiotensin II-induced constriction in MA. Small interfering RNA targeting RGS5 caused augmented myogenic tone in intact mesenteric arteries and increased activation of protein kinase C in smooth muscle cell cultures. PPARγ and PPARδ each bind to a PPAR response element close to the RGS5 promoter. RGS5 expression in nontransgenic MA was induced after activation of either PPARγ or PPARδ, an effect that was markedly blunted by DN PPARγ. CONCLUSIONS: We conclude that RGS5 in smooth muscle is a PPARγ and PPARδ target, which when activated blunts angiotensin II-mediated activation of protein kinase C, and preserves the large conductance Ca(2+)-activated K(+) channel activity, thus providing tight control of myogenic tone in the microcirculation.


Asunto(s)
Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Arterias Mesentéricas/fisiología , PPAR gamma/fisiología , Proteína Quinasa C/metabolismo , Proteínas RGS/metabolismo , Angiotensina II/farmacología , Animales , Western Blotting , Femenino , Perfilación de la Expresión Génica , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Mutación , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Naftalenos/farmacología , PPAR gamma/genética , PPAR gamma/metabolismo , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteínas RGS/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tetraetilamonio/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA