Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Vasc Res ; 59(5): 288-302, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35947969

RESUMEN

BACKGROUND: The mechanism of the perivascular adipose tissue (PVAT) anticontractile effect is well characterized in rodent visceral vascular beds; however, little is known about the mechanism of PVAT anticontractile function in subcutaneous vessels. In addition, we have previously shown that PVAT anticontractile function is nitric oxide synthase (NOS) dependent but have not investigated the roles of NOS isoforms. OBJECTIVE: Here, we examined PVAT anticontractile function in the mouse gracilis artery, a subcutaneous fat depot, in lean control and obese mice and investigated the mechanism in comparison to a visceral depot. METHOD: Using the wire myograph, we generated responses to noradrenaline and electrical field stimulation in the presence of pharmacological tools targeting components of the known PVAT anticontractile mechanism. In addition, we performed ex vivo "fat transplants" in the organ bath. RESULTS: The mechanism of PVAT anticontractile function is similar between subcutaneous and visceral PVAT depots. Both endothelial and neuronal NOS isoforms mediated the PVAT anticontractile effect. Loss of PVAT anticontractile function in obesity is independent of impaired vasoreactivity, and function can be restored in visceral PVAT by NOS activation. CONCLUSIONS: Targeting NOS isoforms may be useful in restoring PVAT anticontractile function in obesity, ameliorating increased vascular tone, and disease.


Asunto(s)
Tejido Adiposo , Obesidad , Ratones , Animales , Óxido Nítrico Sintasa de Tipo I/farmacología , Norepinefrina/farmacología , Ratones Obesos , Óxido Nítrico Sintasa , Isoformas de Proteínas/farmacología , Óxido Nítrico , Vasoconstricción
2.
Cardiovasc Drugs Ther ; 35(6): 1291-1304, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33687595

RESUMEN

PURPOSE: Perivascular adipose tissue (PVAT) exerts an anti-contractile effect which is vital in regulating vascular tone. This effect is mediated via sympathetic nervous stimulation of PVAT by a mechanism which involves noradrenaline uptake through organic cation transporter 3 (OCT3) and ß3-adrenoceptor-mediated adiponectin release. In obesity, autonomic dysfunction occurs, which may result in a loss of PVAT function and subsequent vascular disease. Accordingly, we have investigated abnormalities in obese PVAT, and the potential for exercise in restoring function. METHODS: Vascular contractility to electrical field stimulation (EFS) was assessed ex vivo in the presence of pharmacological tools in ±PVAT vessels from obese and exercised obese mice. Immunohistochemistry was used to detect changes in expression of ß3-adrenoceptors, OCT3 and tumour necrosis factor-α (TNFα) in PVAT. RESULTS: High fat feeding induced hypertension, hyperglycaemia, and hyperinsulinaemia, which was reversed using exercise, independent of weight loss. Obesity induced a loss of the PVAT anti-contractile effect, which could not be restored via ß3-adrenoceptor activation. Moreover, adiponectin no longer exerts vasodilation. Additionally, exercise reversed PVAT dysfunction in obesity by reducing inflammation of PVAT and increasing ß3-adrenoceptor and OCT3 expression, which were downregulated in obesity. Furthermore, the vasodilator effects of adiponectin were restored. CONCLUSION: Loss of neutrally mediated PVAT anti-contractile function in obesity will contribute to the development of hypertension and type II diabetes. Exercise training will restore function and treat the vascular complications of obesity.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Obesidad/fisiopatología , Obesidad/terapia , Condicionamiento Físico Animal/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hiperglucemia/inducido químicamente , Hiperinsulinismo/inducido químicamente , Hipertensión/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Receptores Adrenérgicos beta 3/efectos de los fármacos , Factor de Necrosis Tumoral alfa/efectos de los fármacos
3.
Am J Physiol Heart Circ Physiol ; 319(6): H1387-H1397, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33035443

RESUMEN

Perivascular adipose tissue (PVAT) depots are metabolically active and play a major vasodilator role in healthy lean individuals. In obesity, they become inflamed and eosinophil-depleted and the anticontractile function is lost with the development of diabetes and hypertension. Moreover, eosinophil-deficient ΔdblGATA-1 mice lack PVAT anticontractile function and exhibit hypertension. Here, we have investigated the effects of inducing eosinophilia on PVAT function in health and obesity. Control, obese, and ΔdblGATA-1 mice were administered intraperitoneal injections of interleukin-33 (IL-33) for 5 days. Conscious restrained blood pressure was measured, and blood was collected for glucose and plasma measurements. Wire myography was used to assess the contractility of mesenteric resistance arteries. IL-33 injections induced a hypereosinophilic phenotype. Obese animals had significant elevations in blood pressure, blood glucose, and plasma insulin, which were normalized with IL-33. Blood glucose and insulin levels were also lowered in lean treated mice. In arteries from control mice, PVAT exerted an anticontractile effect on the vessels, which was enhanced with IL-33 treatment. In obese mice, loss of PVAT anticontractile function was rescued by IL-33. Exogenous application of IL-33 to isolated arteries induced a rapidly decaying endothelium-dependent vasodilation. The therapeutic effects were not seen in IL-33-treated ΔdblGATA-1 mice, thereby confirming that the eosinophil is crucial. In conclusion, IL-33 treatment restored PVAT anticontractile function in obesity and reversed development of hypertension, hyperglycemia, and hyperinsulinemia. These data suggest that targeting eosinophil numbers in PVAT offers a novel approach to the treatment of hypertension and type 2 diabetes in obesity.NEW & NOTEWORTHY In this study, we have shown that administering IL-33 to obese mice will restore PVAT anticontractile function, and this is accompanied by normalized blood pressure, blood glucose, and plasma insulin. Moreover, the PVAT effect is enhanced in control mice given IL-33. IL-33 induced a hypereosinophilic phenotype in our mice, and the effects of IL-33 on PVAT function, blood pressure, and blood glucose are absent in eosinophil-deficient mice, suggesting that the effects of IL-33 are mediated via eosinophils.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Hipertensión/prevención & control , Interleucina-33/farmacología , Arterias Mesentéricas/efectos de los fármacos , Obesidad/tratamiento farmacológico , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Animales , Presión Arterial/efectos de los fármacos , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus Tipo 2/prevención & control , Modelos Animales de Enfermedad , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipoglucemiantes/farmacología , Masculino , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/metabolismo , Obesidad/fisiopatología
4.
Noncoding RNA ; 6(1)2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31979312

RESUMEN

Adipose tissue is an endocrine organ, capable of regulating distant physiological processes in other tissues via the release of adipokines into the bloodstream. Recently, circulating adipose-derived microRNAs (miRNAs) have been proposed as a novel class of adipokine, due to their capacity to regulate gene expression in tissues other than fat. Circulating levels of adipokines are known to be altered in obese individuals compared with typical weight individuals and are linked to poorer health outcomes. For example, obese individuals are known to be more prone to the development of some cancers, and less likely to achieve event-free survival following chemotherapy. The purpose of this review was twofold; first to identify circulating miRNAs which are reproducibly altered in obesity, and secondly to identify mechanisms by which these obesity-linked miRNAs might influence the sensitivity of tumors to treatment. We identified 8 candidate circulating miRNAs with altered levels in obese individuals (6 increased, 2 decreased). A second literature review was then performed to investigate if these candidates might have a role in mediating resistance to cancer treatment. All of the circulating miRNAs identified were capable of mediating responses to cancer treatment at the cellular level, and so this review provides novel insights which can be used by future studies which aim to improve obese patient outcomes.

5.
J Vasc Res ; 56(6): 320-332, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31550717

RESUMEN

BACKGROUND: Perivascular adipose tissue (PVAT) reduces vascular tone in isolated arteries in vitro, however there are no studies of PVAT effects on vascular tone in vivo. In vitro adipocyte ß3-adrenoceptors play a role in PVAT function via secretion of the vasodilator adiponectin. OBJECTIVE: We have investigated the effects of PVAT on vessel diameter in vivo, and the contributions of ß3-adrenoceptors and adiponectin. METHOD: In anaesthetised rats, sections of the intact mesenteric bed were visualised and the diameter of arteries was recorded. Arteries were stimulated with electrical field stimulation (EFS), noradrenaline (NA), arginine-vasopressin (AVP), and acetylcholine (Ach). RESULTS: We report that in vivo, stimulation of PVAT with EFS, NA, and AVP evokes a local anti-constrictive effect on the artery, whilst PVAT exerts a pro-contractile effect on arteries subjected to Ach. The anti-constrictive effect of PVAT stimulated with EFS and NA was significantly reduced using ß3-adrenoceptor inhibition, and activation of ß3-adrenoceptors potentiated the anti-constrictive effect of vessels stimulated with EFS, NA, and AVP. The ß3-adrenoceptor agonist had no effect on mesenteric arteries with PVAT removed. A blocking peptide for adiponectin receptor 1 polyclonal antibody reduced the PVAT anti-constrictive effect in arteries stimulated with EFS and NA, indicating that adiponectin may be the anti-constrictive factor released upon ß3-adrenoceptor activation. CONCLUSIONS: These results clearly demonstrate that PVAT plays a paracrine role in regulating local vascular tone in vivo, and therefore may contribute to the modulation of blood pressure. This effect is mediated via adipocyte ß3-adrenoceptors, which may trigger release of the vasodilator adiponectin.


Asunto(s)
Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Arterias Mesentéricas/metabolismo , Comunicación Paracrina , Receptores Adrenérgicos beta 3/metabolismo , Vasoconstricción , Vasodilatación , Tejido Adiposo/efectos de los fármacos , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Antagonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Estimulación Eléctrica , Masculino , Arterias Mesentéricas/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Ratas Wistar , Receptores Adrenérgicos beta 3/efectos de los fármacos , Transducción de Señal , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
6.
Physiol Rev ; 99(4): 1701-1763, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31339053

RESUMEN

Obesity is increasingly prevalent and is associated with substantial cardiovascular risk. Adipose tissue distribution and morphology play a key role in determining the degree of adverse effects, and a key factor in the disease process appears to be the inflammatory cell population in adipose tissue. Healthy adipose tissue secretes a number of vasoactive adipokines and anti-inflammatory cytokines, and changes to this secretory profile will contribute to pathogenesis in obesity. In this review, we discuss the links between adipokine dysregulation and the development of hypertension and diabetes and explore the potential for manipulating adipose tissue morphology and its immune cell population to improve cardiovascular health in obesity.


Asunto(s)
Tejido Adiposo/fisiopatología , Presión Sanguínea , Diabetes Mellitus/fisiopatología , Hipertensión/fisiopatología , Obesidad/fisiopatología , Adipoquinas/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Adiposidad , Animales , Diabetes Mellitus/epidemiología , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Humanos , Hipertensión/epidemiología , Hipertensión/inmunología , Hipertensión/metabolismo , Mediadores de Inflamación/metabolismo , Obesidad/epidemiología , Obesidad/inmunología , Obesidad/metabolismo , Fenotipo , Medición de Riesgo , Factores de Riesgo , Transducción de Señal , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología
7.
Cardiovasc Drugs Ther ; 33(2): 245-259, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30747398

RESUMEN

Perivascular adipose tissue (PVAT) is no longer recognised as simply a structural support for the vasculature, and we now know that PVAT releases vasoactive factors which modulate vascular function. Since the discovery of this function in 1991, PVAT research is rapidly growing and the importance of PVAT function in disease is becoming increasingly clear. Obesity is associated with a plethora of vascular conditions; therefore, the study of adipocytes and their effects on the vasculature is vital. PVAT contains an adrenergic system including nerves, adrenoceptors and transporters. In obesity, the autonomic nervous system is dysfunctional; therefore, sympathetic innervation of PVAT may be the key mechanistic link between increased adiposity and vascular disease. In addition, not all obese people develop vascular disease, but a common feature amongst those that do appears to be the inflammatory cell population in PVAT. This review will discuss what is known about sympathetic innervation of PVAT, and the links between nerve activation and inflammation in obesity. In addition, we will examine the therapeutic potential of exercise in sympathetic stimulation of adipose tissue.


Asunto(s)
Tejido Adiposo/inervación , Enfermedades Cardiovasculares/fisiopatología , Inflamación/fisiopatología , Obesidad/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Adipocitos/metabolismo , Adipoquinas/metabolismo , Fibras Adrenérgicas/metabolismo , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/prevención & control , Metabolismo Energético , Terapia por Ejercicio , Humanos , Inflamación/metabolismo , Inflamación/prevención & control , Mediadores de Inflamación/metabolismo , Obesidad/metabolismo , Obesidad/terapia , Sistema Nervioso Simpático/metabolismo
8.
Br J Pharmacol ; 175(18): 3685-3698, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29980164

RESUMEN

BACKGROUND AND PURPOSE: In response to noradrenaline, healthy perivascular adipose tissue (PVAT) exerts an anticontractile effect on adjacent small arterial tissue. Organ bath solution transfer experiments have demonstrated the release of PVAT-derived relaxing factors that mediate this function. The present studies were designed to investigate the mechanism responsible for the noradrenaline-induced PVAT anticontractile effect. EXPERIMENTAL APPROACH: In vitro rat small arterial contractile function was assessed using wire myography in the presence and absence of PVAT and the effects of sympathomimetic stimulation on the PVAT environment explored using Western blotting and assays of organ bath buffer. KEY RESULTS: PVAT elicited an anticontractile effect in response to noradrenaline but not phenylephrine stimulation. In arteries surrounded by intact PVAT, the ß3 -adrenoceptor agonist, CL-316243, reduced the vasoconstrictor effect of phenylephrine but not noradrenaline. Kv 7 channel inhibition using XE 991 reversed the noradrenaline-induced anticontractile effect in exogenously applied PVAT studies. Adrenergic stimulation of PVAT with noradrenaline and CL-316243, but not phenylephrine, was associated with increased adipocyte-derived NO production, and the contractile response to noradrenaline was augmented following incubation of exogenous PVAT with L-NMMA. PVAT from eNOS-/- mice had no anticontractile effect. Assays of adipocyte cAMP demonstrated an increase with noradrenaline stimulation implicating Gαs signalling in this process. CONCLUSIONS AND IMPLICATIONS: We have shown that adipocyte-located ß3 -adrenoceptor stimulation leads to activation of Gαs signalling pathways with increased cAMP and the release of adipocyte-derived NO. This process is dependent upon Kv 7 channel function. We conclude that adipocyte-derived NO plays a central role in anticontractile activity when rodent PVAT is stimulated by noradrenaline.


Asunto(s)
Adipocitos/efectos de los fármacos , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Arterias/efectos de los fármacos , Vasos Sanguíneos/citología , Óxido Nítrico/metabolismo , Receptores Adrenérgicos beta 3/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Adipocitos/metabolismo , Adiponectina/metabolismo , Animales , Arterias/fisiología , Vasos Sanguíneos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Masculino , Óxido Nítrico/biosíntesis , Norepinefrina/farmacología , Canales de Potasio/agonistas , Ratas , Ratas Sprague-Dawley , Transducción de Señal
10.
Arterioscler Thromb Vasc Biol ; 38(4): 880-891, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29496660

RESUMEN

OBJECTIVE: Healthy perivascular adipose tissue (PVAT) exerts an anticontractile effect on resistance arteries which is vital in regulating arterial tone. Activation of ß3-adrenoceptors by sympathetic nerve-derived NA (noradrenaline) may be implicated in this effect and may stimulate the release of the vasodilator adiponectin from adipocytes. Understanding the mechanisms responsible is vital for determining how PVAT may modify vascular resistance in vivo. APPROACH AND RESULTS: Electrical field stimulation profiles of healthy C57BL/6J mouse mesenteric resistance arteries were characterized using wire myography. During electrical field stimulation, PVAT elicits a reproducible anticontractile effect, which is endothelium independent. To demonstrate the release of an anticontractile factor, the solution surrounding stimulated exogenous PVAT was transferred to a PVAT-denuded vessel. Post-transfer contractility was significantly reduced confirming that stimulated PVAT releases a transferable anticontractile factor. Sympathetic denervation of PVAT using tetrodotoxin or 6-hydroxydopamine completely abolished the anticontractile effect. ß3-adrenoceptor antagonist SR59203A reduced the anticontractile effect, although the PVAT remained overall anticontractile. When the antagonist was used in combination with an OCT3 (organic cation transporter 3) inhibitor, corticosterone, the anticontractile effect was completely abolished. Application of an adiponectin receptor-1 blocking peptide significantly reduced the anticontractile effect in +PVAT arteries. When used in combination with the ß3-adrenoceptor antagonist, there was no further reduction. In adiponectin knockout mice, the anticontractile effect is absent. CONCLUSIONS: The roles of PVAT are 2-fold. First, sympathetic stimulation in PVAT triggers the release of adiponectin via ß3-adrenoceptor activation. Second, PVAT acts as a reservoir for NA, preventing it from reaching the vessel and causing contraction.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/inervación , Tejido Adiposo/metabolismo , Arterias Mesentéricas/metabolismo , Norepinefrina/metabolismo , Comunicación Paracrina , Sistema Nervioso Simpático/metabolismo , Vasodilatación , Adiponectina/genética , Adiponectina/metabolismo , Animales , Técnicas In Vitro , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Adrenérgicos beta 3/metabolismo , Transducción de Señal , Vasoconstricción
11.
Sci Rep ; 7: 44571, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28303919

RESUMEN

Obesity impairs the relaxant capacity of adipose tissue surrounding the vasculature (PVAT) and has been implicated in resultant obesity-related hypertension and impaired glucose intolerance. Resident immune cells are thought to regulate adipocyte activity. We investigated the role of eosinophils in mediating normal PVAT function. Healthy PVAT elicits an anti-contractile effect, which was lost in mice deficient in eosinophils, mimicking the obese phenotype, and was restored upon eosinophil reconstitution. Ex vivo studies demonstrated that the loss of PVAT function was due to reduced bioavailability of adiponectin and adipocyte-derived nitric oxide, which was restored after eosinophil reconstitution. Mechanistic studies demonstrated that adiponectin and nitric oxide are released after activation of adipocyte-expressed ß3 adrenoceptors by catecholamines, and identified eosinophils as a novel source of these mediators. We conclude that adipose tissue eosinophils play a key role in the regulation of normal PVAT anti-contractile function.


Asunto(s)
Tejido Adiposo/metabolismo , Eosinófilos/metabolismo , Hipertensión/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Tejido Adiposo/patología , Animales , Aorta/metabolismo , Aorta/patología , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Catecolaminas/metabolismo , Dieta Alta en Grasa , Humanos , Hipertensión/complicaciones , Hipertensión/patología , Ratones , Óxido Nítrico/metabolismo , Obesidad/complicaciones , Obesidad/patología , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 36(7): 1377-85, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27174097

RESUMEN

OBJECTIVE: Perivascular adipose tissue (PVAT) exerts an anticontractile effect in response to various vasoconstrictor agonists, and this is lost in obesity. A recent study reported that bariatric surgery reverses the damaging effects of obesity on PVAT function. However, PVAT function has not been characterized after weight loss induced by caloric restriction, which is often the first line treatment for obesity. APPROACH AND RESULTS: Contractility studies were performed using wire myography on small mesenteric arteries with and without PVAT from control, diet-induced obese, calorie restricted and sustained weight loss rats. Changes in the PVAT environment were assessed using immunohistochemistry. PVAT from healthy animals elicited an anticontractile effect in response to norepinephrine. This was abolished in diet-induced obesity through a mechanism involving increased local tumor necrosis factor-α and reduced nitric oxide bioavailability within PVAT. Sustained weight loss led to improvement in PVAT function associated with restoration of adipocyte size, reduced tumor necrosis factor-α, and increased nitric oxide synthase function. This was associated with reversal of obesity-induced hypertension and normalization of plasma adipokine levels, including leptin and insulin. CONCLUSIONS: We have shown that diet-induced weight loss reverses obesity-induced PVAT damage through a mechanism involving reduced inflammation and increased nitric oxide synthase activity within PVAT. These data reveal inflammation and nitric oxide synthase, particularly endothelial nitric oxide synthase, as potential targets for the treatment of PVAT dysfunction associated with obesity and metabolic syndrome.


Asunto(s)
Tejido Adiposo/fisiopatología , Adiposidad , Restricción Calórica , Arterias Mesentéricas/fisiopatología , Obesidad/dietoterapia , Vasoconstricción , Pérdida de Peso , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Mediadores de Inflamación/metabolismo , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Obesidad/metabolismo , Obesidad/patología , Obesidad/fisiopatología , Comunicación Paracrina , Ratas Sprague-Dawley , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
13.
J Hypertens ; 33(5): 1039-45, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25909701

RESUMEN

BACKGROUND/AIMS: In patients with obesity, there is increased inflammation with attendant oxidative stress in perivascular adipose tissue. This has functional consequences with loss of vasodilator adipokine bioavailability. Part of the inflammatory response is mediated by increased activation of the renin-angiotensin-aldosterone axis. Therefore, this study was designed to investigate whether angiotensin-converting enzyme inhibitors or angiotensin receptor blockers can improve the anticontractile function of perivascular adipose tissue. METHODS: Segments of rat mesenteric small artery were dissected and mounted in a wire myograph and contracted to incremental doses of norepinephrine in the presence and absence of perivascular adipose tissue and in conditions of normal oxygenation or after hypoxia and incubated with captopril or telmisartan. RESULTS: Vessels with perivascular adipose tissue contracted significantly less than arteries with perivascular adipose tissue removed under normal oxygenation conditions, indicating that perivascular adipose tissue exerts an anticontractile effect. Hypoxia induced a loss of this anticontractile effect which could be completely prevented with captopril or telmisartan. CONCLUSION: The in-vitro creation of a hypoxic environment can simulate the loss of anticontractile perivascular adipose tissue function seen in vivo in obese patients, and this can be prevented using inhibitors of the renin-angiotensin cascade.


Asunto(s)
Tejido Adiposo/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Arteriolas/efectos de los fármacos , Obesidad/metabolismo , Vasoconstricción/efectos de los fármacos , Adipoquinas/metabolismo , Adulto , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Hipoxia , Técnicas In Vitro , Inflamación/metabolismo , Masculino , Arterias Mesentéricas/efectos de los fármacos , Norepinefrina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Sistema Renina-Angiotensina/efectos de los fármacos
14.
J Vasc Res ; 51(4): 305-14, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25300309

RESUMEN

Human essential hypertension is characterized by eutrophic inward remodeling of the resistance arteries with little evidence of hypertrophy. Upregulation of αVß3 integrin is crucial during this process. In order to investigate the role of focal adhesion kinase (FAK) activation in this process, the level of FAK Y397 autophosphorylation was studied in small blood vessels from young TGR(mRen2)27 animals as blood pressure rose and eutrophic inward remodeling took place. Between weeks 4 and 5, this process was completed and accompanied by a significant increase in FAK phosphorylation compared with normotensive control animals. Phosphorylated (p)FAK Y397 was coimmunoprecipitated with both ß1- and ß3-integrin-specific antibodies. In contrast, only a fraction (<10-fold) was coprecipitated with the ß3 integrin subunit in control vessels. Inhibition of eutrophic remodeling by cRGDfV treatment of TGR(mRen2)27 rats resulted in the development of smooth-muscle-cell hypertrophy and a significant further enhancement of FAK Y397 phosphorylation, but this time with exclusive coassociation of pFAK Y397 with integrin ß1. We established that phosphorylation of FAK Y397 with association with ß1 and ß3 integrins occurs with pressure-induced eutrophic remodeling. Inhibiting this process leads to an adaptive hypertrophic vascular response induced by a distinct ß1-mediated FAK phosphorylation pattern.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Hipertensión/fisiopatología , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Remodelación Vascular/fisiología , Resistencia Vascular/fisiología , Animales , Arterias/metabolismo , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Integrina alfaV/metabolismo , Fosforilación/fisiología , Ratas Sprague-Dawley , Ratas Transgénicas
15.
Arterioscler Thromb Vasc Biol ; 34(8): 1637-42, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24855062

RESUMEN

The concept that fat cells could influence the circulation and indeed cardiac function has been in existence for ≥20 years and has gained a wide interest and no less excitement as evidence has accrued to suggest that such effects may be profound enough to explain disease states, such as hypertension and metabolic changes associated with obesity and type II diabetes mellitus. This ATVB in Focus intends to examine our current knowledge in this field, and suggests mechanisms that may be responsible for normal perivascular function and how they become disordered in obesity. There is the tantalizing prospect of developing new therapeutic approaches to keep obese individuals healthy and redesignating type II diabetes mellitus as a vascular disease.


Asunto(s)
Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Vasos Sanguíneos/metabolismo , Obesidad/metabolismo , Comunicación Paracrina , Transducción de Señal , Enfermedades Vasculares/metabolismo , Tejido Adiposo/fisiopatología , Animales , Vasos Sanguíneos/fisiopatología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Síndrome Metabólico/metabolismo , Síndrome Metabólico/fisiopatología , Obesidad/epidemiología , Obesidad/fisiopatología , Obesidad/terapia , Factores de Riesgo , Enfermedades Vasculares/epidemiología , Enfermedades Vasculares/fisiopatología , Enfermedades Vasculares/terapia , Pérdida de Peso
16.
J Hypertens ; 32(6): 1264-74, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24751595

RESUMEN

AIMS: It has been demonstrated previously that inflammation in perivascular adipose tissue (PVAT) may be implicated in vascular dysfunction. The aim of this study was to investigate the functional responses of small mesenteric arteries in a hyperphagic animal model of obesity after chronic treatment with melatonin, an endogenous hormone with antioxidant and vasculoprotective properties. METHODS AND RESULTS: Ten obese mice (ob/ob) and 10 control lean mice (CLM) were treated with melatonin 100  mg/kg per day in the drinking water for 8 weeks. Mesenteric small resistance arteries were dissected and mounted on a wire myograph and a concentration-response to norepinephrine was evaluated in vessels with intact PVAT and after PVAT was removed and in the presence of iberiotoxin, a selective blocker of BKCA channels as well as under conditions of induced hypoxia in vitro. The presence of PVAT reduced the contractile response to norepinephrine in both ob/ob and CLM; however, the effect was significantly reduced in ob/ob. The anticontractile effect of PVAT completely disappeared with iberiotoxin preincubation. After melatonin treatment, inflammation was significantly ameliorated, and the contractile response in ob/ob and CLM was significantly reduced when PVAT was removed. Anticontractile effect of PVAT that is lost in obesity can be rescued using melatonin. A reduced expression of adiponectin and adiponectin receptor was observed in perivascular fat of ob/ob, whereas significant increase was observed in ob/ob treated with melatonin. CONCLUSION: Melatonin seems to exert a protective effect on arteries from both ob/ob and CLM, counteracting the adverse effect of hypoxia and iberiotoxin.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Melatonina/farmacología , Adiponectina/metabolismo , Tejido Adiposo/patología , Animales , Antioxidantes/uso terapéutico , Peso Corporal , Hipoxia/patología , Inflamación , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Ratones , Ratones Obesos , Microscopía Fluorescente , Norepinefrina/metabolismo , Obesidad/fisiopatología , Estrés Oxidativo , Péptidos/farmacología , Receptores de Adiponectina/metabolismo , Vasoconstricción/efectos de los fármacos
17.
Cardiovasc Res ; 101(1): 130-7, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24095868

RESUMEN

AIM: The aim of this study was to investigate the role of cGMP-dependent protein kinase (PKG) in mediating the anticontractile function of perivascular adipose tissue (PVAT) and whether its activation can rescue PVAT activity which is lost in an experimental model of inflammation. METHODS AND RESULTS: Contractile responses to norepinephrine were assessed using wire myography from small arterial segments obtained from PKG(-/-), PKG(+/+), adipo(-/-), and C57Bl6/J mice with and without PVAT during normal oxygenation and hypoxia. An anticontractile effect of PVAT was observed in control blood vessels. This was not present in arteries from PKG(-/-) or PKG(+/+) with inhibition of PKG signalling using DT-2/ODQ. Hypoxia-induced loss of PVAT function was rescued by ANP activation of PKG as there was no effect in blood vessels from PKG(-/-) mice or in the presence of DT-2. Solution transfer studies demonstrated that PKG was necessary for the normal paracrine effects of PVAT on smooth muscle and endothelium. PKG activation by atrial natriuretic peptide (ANP) did not restore the absent PVAT anticontractile capacity in arteries from adiponectin(-/-) mice; however, inhibition of PKG did not further abrogate this effect suggesting dysregulation of PKG signalling pathways in this model. The absence of PKG was associated with reduced adipocyte adiponectin expression. CONCLUSION: PKG plays a key role in regulating normal PVAT function both in modulating anticontractile factor release from adipocytes as well as being essential for its downstream dilator function in arterial smooth muscle.


Asunto(s)
Tejido Adiposo/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Hipoxia/fisiopatología , Arterias Mesentéricas/fisiología , Vasoconstricción , Adiponectina/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miografía , Superóxido Dismutasa/metabolismo
18.
Am J Physiol Heart Circ Physiol ; 304(6): H786-95, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23292715

RESUMEN

This study aims to identify the potential mechanisms by which perivascular adipose tissue (PVAT) reduces tone in small arteries. Small mesenteric arteries from wild-type and large-conductance Ca(2+)-activated K(+) (BKCa) channel knockout mice were mounted on a wire myograph in the presence and absence of PVAT, and contractile responses to norepinephrine were assessed. Electrophysiology studies were performed in isolated vessels to measure changes in membrane potential produced by adiponectin. Contractile responses from wild-type mouse small arteries were significantly reduced in the presence of PVAT. This was not observed in the presence of a BKCa channel inhibitor or with nitric oxide synthase (NOS) inhibition or in BKCa or adiponectin knockout mice. Solution transfer experiments demonstrated the presence of an anticontractile factor released from PVAT. Adiponectin-induced vasorelaxation and hyperpolarization in wild-type arteries were not evident in the absence of or after inhibition of BKCa channels. PVAT from BKCa or adiponectin knockout mice failed to elicit an anticontractile response in wild-type arteries. PVAT releases adiponectin, which is an anticontractile factor. Its effect on vascular tone is mediated by activation of BKCa channels on vascular smooth muscle cells and adipocytes and by endothelial mechanisms.


Asunto(s)
Adiponectina/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Contracción Muscular/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Potenciales de la Membrana , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Óxido Nítrico/antagonistas & inhibidores , Norepinefrina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Vasoconstrictores/farmacología , Vasodilatación
19.
J Cardiovasc Dis Res ; 4(3): 164-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24396255

RESUMEN

INTRODUCTION: The aim of this study was to investigate the effect of erythropoietin on vascular contractility using an in vitro model of hypoxia replicating the hypoxic environment of blood vessels and surrounding adipose tissue in obesity. METHODS AND RESULTS: Pharmacological in vitro studies were carried out on small mesenteric arterial segments from male Wistar rats with and without perivascular fat and endothelium. Contractile responses were investigated by wire myography under normoxia, experimental hypoxia ± erythropoietin and l-NNA. Perivascular fat exerted an anticontractile effect which was lost following the induction of experimental hypoxia. Erythropoietin prevented the loss of the anticontractile capacity when vessels were incubated for one hour before the induction of hypoxia or throughout the period of hypoxia; this was found to be independent of the function of perivascular fat, as fat denuded arteries had a similar reduction in contractility (artery no fat + hypoxia vs. artery no fat + hypoxia + erythropoietin). The mechanism by which erythropoietin was exerting its effect was found to be partially endothelium dependent and associated with an increase of nitric oxide bioavailability as nitric oxide synthase inhibition prevented the effect. CONCLUSIONS: Whilst erythropoietin is working downstream from perivascular fat, it is possible that it may be therapeutically useful in obesity when hypoxia and inflammation reduce the normal activity of perivascular fat.

20.
Front Biosci (Elite Ed) ; 3(3): 968-77, 2011 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-21622105

RESUMEN

C-Myb is a DNA-binding transcription factor that functions in apoptosis, proliferation and differentiation. The role of c-Myb in vascular injury has been investigated previously both in vitro and in vivo, where knock-down of c-Myb is known to lead to a reduction in proliferation and an increase in apoptosis of vascular smooth muscle cells (VSMCs). Reduction of c-Myb activity has also been shown to decrease neointimal formation in vivo, by reducing VSMC proliferation. In contrast, over-expression of c-Myb in vivo leads to increased survival rates in certain cell types. This review will look mainly at studies investigating c-Myb function in the vasculature, and evidence of signalling interactions which may be considered with regard to c-Myb as a possible target in the treatment of vasculoproliferative diseases.


Asunto(s)
Vasos Sanguíneos/fisiología , Endotelio Vascular/fisiología , Proteínas Proto-Oncogénicas c-myb/fisiología , Túnica Íntima/fisiología , Animales , Hematopoyesis , Humanos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...