Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
3.
Hypertension ; 81(1): 24-33, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37937425

RESUMEN

Alterations in microcirculation play a crucial role in the pathogenesis of cardiovascular and metabolic disorders such as obesity and hypertension. The small resistance arteries of these patients show a typical remodeling, as indicated by an increase of media or total wall thickness to lumen diameter ratio that impairs organ flow reserve. The majority of blood vessels are surrounded by a fat depot which is termed perivascular adipose tissue (PVAT). In recent years, data from several studies have indicated that PVAT is an endocrine organ that can produce a variety of adipokines and cytokines, which may participate in the regulation of vascular tone, and the secretory profile varies with adipocyte phenotype and disease status. The PVAT of lean humans largely secretes the vasodilator adiponectin, which will act in a paracrine fashion to reduce peripheral resistance and improve nutrient uptake into tissues, thereby protecting against the development of hypertension and diabetes. In obesity, PVAT becomes enlarged and inflamed, and the bioavailability of adiponectin is reduced. The inevitable consequence is a rise in peripheral resistance with higher blood pressure. The interrelationship between obesity and hypertension could be explained, at least in part, by a cross-talk between microcirculation and PVAT. In this article, we propose an integrated pathophysiological approach of this relationship, in order to better clarify its role in obesity and hypertension, as the basis for effective and specific prevention and treatment.


Asunto(s)
Adiponectina , Hipertensión , Humanos , Adiponectina/metabolismo , Microcirculación , Tejido Adiposo/patología , Obesidad
4.
J Hypertens ; 41(10): 1668-1669, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37642596
5.
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
6.
J Hypertens ; 39(10): 1933, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34326280
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Am Heart J ; 204: 102-108, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30092411

RESUMEN

BACKGROUND: Ethnicity, along with a variety of genetic and environmental factors, is thought to influence the efficacy of antihypertensive therapies. Current UK guidelines use a "black versus white" approach; in doing so, they ignore the United Kingdom's largest ethnic minority: Asians from South Asia. STUDY DESIGN: The primary purpose of the AIM-HY INFORM trial is to identify potential differences in response to antihypertensive drugs used as mono- or dual therapy on the basis of self-defined ethnicity. A multicenter, prospective, open-label, randomized study with 2 parallel, independent trial arms (mono- and dual therapy), AIM-HY INFORM plans to enroll a total of 1,320 patients from across the United Kingdom. Those receiving monotherapy (n = 660) will enter a 3-treatment (amlodipine 10 mg od; lisinopril 20 mg od; chlorthalidone 25 mg od), 3-period crossover, lasting 24 weeks, whereas those receiving dual therapy (n = 660) will enter a 4-treatment (amlodipine 5 mg od and lisinopril 20 mg od; amlodipine 5 mg od and chlorthalidone 25 mg od; lisinopril 20 mg od and chlorthalidone 25 mg od; amiloride 10 mg od and chlorthalidone 25 mg od), 4-period crossover, lasting 32 weeks. Equal numbers of 3 ethnic groups (white, black/black British, and Asian/Asian British) will ultimately be recruited to each of the trial arms (ie, 220 participants per ethnic group per arm). Seated, automated, unattended, office, systolic blood pressure measured 8 weeks after each treatment period begins will serve as the primary outcome measure. CONCLUSION: AIM-HY INFORM is a prospective, open-label, randomized trial which aims to evaluate first- and second-line antihypertensive therapies for multiethnic populations.


Asunto(s)
Antihipertensivos/uso terapéutico , Hipertensión/tratamiento farmacológico , Hipertensión/etnología , Adolescente , Adulto , Anciano , Amlodipino/uso terapéutico , Pueblo Asiatico , Población Negra , Clortalidona/uso terapéutico , Estudios Cruzados , Esquema de Medicación , Quimioterapia Combinada , Hemodinámica , Humanos , Hipertensión/fisiopatología , Lisinopril/uso terapéutico , Persona de Mediana Edad , Estudios Prospectivos , Reino Unido , Población Blanca , Adulto Joven
13.
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
14.
Curr Hypertens Rep ; 20(5): 44, 2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29736674

RESUMEN

PURPOSE OF REVIEW: In this review, we discuss the role of perivascular adipose tissue (PVAT) in the modulation of vascular contractility and arterial pressure, focusing on the role of the renin-angiotensin-aldosterone system and oxidative stress/inflammation. RECENT FINDINGS: PVAT possesses a relevant endocrine-paracrine activity, which may be altered in several pathophysiological and clinical conditions. During the last two decades, it has been shown that PVAT may modulate vascular reactivity. It has also been previously demonstrated that inflammation in adipose tissue may be implicated in vascular dysfunction. In particular, adipocytes secrete a number of adipokines with various functions, as well as several vasoactive factors, together with components of the renin-angiotensin system which may act at local or at systemic level. It has been shown that the anti-contractile effect of PVAT is lost in obesity, probably as a consequence of the development of adipocyte hypertrophy, inflammation, and oxidative stress. Adipose tissue dysfunction is interrelated with inflammation and oxidative stress, thus contributing to endothelial dysfunction observed in several pathological and clinical conditions such as obesity and hypertension. Decreased local adiponectin level, macrophage recruitment and infiltration, and activation of renin-angiotensin-aldosterone system could play an important role in this regard.


Asunto(s)
Tejido Adiposo/fisiopatología , Vasos Sanguíneos/fisiopatología , Hipertensión/fisiopatología , Inflamación/fisiopatología , Obesidad/fisiopatología , Adipocitos/fisiología , Adipoquinas/fisiología , Animales , Humanos , Estrés Oxidativo , Sistema Renina-Angiotensina/fisiología , Vasodilatación/fisiología
16.
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
17.
J Am Heart Assoc ; 6(4)2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28400366

RESUMEN

BACKGROUND: Small artery pathophysiology is frequently invoked as a cause of obesity-related diastolic heart failure. However, evidence to support this hypothesis is scant, particularly in humans. METHODS AND RESULTS: To address this, we studied human small artery structure and function in obesity and looked for correlations between vascular parameters and diastolic function. Seventeen obese patients with metabolic syndrome and 5 control participants underwent echocardiography and subcutaneous gluteal fat biopsy. Small arteries were isolated from the biopsy and pressure myography was used to study endothelial function and wall structure. In comparison with the control group, small arteries from obese participants exhibited significant endothelial dysfunction, assessed as the vasodilatory response to acetylcholine and also pathological growth of the wall. For the obese participants, multiple regression analysis revealed an association between left atrial volume and both the small artery wall thickness (ß=0.718, P=0.02) and wall-to-lumen ratio (ß=0.605, P=0.02). Furthermore, the E:E' ratio was associated with wall-to-lumen ratio (ß=0.596, P=0.02) and inversely associated with interleukin-6 (ß=-0.868, P=0.03). By contrast, endothelial function did not correlate with any of the echocardiographic parameters studied. CONCLUSIONS: Although the small arteries studied were not cardiac in origin, our results support a role for small artery remodeling in the development of diastolic dysfunction in humans. Further direct examination of the structure and function of the myocardial resistance vasculature is now warranted, to elucidate the temporal association between metabolic risk factors, small artery injury, and diastolic impairment.


Asunto(s)
Arterias/fisiopatología , Síndrome Metabólico/fisiopatología , Obesidad/fisiopatología , Grasa Subcutánea/irrigación sanguínea , Remodelación Vascular , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda , Adulto , Anciano , Arterias/patología , Biopsia , Nalgas , Estudios de Casos y Controles , Diástole , Ecocardiografía Doppler , Humanos , Síndrome Metabólico/diagnóstico , Persona de Mediana Edad , Obesidad/diagnóstico , Factores de Riesgo , Vasodilatación , Disfunción Ventricular Izquierda/diagnóstico por imagen
18.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...