Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 27(6): 2244-55, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23457218

RESUMEN

We previously reported that the sympathetic neurotransmitter neuropeptide Y (NPY) is potently angiogenic, primarily through its Y2 receptor, and that endogenous NPY is crucial for capillary angiogenesis in rodent hindlimb ischemia. Here we sought to identify the source of NPY responsible for revascularization and its mechanisms of action. At d 3, NPY(-/-) mice demonstrated delayed recovery of blood flow and limb function, consistent with impaired collateral conductance, while ischemic capillary angiogenesis was reduced (~70%) at d 14. This biphasic temporal response was confirmed by 2 peaks of NPY activation in rats: a transient early increase in neuronally derived plasma NPY and increase in platelet NPY during late-phase recovery. Compared to NPY-null platelets, collagen-activated NPY-rich platelets were more mitogenic (~2-fold vs. ~1.6-fold increase) for human microvascular endothelial cells, and Y2/Y5 receptor antagonists ablated this difference in proliferation. In NPY(+/+) mice, ischemic angiogenesis was prevented by platelet depletion and then restored by transfusion of platelets from NPY(+/+) mice, but not NPY(-/-) mice. In thrombocytopenic NPY(-/-) mice, transfusion of wild-type platelets fully restored ischemia-induced angiogenesis. These findings suggest that neuronally derived NPY accelerates the early response to femoral artery ligation by promoting collateral conductance, while platelet-derived NPY is critical for sustained capillary angiogenesis.


Asunto(s)
Plaquetas/metabolismo , Isquemia/sangre , Neovascularización Fisiológica , Neuropéptido Y/fisiología , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Miembro Posterior , Humanos , Isquemia/genética , Isquemia/fisiopatología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Neovascularización Fisiológica/genética , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Ratas , Ratas Wistar
2.
Nat Med ; 13(7): 803-11, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17603492

RESUMEN

The relationship between stress and obesity remains elusive. In response to stress, some people lose weight, whereas others gain. Here we report that stress exaggerates diet-induced obesity through a peripheral mechanism in the abdominal white adipose tissue that is mediated by neuropeptide Y (NPY). Stressors such as exposure to cold or aggression lead to the release of NPY from sympathetic nerves, which in turn upregulates NPY and its Y2 receptors (NPY2R) in a glucocorticoid-dependent manner in the abdominal fat. This positive feedback response by NPY leads to the growth of abdominal fat. Release of NPY and activation of NPY2R stimulates fat angiogenesis, macrophage infiltration, and the proliferation and differentiation of new adipocytes, resulting in abdominal obesity and a metabolic syndrome-like condition. NPY, like stress, stimulates mouse and human fat growth, whereas pharmacological inhibition or fat-targeted knockdown of NPY2R is anti-angiogenic and anti-adipogenic, while reducing abdominal obesity and metabolic abnormalities. Thus, manipulations of NPY2R activity within fat tissue offer new ways to remodel fat and treat obesity and metabolic syndrome.


Asunto(s)
Dieta , Neuropéptido Y/genética , Obesidad/metabolismo , Estrés Fisiológico/metabolismo , Células 3T3-L1 , Tejido Adiposo Blanco/metabolismo , Animales , Frío , Grasas de la Dieta , Eliminación de Gen , Regulación de la Expresión Génica , Masculino , Síndrome Metabólico , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/genética , Regulación hacia Arriba
3.
Lancet ; 380(9841): 572-80, 2012 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-22607825

RESUMEN

BACKGROUND: High plasma HDL cholesterol is associated with reduced risk of myocardial infarction, but whether this association is causal is unclear. Exploiting the fact that genotypes are randomly assigned at meiosis, are independent of non-genetic confounding, and are unmodified by disease processes, mendelian randomisation can be used to test the hypothesis that the association of a plasma biomarker with disease is causal. METHODS: We performed two mendelian randomisation analyses. First, we used as an instrument a single nucleotide polymorphism (SNP) in the endothelial lipase gene (LIPG Asn396Ser) and tested this SNP in 20 studies (20,913 myocardial infarction cases, 95,407 controls). Second, we used as an instrument a genetic score consisting of 14 common SNPs that exclusively associate with HDL cholesterol and tested this score in up to 12,482 cases of myocardial infarction and 41,331 controls. As a positive control, we also tested a genetic score of 13 common SNPs exclusively associated with LDL cholesterol. FINDINGS: Carriers of the LIPG 396Ser allele (2·6% frequency) had higher HDL cholesterol (0·14 mmol/L higher, p=8×10(-13)) but similar levels of other lipid and non-lipid risk factors for myocardial infarction compared with non-carriers. This difference in HDL cholesterol is expected to decrease risk of myocardial infarction by 13% (odds ratio [OR] 0·87, 95% CI 0·84-0·91). However, we noted that the 396Ser allele was not associated with risk of myocardial infarction (OR 0·99, 95% CI 0·88-1·11, p=0·85). From observational epidemiology, an increase of 1 SD in HDL cholesterol was associated with reduced risk of myocardial infarction (OR 0·62, 95% CI 0·58-0·66). However, a 1 SD increase in HDL cholesterol due to genetic score was not associated with risk of myocardial infarction (OR 0·93, 95% CI 0·68-1·26, p=0·63). For LDL cholesterol, the estimate from observational epidemiology (a 1 SD increase in LDL cholesterol associated with OR 1·54, 95% CI 1·45-1·63) was concordant with that from genetic score (OR 2·13, 95% CI 1·69-2·69, p=2×10(-10)). INTERPRETATION: Some genetic mechanisms that raise plasma HDL cholesterol do not seem to lower risk of myocardial infarction. These data challenge the concept that raising of plasma HDL cholesterol will uniformly translate into reductions in risk of myocardial infarction. FUNDING: US National Institutes of Health, The Wellcome Trust, European Union, British Heart Foundation, and the German Federal Ministry of Education and Research.


Asunto(s)
HDL-Colesterol/sangre , Análisis de la Aleatorización Mendeliana/métodos , Infarto del Miocardio/sangre , Biomarcadores/sangre , Estudios de Casos y Controles , LDL-Colesterol/sangre , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Lipasa/genética , Infarto del Miocardio/epidemiología , Infarto del Miocardio/etiología , Infarto del Miocardio/genética , Polimorfismo de Nucleótido Simple , Estudios Prospectivos , Factores de Riesgo
4.
Lancet ; 377(9763): 383-92, 2011 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-21239051

RESUMEN

BACKGROUND: We tested whether genetic factors distinctly contribute to either development of coronary atherosclerosis or, specifically, to myocardial infarction in existing coronary atherosclerosis. METHODS: We did two genome-wide association studies (GWAS) with coronary angiographic phenotyping in participants of European ancestry. To identify loci that predispose to angiographic coronary artery disease (CAD), we compared individuals who had this disorder (n=12,393) with those who did not (controls, n=7383). To identify loci that predispose to myocardial infarction, we compared patients who had angiographic CAD and myocardial infarction (n=5783) with those who had angiographic CAD but no myocardial infarction (n=3644). FINDINGS: In the comparison of patients with angiographic CAD versus controls, we identified a novel locus, ADAMTS7 (p=4·98×10(-13)). In the comparison of patients with angiographic CAD who had myocardial infarction versus those with angiographic CAD but no myocardial infarction, we identified a novel association at the ABO locus (p=7·62×10(-9)). The ABO association was attributable to the glycotransferase-deficient enzyme that encodes the ABO blood group O phenotype previously proposed to protect against myocardial infarction. INTERPRETATION: Our findings indicate that specific genetic predispositions promote the development of coronary atherosclerosis whereas others lead to myocardial infarction in the presence of coronary atherosclerosis. The relation to specific CAD phenotypes might modify how novel loci are applied in personalised risk assessment and used in the development of novel therapies for CAD. FUNDING: The PennCath and MedStar studies were supported by the Cardiovascular Institute of the University of Pennsylvania, by the MedStar Health Research Institute at Washington Hospital Center and by a research grant from GlaxoSmithKline. The funding and support for the other cohorts contributing to the paper are described in the webappendix.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/genética , Proteínas ADAM/genética , Enfermedad de la Arteria Coronaria/genética , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Infarto del Miocardio/genética , Polimorfismo de Nucleótido Simple , Proteína ADAMTS7 , Adulto , Anciano , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Infarto del Miocardio/sangre , Infarto del Miocardio/complicaciones , Infarto del Miocardio/diagnóstico por imagen
5.
Arterioscler Thromb Vasc Biol ; 31(8): 1748-56, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21617137

RESUMEN

OBJECTIVE: Aging is a major risk factor for increased ischemic tissue injury. Whether collateral rarefaction and impaired remodeling contribute to this is unknown. We quantified the number and diameter of native collaterals and their remodeling in 3-, 16-, 24-, and 31-month-old mice. METHODS AND RESULTS: Aging caused an "age-dose-dependent" greater drop in perfusion immediately after femoral artery ligation, followed by a diminished recovery of flow and increase in tissue injury. These effects were associated with a decline in collateral number, diameter, and remodeling. Angiogenesis was also impaired. Mechanistically, these changes were not accompanied by reduced recruitment of T cells or macrophages to remodeling collaterals. However, endothelial nitric oxide synthase signaling was dysfunctional, as indicated by increased protein nitrosylation and less phosphorylated endothelial nitric oxide synthase and vasodilator-stimulated phosphoprotein in collateral wall cells. The cerebral circulation exhibited a similar age-dose-dependent loss of collateral number and diameter and increased tortuosity, resulting in an increase in collateral resistance and infarct volume (eg, 6- and 3-fold, respectively, in 24-month-old mice) after artery occlusion. This was not associated with rarefaction of similarly sized arterioles. Collateral remodeling was also reduced. CONCLUSIONS: Our findings demonstrate that aging causes rarefaction and insufficiency of the collateral circulation in multiple tissues, resulting in more severe ischemic tissue injury.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Circulación Colateral , Isquemia/patología , Isquemia/fisiopatología , Animales , Encéfalo/irrigación sanguínea , Infarto Encefálico/patología , Infarto Encefálico/fisiopatología , Modelos Animales de Enfermedad , Arteria Femoral/lesiones , Miembro Posterior/irrigación sanguínea , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Transducción de Señal , Resistencia Vascular
6.
PLoS Genet ; 5(12): e1000768, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20011104

RESUMEN

The adipocyte-derived protein adiponectin is highly heritable and inversely associated with risk of type 2 diabetes mellitus (T2D) and coronary heart disease (CHD). We meta-analyzed 3 genome-wide association studies for circulating adiponectin levels (n = 8,531) and sought validation of the lead single nucleotide polymorphisms (SNPs) in 5 additional cohorts (n = 6,202). Five SNPs were genome-wide significant in their relationship with adiponectin (P< or =5x10(-8)). We then tested whether these 5 SNPs were associated with risk of T2D and CHD using a Bonferroni-corrected threshold of P< or =0.011 to declare statistical significance for these disease associations. SNPs at the adiponectin-encoding ADIPOQ locus demonstrated the strongest associations with adiponectin levels (P-combined = 9.2x10(-19) for lead SNP, rs266717, n = 14,733). A novel variant in the ARL15 (ADP-ribosylation factor-like 15) gene was associated with lower circulating levels of adiponectin (rs4311394-G, P-combined = 2.9x10(-8), n = 14,733). This same risk allele at ARL15 was also associated with a higher risk of CHD (odds ratio [OR] = 1.12, P = 8.5x10(-6), n = 22,421) more nominally, an increased risk of T2D (OR = 1.11, P = 3.2x10(-3), n = 10,128), and several metabolic traits. Expression studies in humans indicated that ARL15 is well-expressed in skeletal muscle. These findings identify a novel protein, ARL15, which influences circulating adiponectin levels and may impact upon CHD risk.


Asunto(s)
Adiponectina/sangre , Estudio de Asociación del Genoma Completo , Adiponectina/genética , Adiponectina/fisiología , Enfermedad Coronaria/genética , Diabetes Mellitus Tipo 2/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Polimorfismo de Nucleótido Simple
7.
Am J Physiol Heart Circ Physiol ; 300(6): H2027-34, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21398592

RESUMEN

Blood flow restoration to ischemic tissue is affected by various risk factors. The aim of this study was to examine gender effects on arteriogenesis and angiogenesis in a mouse ischemic hindlimb model. C57BL/6J mice were subjected to unilateral hindlimb ischemia. Flow recovery was less and hindlimb use impairment was greater in females. No gender difference in vessel number was found at baseline, although 7 days postsurgery females had fewer α-smooth muscle actin-positive vessels in the midpoint of the adductor region. Females had higher hindlimb vascular resistance, were less responsive to vasodilators, and were more sensitive to vasoconstrictors postligation. Western blotting showed that females had higher baseline levels of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS) in the calf, while 7 days postligation males had higher levels of VEGF, eNOS, and phosphorylated vasodilator stimulated phosphoprotein. Females had less angiogenesis in a Matrigel plug assay and less endothelial cell proliferation in vitro. Females have impaired recovery of flow, a finding presumably caused by multiple factors including decreased collateral remodeling, less angiogenesis, impaired vasodilator response, and increased vasoconstrictor activity; our results also suggest the possibility that new collateral formation, from capillaries, is impaired in females.


Asunto(s)
Arteria Femoral/fisiología , Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Flujo Sanguíneo Regional/fisiología , Caracteres Sexuales , Animales , Femenino , Miembro Posterior/metabolismo , Isquemia/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Resistencia Vascular/fisiología
8.
Arterioscler Thromb Vasc Biol ; 30(3): 477-82, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20056912

RESUMEN

OBJECTIVE: In a previous study we identified metallothionein (MT) as a candidate gene potentially influencing collaterogenesis. In this investigation, we determined the effect of MT on collaterogenesis and examined the mechanisms contributing to the effects we found. METHODS AND RESULTS: Collateral blood flow recovery was assessed using laser Doppler perfusion imaging, and angiogenesis was measured using a Matrigel plug assay. Smooth muscle cells were isolated from MT knockout (KO) mice for functional assays. Gene expression of matrix metalloproteinase-9, platelet-derived growth factor, vascular endothelial growth factor, and Fat cadherin in smooth muscle cells was measured by real-time polymerase chain reaction, and protein levels of vascular endothelial growth factor and matrix metalloproteinase-9 were determined using enzyme-linked immunosorbent assay and Western blot. CD11b(+) macrophages were tested for invasiveness using a real-time impedance assay. Both flow recovery and angiogenesis were impaired in MT KO mice. Proliferation, migration, and invasion were decreased in MT KO smooth muscle cells, and matrix metalloproteinase-9, platelet-derived growth factor, and vascular endothelial growth factor expression were also decreased, whereas FAT-1 cadherin expression was elevated. MT KO CD11b(+) cells were more invasive than wild-type cells. CONCLUSIONS: MT plays an important role in collateral flow recovery and angiogenesis, an activity that appears to be mediated, in part, by the effects of MT on the functionality of 3 cell types essential for these processes: endothelial cells, smooth muscle cells, and macrophages.


Asunto(s)
Arterias/crecimiento & desarrollo , Macrófagos/fisiología , Metalotioneína/fisiología , Músculo Liso Vascular/fisiología , Neovascularización Fisiológica/fisiología , Animales , Arterias/citología , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Miembro Posterior/irrigación sanguínea , Macrófagos/citología , Masculino , Metalotioneína/genética , Ratones , Ratones Noqueados , Modelos Animales , Músculo Liso Vascular/citología , Flujo Sanguíneo Regional/fisiología
9.
Cardiovasc Res ; 78(2): 257-64, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18178573

RESUMEN

Animal and early clinical studies of gene therapy for tissue ischaemia suggested that this approach might provide benefit to patients with coronary artery disease not amenable to traditional revascularization. This enthusiasm was then tempered by the subsequent disappointing results of randomized clinical trials and led researchers to develop strategies using progenitor cells as an alternative to improve collateral function. However, the recent publication of several randomized clinical trials reporting either negative or weakly positive results using this approach have led to questions regarding its effectiveness. There are several factors that need to be considered in explaining the discordance between the positive studies of such treatments in animals and the disappointing results seen in randomized patient trials. Aside from the practical issues of arteriogenic therapies, such as effective delivery, vascular remodelling is an extraordinarily complex process, and the administration of a single agent or cell in the hope that it would lead to lasting physiological effects may be far too simplistic an approach. In addition, however, evidence now suggests that many of the traditional cardiovascular risk factors-such as age and hypercholesterolemia-may impair the host response not only to ischaemia but, critically, also to treatment as well. This review discusses the evidence and mechanisms for these observations and highlights future directions that might be taken in an effort to provide more effective therapies.


Asunto(s)
Circulación Colateral , Vasos Coronarios/fisiopatología , Isquemia Miocárdica/fisiopatología , Revascularización Miocárdica , Neovascularización Fisiológica , Animales , Enfermedad de la Arteria Coronaria/fisiopatología , Enfermedad de la Arteria Coronaria/terapia , Terapia Genética , Humanos , Isquemia Miocárdica/etiología , Isquemia Miocárdica/terapia , Revascularización Miocárdica/métodos , Selección de Paciente , Factores de Riesgo , Trasplante de Células Madre , Insuficiencia del Tratamiento
11.
Circulation ; 113(1): 118-24, 2006 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-16380545

RESUMEN

BACKGROUND: Previous studies have demonstrated that macrophages and CD4+ T lymphocytes play pivotal roles in collateral development. Indirect evidence suggests that CD8+ T cells also play a role. Thus, after acute cerebral ischemia, CD8+ T cells infiltrate the perivascular space and secrete interleukin-16 (IL-16), a potent chemoattractant for monocytes and CD4+ T cells. We tested whether CD8+ T lymphocytes contribute to collateral vessel development and whether the lack of circulating CD8+ T cells prevents IL-16 expression, impairs CD4+ mononuclear cell recruitment, and reduces collateral vessel growth after femoral artery ligation in CD8(-/-) mice. METHODS AND RESULTS: After surgical excision of the femoral artery, laser Doppler perfusion imaging demonstrated reduced blood flow recovery in CD8(-/-) mice compared with C57/BL6 mice (ischemic/nonischemic limb at day 28, 0.66+/-0.04 versus 0.87+/-0.04, respectively; P<0.01). This resulted in greater calf muscle atrophy (mean fiber area, 785+/-68 versus 1067+/-69 microm2, respectively; P<0.01) and increased fibrotic tissue content (10.8+/-1.2% versus 7+/-1%, respectively; P<0.01). Moreover, CD8(-/-) mice displayed reduced IL-16 expression and decreased CD4+ T-cell recruitment at the site of collateral vessel development. Exogenous CD8+ T cells, infused into CD8(-/-) mice immediately after femoral artery ligation, selectively homed to the ischemic hind limb and expressed IL-16. The restoration of IL-16 expression resulted in significant CD4+ mononuclear cell infiltration of the ischemic limb, faster blood flow recovery, and reduced hindlimb muscle atrophy/fibrosis. When exogenous CD8+ T cells deficient in IL-16 (IL-16(-/-)) were infused into CD8(-/-) mice immediately after femoral artery ligation, they selectively homed to the ischemic hind limb but were unable to recruit CD4+ mononuclear cells and did not improve blood flow recovery. CONCLUSIONS: These results demonstrate that CD8+ T cells importantly contribute to the early phase of collateral development. After femoral artery ligation, CD8+ T cells infiltrate the site of collateral vessel growth and recruit CD4+ mononuclear cells through the expression of IL-16. Our study provides further evidence of the significant role of the immune system in modulating collateral development in response to peripheral ischemia.


Asunto(s)
Arterioloesclerosis/etiología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/fisiología , Quimiotaxis de Leucocito/fisiología , Circulación Colateral/inmunología , Interleucina-16/fisiología , Isquemia/complicaciones , Animales , Arterioloesclerosis/inmunología , Antígenos CD8/genética , Modelos Animales de Enfermedad , Arteria Femoral , Sistema Inmunológico/fisiología , Interleucina-16/biosíntesis , Ratones , Ratones Noqueados
12.
J Clin Endocrinol Metab ; 91(1): 64-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16249281

RESUMEN

CONTEXT: Recently, resistin was found to be present in atherosclerotic lesions in apoE(-/-) mice. Resistin may be associated with inflammation and atherosclerosis in humans; however, the role of resistin in human disease remains controversial. OBJECTIVE: This study assesses cross-sectional relationships of resistin with coronary heart disease (CHD). DESIGN, SETTING, AND PARTICIPANTS: Blood samples from the third examination of the Strong Heart Study (SHS)--the largest study of CHD in American Indians--were used. Cases who had suffered previous myocardial infarction (n = 100) were selected randomly from the three SHS sites and matched for study site and sex with controls who had no history of cardiovascular disease (CHD or stroke) (n = 100). MAIN OUTCOME MEASURE: Resistin levels by enzyme-linked immunosorbent assay method in cases and controls was the main outcome measure. RESULTS: Resistin levels were higher in cases than controls [median (interquartile range): 3.4 (2.5-4.7) vs. 2.8 (2.1-4.0) ng/ml; P = 0.003] and had univariate correlations with age (Spearman r = 0.21; P < 0.002), fasting insulin (r = 0.21; P = 0.003), insulin resistance by homeostasis model (r = 0.22; P = 0.04), albumin to creatinine ratio (r = 0.19; P = 0.01), and fibrinogen (r = 0.34; P < 0.0001). Cases were more likely to have diabetes (cases 67%; controls 41%; P < 0.0001) but had similar body mass index (cases 31.4 +/- 5.4; controls 30.7 +/- 6.3; P = 0.85). Resistin levels were higher in participants with established nephropathy (albumin to creatinine ratio >300 mg/g, n = 26) compared with those with normo- (n = 122) or microalbuminuria (n = 42). In multivariate analysis, nephropathy (P = 0.0013) but not previous myocardial infarction (P = 0.12) was significantly associated with resistin. CONCLUSIONS: Resistin is not independently associated with CHD. Resistin is elevated in survivors of myocardial infarction; however, this reflects a novel association of raised resistin with diabetic nephropathy.


Asunto(s)
Enfermedad Coronaria/sangre , Enfermedad Coronaria/diagnóstico , Resistencia a la Insulina , Resistina/sangre , Anciano , Índice de Masa Corporal , Estudios de Casos y Controles , Enfermedad Coronaria/epidemiología , Estudios Transversales , Complicaciones de la Diabetes/epidemiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Hipertensión/complicaciones , Indígenas Norteamericanos/estadística & datos numéricos , Insulina/sangre , Masculino , Persona de Mediana Edad , Infarto del Miocardio/epidemiología , Estados Unidos/epidemiología
13.
Circ Res ; 95(4): 354-63, 2004 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-15321945

RESUMEN

Initial animal studies of single angiogenic agents, such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), generated enthusiasm for the concept that these agents might enhance collateral development and thereby provide alternative therapies for patients with vascular disease not amenable to traditional revascularization. The enthusiasm, apparently justified by the subsequent results of small nonrandomized phase-I clinical trials, was then tempered by the subsequent disappointing results of randomized clinical trials. In light of these disappointing results, investigators have pursued alternative strategies in an attempt to improve tissue perfusion. One such strategy is the utilization of bone marrow-derived cell therapy. This review discusses mechanistic pathways mediating the effects of such cell therapy, summarizes the animal and early clinical experience, and speculates on the potential of genetic manipulation of bone marrow-derived cells in an attempt to further enhance their potency.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas , Neovascularización Fisiológica , Factores de Edad , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Ensayos Clínicos Fase I como Asunto , Circulación Colateral/fisiología , Endotelio Vascular/citología , Predicción , Sustancias de Crecimiento/fisiología , Humanos , Isquemia/fisiopatología , Ratones , Músculo Liso Vascular/citología , Infarto del Miocardio/terapia , Isquemia Miocárdica/terapia , Óxido Nítrico/fisiología , Comunicación Paracrina , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Células del Estroma/citología , Células del Estroma/trasplante
14.
Circulation ; 109(7): 893-7, 2004 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-14757699

RESUMEN

BACKGROUND: The possible etiologic role of infection in cardiovascular disease is still debated. Having previously demonstrated that murine cytomegalovirus (MCMV) infection of apolipoprotein (apo) E-/- mice increases atherosclerotic lesion size, we determined if MCMV infection produces proatherogenic changes in aortic gene expression. Additionally, in cholesterol-fed C57BL/6J mice, we examined the effects of MCMV infection on aortic lesion area. METHODS AND RESULTS: C57BL/6J apoE-/- and wild-type C57BL/6J mice were infected with MCMV. At various time points, aortas were collected and pooled. Total RNA was extracted and hybridized to Affymetrix murine chips or analyzed for specific gene expression using TaqMan reverse transcription-polymerase chain reaction. Data from infected and uninfected mice were compared. A separate group of cholesterol-fed C57BL/6J mice were infected with MCMV, and lesion area in the aortic sinus was assessed using oil red O staining. Acute MCMV infection altered aortic expression of atherogenic genes in young apoE-/- and C57BL/6J mice-specifically, monocyte chemoattractant protein-1, monokine induced by interferon-gamma, and interferon-gamma inducible protein 10. Acute infection in adult 9-month-old apoE-/- mice with well-established lesions increased aortic expression of monocyte chemoattractant protein-1. Atherosclerotic lesion area in cholesterol-fed C57BL/6J mice was increased after infection with MCMV. CONCLUSIONS: MCMV infection significantly increases atherosclerotic lesion area and aortic expression of atherogenic genes. These infection-induced effects indicate mechanisms by which cytomegalovirus may contribute to atherosclerotic disease initiation and progression and to the precipitation of clinical events. These results additionally add to data compatible with the concept that infection does play an important role in atherosclerotic disease.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/etiología , Arteriosclerosis/etiología , Quimiocina CCL2/biosíntesis , Quimiocinas CXC/biosíntesis , Infecciones por Citomegalovirus/genética , Regulación de la Expresión Génica , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Arteriosclerosis/genética , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Quimiocina CCL2/genética , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/genética , Colesterol en la Dieta/farmacología , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/metabolismo , Dieta Aterogénica , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Bazo/patología , Linfocitos T/metabolismo
15.
Circulation ; 108(2): 205-10, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12821542

RESUMEN

BACKGROUND: T lymphocytes, components of the immune and inflammatory systems, are involved in such normal processes as wound healing and host defense against infection and in such pathological processes as tumor growth and atherosclerotic plaque development. Angiogenesis is a mechanism common to each. Because CD4+ T lymphocytes are active in regulating humoral and cellular responses of the immune system, we determined whether CD4+ cells contribute to collateral vessel development by using the mouse ischemic hindlimb model. METHODS AND RESULTS: One week after ischemia, CD4-/- mice showed reduced collateral flow induction, macrophage number, and vascular endothelial growth factor levels in the ischemic muscle compared with wild-type mice. There was also delayed recovery of hindlimb function and increased muscle atrophy/fibrosis. Spleen-derived purified CD4+ T cells infused into CD4-/- mice selectively localized to the ischemic limb and significantly increased collateral flow as well as macrophage number and vascular endothelial growth factor levels in the ischemic muscle. Muscle function and damage also improved. CONCLUSIONS: These results indicate an important role of CD4+ cells in collateral development, as demonstrated by a 25% decrease in blood flow recovery after femoral artery ligation. Our data also suggest that CD4+ T cells control the arteriogenic response to acute hindlimb ischemia, at least in part, by recruiting macrophages to the site of active collateral artery formation, which in turn triggers the development of collaterals through the synthesis of arteriogenic cytokines.


Asunto(s)
Arterias/fisiopatología , Antígenos CD4/genética , Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Neovascularización Fisiológica , Enfermedad Aguda , Animales , Arterias/patología , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD4-Positivos/trasplante , Células Cultivadas , Circulación Colateral/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Arteria Femoral/fisiopatología , Citometría de Flujo , Miembro Posterior/patología , Miembro Posterior/fisiopatología , Inflamación/patología , Isquemia/patología , Isquemia/terapia , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Recuperación de la Función , Flujo Sanguíneo Regional
16.
J Am Coll Cardiol ; 43(3): 474-82, 2004 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-15013133

RESUMEN

OBJECTIVES: We sought to understand the genomic program leading to collateral vessel formation. BACKGROUND: Recently, technology has advanced to the point that it is now possible to elucidate the large array of genes that must be expressed, as well as the temporal expression pattern, for the development of functionally important collateral vessels. In this investigation, we used deoxyribonucleic acid array expression profiling to determine the time course of differential expression of 12,000 genes after femoral artery ligation in C57BL/6 mice. METHODS: Ribonucleic acid was extracted from the adductor muscle, which showed no signs of ischemia. Sampling was at baseline, 6 h, and 1, 3, 7, and 14 days after femoral artery ligation or sham operation. RESULTS: Femoral artery ligation caused the differential expression (>2-fold) of 783 genes at one or multiple time points: 518 were induced and 265 were repressed. Cluster analysis generated four temporal patterns: 1) early upregulated (6 to 24 h)-immediate early transcriptional factors, angiogenesis, inflammation, and stress-related genes; 2) mid-phase upregulated (day 3)-cell cycle and cytoskeletal and inflammatory genes; 3) late upregulated (days 7 to 14)-angiostatic, anti-inflammatory, and extracellular matrix-associated genes; and 4) downregulated-genes involved in energy metabolism, water channel, and muscle contraction. Microarray data were validated using quantitative reverse transcription polymerase chain reaction. CONCLUSIONS: This study documents the large number of genes whose differential expression and temporal functional clustering appear to contribute to collateral formation. These results can serve as a genomic model for arteriogenesis and as a database for developing new therapeutic strategies.


Asunto(s)
Circulación Colateral/genética , Expresión Génica/genética , Miembro Posterior/irrigación sanguínea , Isquemia/genética , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/genética , Animales , Arteria Femoral/cirugía , Perfilación de la Expresión Génica/métodos , Isquemia/fisiopatología , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Músculo Esquelético/fisiopatología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Factores de Tiempo
17.
J Am Coll Cardiol ; 41(10): 1812-9, 2003 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-12767669

RESUMEN

OBJECTIVES: We examined whether selective cyclooxygenase-2 (COX-2) inhibition in apolipoprotein-E (apoE) deficient mice reduces cytomegalovirus (CMV) replication, and determined whether COX-2 anti-inflammatory activity leads to decreased atherosclerosis. BACKGROUND: Evidence suggests that CMV infection contributes to atherosclerosis and that this occurs in part through inflammatory mechanisms. Cyclooxygenase-2 inhibitors are potent anti-inflammatory agents. They also inhibit CMV replication in vitro. METHODS: The apoE deficient mice were either treated or not treated with a selective COX-2 inhibitor, and either infected or not infected with CMV. Viral deoxyribonucleic acid load in salivary glands was determined by quantitative polymerase chain reaction. Atherosclerotic lesion analysis was performed by standard methods. RESULTS: In vivo COX-2 inhibition, unexpectedly increased viral load: in the CMV-infected animals viral load was 2.58 +/- 1.0 in the nontreated group, 4.74 +/- 1.38 in the group treated with 12 mg/kg/day MF-tricyclic, and 6.51 +/- 1.64 in the group treated with 24 mg/kg/day MF-tricyclic (p trend = 0.050). This increased viral load was paralleled by increased anti-CMV antibody titers. Most surprisingly, COX-2 inhibition significantly increased early atherosclerotic lesion area, independent of viral infection. CONCLUSIONS: Our study demonstrates that selective inhibition of COX-2 in vivo increases viral load. The finding that inhibition of COX-2 increases atherosclerosis development in apoE deficient mice suggests, unexpectedly, that this enzyme exerts antiatherosclerosis activity, at least in this model.


Asunto(s)
Apolipoproteínas E/deficiencia , Arteriosclerosis/patología , Inhibidores de la Ciclooxigenasa/farmacología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/fisiología , Furanos/farmacología , Isoenzimas/fisiología , Prostaglandina-Endoperóxido Sintasas/fisiología , Replicación Viral , Animales , Anticuerpos Antivirales/análisis , Arteriosclerosis/fisiopatología , Arteriosclerosis/virología , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Citocinas/sangre , Citomegalovirus/inmunología , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/virología , ADN Viral/análisis , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Relación Dosis-Respuesta Inmunológica , Inmunoglobulina G/análisis , Isoenzimas/antagonistas & inhibidores , Ratones , Ratones Noqueados , Carga Viral
18.
Atherosclerosis ; 182(2): 241-8, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16159596

RESUMEN

Resistin, an adipocyte-derived cytokine linked to insulin resistance and obesity, has recently been shown to activate endothelial cells (ECs). Using microarrays, we found that along with numerous other pro-atherosclerotic genes, resistin expression levels are elevated in the aortas of C57BL/6J apoE-/- mice; these findings led us to further explore the relation between resistin and atherosclerosis. Using TaqMan PCR and immunohistochemistry, we found that ApoE-/- mice had significantly higher resistin mRNA and protein levels in their aortas, and elevated serum resistin levels, compared to C57BL/6J wild-type mice. Incubation of murine aortic ECs with recombinant resistin increased monocyte chemoattractant protein (MCP)-1 and soluble vascular cell adhesion molecule (sVCAM)-1 protein levels in the conditioned medium. Furthermore, human carotid endarterectomy samples stained positive for resistin protein, while internal mammary artery did not show strong staining. Patients diagnosed with premature coronary artery disease (PCAD) were found to have higher serum levels of resistin than normal controls. In summary, resistin protein is present in both murine and human atherosclerotic lesions, and mRNA levels progressively increase in the aortas of mice developing atherosclerosis. Resistin induces increases in MCP-1 and sVCAM-1 expression in murine vascular endothelial cells, suggesting a possible mechanism by which resistin might contribute to atherogenesis. Finally, PCAD patients exhibited increased serum levels of resistin when compared to controls. These findings suggest a possible role of resistin in cardiovascular disease.


Asunto(s)
Enfermedades de las Arterias Carótidas/fisiopatología , Enfermedad de la Arteria Coronaria/fisiopatología , Resistina/sangre , Resistina/genética , Adulto , Animales , Aorta/citología , Aorta/metabolismo , Apolipoproteínas E/genética , Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Células Cultivadas , Enfermedad de la Arteria Coronaria/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Arterias Mamarias/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
19.
Atherosclerosis ; 170(2): 223-8, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14612201

RESUMEN

Atherosclerosis is an inflammatory disease. One of the candidate inflammatory triggers is infection. To further characterize the interaction between infection, cytokine induction, and atherosclerosis, we tested the hypothesis that cytomegalovirus (CMV) infection induces the pro-inflammatory cytokine interleukin-6 (IL-6), which in turn induces "pro-atherosclerotic" changes in vascular endothelial cells (ECs). ELISA was used to determine the levels of monocyte chemoattractant protein-1 (MCP-1) in the supernatant of mouse and human ECs incubated with IL-6, and IL-6 levels in supernatants of splenocytes, derived from CMV-infected and uninfected mice, stimulated with mice CMV antigens. IL-6 induced, in a dose response fashion, MCP-1 expression in human ECs: 0, 2, 10, and 50 pg/ml IL-6 increased MCP-1 levels in EC conditioned medium from 1120+/-65 to 1148+/-105, 1395+/-40, and 2119+/-130 pg/ml, respectively (P<0.001). IL-6 also induced MCP-1 expression in mouse ECs (P<0.002). Importantly, IL-6 concentration in the supernatants of splenocytes stimulated with CMV antigens rose from undetectable levels in uninfected mice to 14.9+/-5 pg/ml in the infected mice (P<0.04). These results suggest a previously unrecognized, but potentially important mechanism whereby CMV, and other pathogens, contribute to atherogenesis: T lymphocytes, clonally expanded in response to antigens presented by CMV infection, home to sites of vascular injury and locally release IL-6 when presented with either pathogen antigens that may be present in the plaque, or when they cross-react with host peptides homologous to the relevant pathogen antigens; IL-6 then triggers ECs to release MCP-1, which recruits more monocytes and T-cells into the vessel wall and thereby exacerbates local inflammation, and thus atherogenesis.


Asunto(s)
Antígenos Virales/inmunología , Arteriosclerosis/virología , Quimiocina CCL2/biosíntesis , Infecciones por Citomegalovirus/metabolismo , Endotelio Vascular/metabolismo , Interleucina-6/biosíntesis , Bazo/metabolismo , Animales , Anticuerpos Antivirales/análisis , Células Cultivadas , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/inmunología , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Humanos , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/citología , Bazo/inmunología
20.
J Cardiovasc Transl Res ; 7(8): 749-55, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25315467

RESUMEN

Chronic stress is associated with increased risk of cardiovascular diseases. Aging is also associated with vascular dysfunction. We hypothesize that chronic stress accelerates collateral dysfunction in old mice. Mice were subjected to either chronic social defeat (CSD) or chronic cold stress (CCS). The CSD mice were housed in a box inside an aggressor's cage and exposed to the aggressor. The CCS group was placed in iced water. After chronic stress, mice underwent femoral artery ligation (FAL) and flow recovery was measured. For the CSD group, appearance and use scores of the foot and a behavioral test were performed. CSD impaired collateral flow recovery after FAL. Further, stressed mice had greater ischemic damage, impaired foot function, and altered behavior. The CCS mice also showed impaired collateral flow recovery. Chronic stress causes hind limb collateral dysfunction in old mice, a conclusion reinforced by the fact that two types of stress produced similar changes.


Asunto(s)
Circulación Colateral , Arteria Femoral/fisiopatología , Isquemia/fisiopatología , Estrés Psicológico/fisiopatología , Animales , Conducta Animal , Enfermedad Crónica , Frío , Modelos Animales de Enfermedad , Arteria Femoral/cirugía , Isquemia/etiología , Isquemia/psicología , Ligadura , Masculino , Ratones Endogámicos C57BL , Recuperación de la Función , Flujo Sanguíneo Regional , Conducta Social , Estrés Psicológico/etiología , Estrés Psicológico/psicología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA