Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 40(9): 2159-2170, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32673527

RESUMEN

OBJECTIVE: Evidence from preclinical and clinical studies has demonstrated that myocardial infarction promotes atherosclerosis progression. The impact of focal vascular inflammation on the progression and phenotype of remote atherosclerosis remains unknown. Approach and Results: We used a novel ApoE-/- knockout mouse model of sustained arterial inflammation, initiated by mechanical injury in the abdominal aorta. Using serial in vivo molecular MRI and ex vivo histology and flow cytometry, we demonstrate that focal arterial inflammation triggered by aortic injury, accelerates atherosclerosis in the remote brachiocephalic artery. The brachiocephalic artery atheroma had distinct histological features including increased plaque size, plaque permeability, necrotic core to collagen ratio, infiltration of more inflammatory monocyte subsets, and reduced collagen content. We also found that arterial inflammation following focal vascular injury evoked a prolonged systemic inflammatory response manifested as a persistent increase in serum IL-6 (interleukin 6). Finally, we demonstrate that 2 therapeutic interventions-pravastatin and minocycline-had distinct anti-inflammatory effects at the plaque and systemic level. CONCLUSIONS: We show for the first time that focal arterial inflammation in response to vascular injury enhances systemic vascular inflammation, accelerates remote atheroma progression and induces plaques more inflamed, lipid-rich, and collagen-poor in the absence of ischemic myocardial injury. This inflammatory cascade is modulated by pravastatin and minocycline treatments, which have anti-inflammatory effects at both plaque and systemic levels that mitigate atheroma progression.


Asunto(s)
Aortitis/complicaciones , Aterosclerosis/etiología , Tronco Braquiocefálico/metabolismo , Mediadores de Inflamación/sangre , Placa Aterosclerótica , Animales , Antiinflamatorios/farmacología , Aortitis/sangre , Aortitis/patología , Aterosclerosis/sangre , Aterosclerosis/patología , Aterosclerosis/prevención & control , Tronco Braquiocefálico/efectos de los fármacos , Tronco Braquiocefálico/patología , Colágeno/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Interleucina-6/sangre , Metabolismo de los Lípidos , Masculino , Ratones Noqueados para ApoE , Minociclina/farmacología , Necrosis , Pravastatina/farmacología , Factores de Tiempo
2.
Atherosclerosis ; 277: 28-33, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30170221

RESUMEN

BACKGROUND AND AIMS: Diets enriched with tree nuts have been demonstrated to reduce the risk of atherosclerosis-related cardiovascular events. Abdominal aortic aneurysm (AAA) shares common risk factors with atherosclerosis and AAA patients commonly have atherosclerosis related cardiovascular events. AAA has some distinct pathological and clinical characteristics to those of atherosclerosis. No previous study has examined the effect of a diet enriched with tree nuts on experimental or clinical AAA. This study investigated the effect of a diet enriched with tree nuts on the development and severity of AAA within an experimental rodent model. METHODS: Male apolipoprotein E deficient mice were allocated to a diet enriched with tree nuts or control diet for 56 days (n = 17 per group). After 28 days, all mice were infused with angiotensin II whilst being maintained on their respective diets. The primary outcome was AAA severity assessed by the supra-renal aortic diameter, measured by ultrasound and ex vivo morphometric analysis. The severity of atherosclerosis was assessed by computer-aided analysis of Sudan IV stained aortic arches and sections of brachiocephalic arteries prepared with Van Gieson's stain. RESULTS: The diet enriched with tree nuts did not influence aortic diameter or aortic rupture incidence. Mice receiving the diet enriched with tree nuts had significantly less atherosclerosis within the brachiocephalic artery (p = 0.033) but not in the aortic arch. CONCLUSIONS: This experimental study suggests that a diet enriched with tree nuts does not reduce the severity of AAA, but does reduce the severity of atherosclerosis within the brachiocephalic artery. The study was not powered to identify a moderate effect of the diet on the primary outcome and therefore this cannot be excluded.


Asunto(s)
Angiotensina II , Alimentación Animal , Aneurisma de la Aorta Abdominal/prevención & control , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Ácidos Grasos Omega-3/administración & dosificación , Nueces , Polifenoles/administración & dosificación , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Dilatación Patológica , Modelos Animales de Enfermedad , Masculino , Ratones Noqueados para ApoE , Valor Nutritivo , Placa Aterosclerótica , Índice de Severidad de la Enfermedad , Factores de Tiempo
3.
Arterioscler Thromb Vasc Biol ; 38(3): 500-508, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29326316

RESUMEN

OBJECTIVE: Fibronectin containing extra domain A (Fn-EDA) is an endogenous ligand of TLR4 (toll-like receptor 4) and is abundant in the extracellular matrix of advanced atherosclerotic lesions in human and mice. Irrespective of sex, deletion of Fn-EDA reduces early atherosclerosis in apolipoprotein E-deficient (Apoe-/-) mice. However, the contribution of Fn-EDA in advanced atherosclerosis remains poorly characterized. We determined the contribution of Fn-EDA in advanced atherosclerotic lesions of aged (1-year-old) Apoe-/- mice. APPROACH AND RESULTS: Plaque composition was determined in the innominate artery, a plaque instability site that is known to mimic several histological features of vulnerable human plaques. Female Apoe-/-, Fn-EDA-/-Apoe-/-, TLR4-/-Apoe-/-, and Fn-EDA-/-TLR4-/-Apoe-/- mice were fed a high-fat Western diet for 44 weeks. Fn-EDA-/-Apoe-/- mice exhibited reduced plaque size characterized by smaller necrotic cores, thick fibrous caps containing abundant vascular smooth muscle cells and collagen, reduced CD68/MMP9 (matrix metalloproteinase 9)-positive content, less accumulation of MMP-cleaved extracellular matrix aggrecan, and decreased vascular smooth muscle cell and macrophage apoptosis (P<0.05 versus Apoe-/- mice). Together these findings suggest that Fn-EDA induces plaque destabilization. Deletion of TLR4 reduced histological features of plaque instability in Apoe-/- mice but did not further reduce features of plaque destabilization in Fn-EDA-/-Apoe-/- mice, suggesting that TLR4 may contribute to Fn-EDA-induced plaque destabilization. Fn-EDA potentiated TLR4-dependent MMP9 expression in bone marrow-derived macrophages, suggesting that macrophage TLR4 may contribute to Fn-EDA-mediated plaque instability. CONCLUSIONS: Fn-EDA induces histological features of plaque instability in established lesions of aged Apoe-/- mice. The abundance of Fn-EDA in advanced atherosclerotic lesions may increase the risk of plaque destabilization.


Asunto(s)
Aterosclerosis/metabolismo , Tronco Braquiocefálico/metabolismo , Fibronectinas/metabolismo , Placa Aterosclerótica , Factores de Edad , Envejecimiento , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Apoptosis , Aterosclerosis/genética , Aterosclerosis/patología , Tronco Braquiocefálico/patología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibronectinas/deficiencia , Fibronectinas/genética , Fibrosis , Macrófagos/metabolismo , Macrófagos/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Necrosis , Rotura Espontánea , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
4.
Atherosclerosis ; 241(2): 297-304, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26062989

RESUMEN

OBJECTIVE: Angiopoietin-2 (Ang-2) blocking agents are currently undergoing clinical trials for use in cancer treatment. Ang-2 has also been associated with rupture-prone atherosclerotic plaques in humans, suggesting a role for Ang-2 in plaque stability. Despite the availability of Ang-2 blocking agents, their clinical use is still lacking. Our aim was to establish if Ang-2 has a role in atheroma development and in the transition of subclinical to clinically relevant atherosclerosis. We investigated the effect of antibody-mediated Ang-2 blockage on atherogenesis after in a mouse model of atherosclerosis. METHODS: Hypercholesterolemic (low-density lipoprotein receptor(-/-) apolipoprotein B(100/100)) mice were subjected to high-cholesterol diet for eight weeks, one group with and one group without Ang-2 blocking antibody treatment during weeks 4-8.To enhance plaque development, a peri-adventitial collar was placed around the carotid arteries at the start of antibody treatment. Aortic root, carotid arteries and brachiocephalic arteries were analyzed to evaluate the effect of Ang-2 blockage on atherosclerotic plaque size and stable plaque characteristics. RESULTS: Anti-Ang-2 treatment reduced the size of fatty streaks in the brachiocephalic artery (-72%, p < 0.05). In addition, antibody-mediated Ang-2 blockage reduced plasma triglycerides (-27%, p < 0.05). In contrast, Ang-2 blockage did not have any effect on the size or composition (collagen content, macrophage percentage, adventitial microvessel density) of pre-existing plaques in the aortic root or collar-induced plaques in the carotid artery. CONCLUSIONS: Ang-2 blockage was beneficial as it decreased fatty streak formation and plasma triglyceride levels, but had no adverse effect on pre-existing atherosclerosis in hypercholesterolemic mice.


Asunto(s)
Angiopoyetina 2/antagonistas & inhibidores , Anticuerpos/farmacología , Aterosclerosis/prevención & control , Tronco Braquiocefálico/efectos de los fármacos , Placa Aterosclerótica , Angiopoyetina 2/inmunología , Angiopoyetina 2/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteína B-100/deficiencia , Apolipoproteína B-100/genética , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Tronco Braquiocefálico/inmunología , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/sangre , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/prevención & control , Colesterol en la Dieta/sangre , Modelos Animales de Enfermedad , Hipercolesterolemia/sangre , Hipercolesterolemia/complicaciones , Hipercolesterolemia/genética , Masculino , Ratones Noqueados , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factores de Tiempo , Triglicéridos/sangre
5.
Arterioscler Thromb Vasc Biol ; 35(4): 779-782, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25810297

RESUMEN

OBJECTIVE: The purpose of this study was to determine the role of the endothelial glucocorticoid receptor in the pathogenesis of atherosclerosis. APPROACH AND RESULTS: Control mice and mice lacking the endothelial glucocorticoid receptor were bred onto an Apoe knockout background and subjected to high-fat diet feeding for 12 weeks. Assessment of body weight and total cholesterol and triglycerides before and after the diet revealed no differences between the 2 groups of mice. However, mice lacking the endothelial glucocorticoid receptor developed more severe atherosclerotic lesions in the aorta, brachiocephalic artery, and aortic sinus, as well as a heightened inflammatory milieu as evidenced by increased macrophage recruitment in the lesions. CONCLUSIONS: These data suggest that the endothelial glucocorticoid receptor is important for tonic inhibition of inflammation and limitation of atherosclerosis progression in this model.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Células Endoteliales/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Peso Corporal , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Colesterol/sangre , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Genotipo , Macrófagos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores de Glucocorticoides/deficiencia , Receptores de Glucocorticoides/genética , Índice de Severidad de la Enfermedad , Factores de Tiempo , Triglicéridos/sangre
6.
BMC Med Genomics ; 7: 51, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25115202

RESUMEN

BACKGROUND: Atherosclerosis, the underlying cause of cardiovascular disease, results from both genetic and environmental factors. METHODS: In the current study we take a systems-based approach using weighted gene co-expression analysis to identify a candidate pathway of genes related to atherosclerosis. Bioinformatic analyses are performed to identify candidate genes and interactions and several novel genes are characterized using in-vitro studies. RESULTS: We identify 1 coexpression module associated with innominate artery atherosclerosis that is also enriched for inflammatory and macrophage gene signatures. Using a series of bioinformatics analysis, we further prioritize the genes in this pathway and identify Cd44 as a critical mediator of the atherosclerosis. We validate our predictions generated by the network analysis using Cd44 knockout mice. CONCLUSION: These results indicate that alterations in Cd44 expression mediate inflammation through a complex transcriptional network involving a number of previously uncharacterized genes.


Asunto(s)
Aterosclerosis/genética , Tronco Braquiocefálico/metabolismo , Biología Computacional , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad/genética , Animales , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Macrófagos/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especificidad de la Especie , Transcripción Genética
7.
Physiol Genomics ; 46(14): 523-32, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24824211

RESUMEN

Estradiol (E2) is a well-known modulator of fetal neuroendocrine activity and has been proposed as a critical endocrine signal readying the fetus for birth and postnatal life. To investigate the modulatory role of E2 on fetal stress responsiveness and the response of the fetal brain to asphyxic stress, we subjected chronically catheterized fetal sheep to a transient (10 min) brachiocephalic artery occlusion (BCO) or sham occlusion. Half of the fetuses received subcutaneous pellets that increased plasma E2 concentrations within the physiological range. Hypothalamic mRNA was analyzed using the Agilent 8x15k ovine array (019921), processed and annotated as previously reported by our laboratory. Analysis of the data by ANOVA revealed that E2 differentially regulated (DR) 561 genes, and BCO DR 894 genes compared with control and E2+BCO DR 1,153 genes compared with BCO alone (all P < 0.05). E2 upregulated epigenetic pathways and downregulated local steroid biosynthesis but did not significantly involve genes known to directly respond to the estrogen receptor. Brachiocephalic occlusion upregulated kinase pathways as well as genes associated with lymphocyte infiltration into the brain and downregulated neuropeptide synthesis. E2 upregulated immune- and apoptosis-related pathways after BCO and reduced kinase and epigenetic pathway responses to the BCO. Responses to BCO are different from responses to hypoxic hypoxia suggesting that mechanisms of responses to these two forms of brain hypoxia are distinct. We conclude that cerebral ischemia caused by BCO might stimulate lymphocyte infiltration into the brain and that this response appears to be modified by estradiol.


Asunto(s)
Tronco Braquiocefálico/efectos de los fármacos , Estradiol/farmacología , Feto/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Animales , Tronco Braquiocefálico/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Feto/metabolismo , Hipotálamo/metabolismo , Hipoxia/embriología , Hipoxia/genética , Linfocitos/efectos de los fármacos , Neuropéptidos/genética , Neuropéptidos/metabolismo , ARN Mensajero/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Ovinos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
8.
J Am Heart Assoc ; 3(2): e000492, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24755147

RESUMEN

BACKGROUND: Bisphenol A (BPA) is a base chemical used extensively in many consumer products. BPA has recently been associated with increased risk of cardiovascular disease (CVD) in multiple large-scale human population studies, but the underlying mechanisms remain elusive. We previously reported that BPA activates the pregnane X receptor (PXR), which acts as a xenobiotic sensor to regulate xenobiotic metabolism and has pro-atherogenic effects in animal models upon activation. Interestingly, BPA is a potent agonist of human PXR but does not activate mouse or rat PXR signaling, which confounds the use of rodent models to evaluate mechanisms of BPA-mediated CVD risk. This study aimed to investigate the atherogenic mechanism of BPA using a PXR-humanized mouse model. METHODS AND RESULTS: A PXR-humanized ApoE deficient (huPXR•ApoE(-/-)) mouse line was generated that respond to human PXR ligands and feeding studies were performed to determine the effects of BPA exposure on atherosclerosis development. Exposure to BPA significantly increased atherosclerotic lesion area in the aortic root and brachiocephalic artery of huPXR•ApoE(-/-) mice by 104% (P<0.001) and 120% (P<0.05), respectively. By contrast, BPA did not affect atherosclerosis development in the control littermates without human PXR. BPA exposure did not affect plasma lipid levels but increased CD36 expression and lipid accumulation in macrophages of huPXR•ApoE(-/-) mice. CONCLUSION: These findings identify a molecular mechanism that could link BPA exposure to increased risk of CVD in exposed individuals. PXR is therefore a relevant target for future risk assessment of BPA and related environmental chemicals in humans.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/inducido químicamente , Compuestos de Bencidrilo/toxicidad , Contaminantes Ambientales/toxicidad , Fenoles/toxicidad , Receptores de Esteroides/agonistas , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/efectos de los fármacos , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Antígenos CD36/metabolismo , Humanos , Lípidos/sangre , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor X de Pregnano , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Medición de Riesgo , Factores de Riesgo
9.
Arterioscler Thromb Vasc Biol ; 34(2): 262-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24334873

RESUMEN

OBJECTIVE: Inbred mouse strains have different susceptibilities to experimental atherosclerosis. The C57BL/6 strain is among the most sensitive and has, therefore, been the most widely used in atherosclerosis studies, whereas many strains are resistant. The FVB/N strain is highly resistant to atherosclerosis on the apolipoprotein E (apoE)- and low-density lipoprotein (LDL) receptor-deficient backgrounds. High-density lipoprotein and its major apoprotein, apoA-I, have been shown to be protective against atherogenesis on the C57BL/6 background. We here examine the influence of genetic background on the atheroprotective nature of apoA-I. APPROACH AND RESULTS: ApoE-deficient/apoA-I-deficient mice were generated in the C57BL/6 and FVB/N strains from apoE-deficient mice. After 6 to 10 weeks on a Western-type diet, plasma lipids and atherosclerotic lesion size were assessed. Macrophage recruitment, cholesterol regulation, and blood monocyte levels were examined as potential mechanisms driving lesion size differences. FVB/N knockout mice had higher plasma very-LDL/LDL cholesterol than their C57BL/6 counterparts. ApoA-I deficiency decreased very-LDL/LDL cholesterol in C57BL/6 mice but not in FVB/N mice. FVB/N single and double knockout mice had less lesion than C57BL/6 6 to 10 weeks on diet. ApoA-I deficiency augmented lesion development only in C57BL/6 mice. Macrophage recruitment to thioglycollate-treated peritoneum and diet-induced blood monocyte levels reflected the pattern of lesion development among the 4 genotypes. ApoA-I deficiency increased macrophage cholesterol content only in C57BL/6. FVB/N plasma was a better acceptor for macrophage cholesterol efflux than C57BL/6. CONCLUSIONS: ApoA-I is atheroprotective only in certain genetic contexts. In the C57BL/6 context, but not FVB/N, apoA-I decreases inflammatory macrophage recruitment and monocytosis, contributors to lesion formation.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Apolipoproteína A-I/metabolismo , Aterosclerosis/prevención & control , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteína A-I/deficiencia , Apolipoproteína A-I/genética , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Línea Celular , Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Especificidad de la Especie , Factores de Tiempo , Triglicéridos/sangre
10.
PLoS One ; 8(9): e74211, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040204

RESUMEN

The osteoprotegerin (OPG) and receptor activator of nuclear factor-κB ligand (RANKL) cytokine system, not only controls bone homeostasis, but has been implicated in regulating vascular calcification. TNF-related apoptosis-inducing ligand (TRAIL) is a second ligand for OPG, and although its effect in vascular calcification in vitro is controversial, its role in vivo is not yet established. This study aimed to investigate the role of TRAIL in vascular calcification in vitro using vascular smooth muscle cells (VSMCs) isolated from TRAIL(-/-) and wild-type mice, as well as in vivo, in advanced atherosclerotic lesions of TRAIL(-/-)ApoE(-/-) mice. The involvement of OPG and RANKL in this process was also examined. TRAIL dose-dependently inhibited calcium-induced calcification of human VSMCs, while TRAIL(-/-) VSMCs demonstrated accelerated calcification induced by multiple concentrations of calcium compared to wild-type cells. Consistent with this, RANKL mRNA was significantly elevated with 24 h calcium treatment, while OPG and TRAIL expression in human VSMCs was inhibited. Brachiocephalic arteries from TRAIL(-/-)ApoE(-/-) and ApoE(-/-) mice fed a high fat diet for 12 w demonstrated increased chondrocyte-like cells in atherosclerotic plaque, as well as increased aortic collagen II mRNA expression in TRAIL(-/-)ApoE(-/-) mice, with significant increases in calcification observed at 20 w. TRAIL(-/-)ApoE(-/-) aortas also had significantly elevated RANKL, BMP-2, IL-1ß, and PPAR-γ expression at 12 w. Our data provides the first evidence that TRAIL deficiency results in accelerated cartilaginous metaplasia and calcification in atherosclerosis, and that TRAIL plays an important role in the regulation of RANKL and inflammatory markers mediating bone turn over in the vasculature.


Asunto(s)
Aorta/metabolismo , Aterosclerosis/genética , Tronco Braquiocefálico/metabolismo , Placa Aterosclerótica/genética , Ligando RANK/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Calcificación Vascular/genética , Animales , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/patología , Calcio/metabolismo , Calcio/farmacología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Dieta Alta en Grasa , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Ligando RANK/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/deficiencia , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
11.
J Am Heart Assoc ; 2(4): e000263, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23900214

RESUMEN

BACKGROUND: Endothelial cell responses during inflammation are heterogeneous and key for selectivity in how leukocytes hone in on specific sites and why vascular diseases are highly bed specific. However, mechanisms for this specificity remain unclear. METHODS AND RESULTS: Here, we exposed human endothelial cells isolated from 5 systemic arterial beds from 1 donor (to overcome donor-to-donor genetic/epigenetic differences), the umbilical vein, and pulmonary microvasculature to TNF-α, LPS, and IL-1ß and assessed acute (ERK1/2 and p65) and chronic (ICAM-1, VCAM-1 total and surface expression) signaling responses and assessed changes in surface N-glycans and monocyte adhesion. Significant diversity in responses was evident by disparate changes in ERK1/2 and p65 NF-κB phosphorylation, which varied up to 5-fold between different cells and in temporal and magnitude differences in ICAM-1 and VCAM-1 expression (maximal VCAM-1 induction typically being observed by 4 hours, whereas ICAM-1 expression was increased further at 24 hours relative to 4 hours). N-glycan profiles both basally and with stimulation were also bed specific, with hypoglycosylated N-glycans correlating with increased THP-1 monocyte adhesion. Differences in surface N-glycan expression tracked with dynamic up- or downregulation of α-mannosidase activity during inflammation. CONCLUSIONS: These results demonstrate a critical role for the vascular bed of origin in controlling endothelial responses and function to inflammatory stimuli and suggest that bed-specific expression of N-linked sugars may provide a signature for select leukocyte recruitment.


Asunto(s)
Células Endoteliales/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Pulmón/irrigación sanguínea , Aorta/inmunología , Aorta/metabolismo , Tronco Braquiocefálico/inmunología , Tronco Braquiocefálico/metabolismo , Arterias Carótidas/inmunología , Arterias Carótidas/metabolismo , Adhesión Celular , Línea Celular , Quimiotaxis de Leucocito , Vasos Coronarios/inmunología , Vasos Coronarios/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Femenino , Glicosilación , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Masculino , Microvasos/inmunología , Microvasos/metabolismo , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Fosforilación , Transducción de Señal , Arteria Subclavia/inmunología , Arteria Subclavia/metabolismo , Factores de Tiempo , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 33(11): 2491-500, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23990207

RESUMEN

OBJECTIVE: Osteoprotegerin (OPG) is a decoy receptor for the osteoclast differentiation factor receptor activator of NF-κB ligand. OPG regulates bone homeostasis, and its inactivation in mice results in severe osteoporosis. OPG deficiency in apolipoprotein E (ApoE)(-/-) mice results in increased atherosclerotic lesion size and calcification. Furthermore, receptor activator of NF-κB ligand enhances macrophage-dependent smooth muscle cell calcification in vitro. Here, we hypothesized that reconstitution of ApoE(-/-)OPG(-/-) mice with ApoE(-/-)OPG(+/+) bone marrow (BM) would be sufficient to rescue lesion progression and vascular calcification. Conversely, reconstitution of ApoE(-/-)OPG(+/+) mice with ApoE(-/-)OPG(-/-) BM may accelerate lesion progression and vascular calcification. APPROACH AND RESULTS: ApoE(-/-)OPG(-/-) mice transplanted with ApoE(-/-)OPG(+/+) BM developed smaller atherosclerotic lesions and deposited less calcium in the innominate artery than that of ApoE(-/-)OPG(-/-) mice transplanted with ApoE(-/-)OPG(-/-) BM. There were no differences in lesion size and calcification in ApoE(-/-)OPG(+/+) mice transplanted with BM from ApoE(-/-)OPG(-/-) or ApoE(-/-)OPG(+/+) mice. The large lesions observed in the ApoE(-/-)OPG(-/-) mice transplanted with OPG(-/-) BM were rich in chondrocyte-like cells, collagen, and proteoglycans. Importantly, the ApoE(-/-)OPG(-/-) mice transplanted with OPG(+/+) BM remained osteoporotic, and the ApoE(-/-)OPG(+/+) mice did not show signs of bone loss regardless of the type of BM received. In coculture experiments, macrophages and mesenchymal stem cells derived from ApoE(-/-)OPG(-/-) BM induced more vascular smooth muscle cell calcification than cells derived from ApoE(-/-)OPG(+/+) mice. CONCLUSIONS: These results indicate that OPG derived either from the BM or from the vessel wall is sufficient to slow down lesion progression and vascular calcification independent of bone turnover.


Asunto(s)
Aterosclerosis , Trasplante de Médula Ósea , Médula Ósea/metabolismo , Osteoprotegerina/metabolismo , Calcificación Vascular , Animales , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/terapia , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Condrocitos/metabolismo , Condrocitos/patología , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Osteoprotegerina/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , Calcificación Vascular/terapia
13.
Eur J Pharmacol ; 697(1-3): 106-16, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23085268

RESUMEN

Hydrogen sulfide (H(2)S) is a novel gaseous mediator that plays important roles in atherosclerosis. The present study investigated the effect of a novel H(2)S-releasing aspirin, ACS14 (2-acetyloxybenzoic acid 4-(3-thioxo-3H-1,2-dithiol-5-yl)phenyl ester), on atherosclerotic plaques in fat-fed apoE(-/-) mice and the underlying mechanism with respect to CX3C chemokine receptor 1 (CX3CR1) in macrophages. Mouse macrophage cell line RAW264.7 or mouse peritoneal macrophages were preincubated with aspirin (50, 100 or 200µM), ACS14 (50, 100 or 200µM) or vehicle for 6h, and then stimulated with interferon (IFN)-γ (500U/ml) or lipopolysaccharide (LPS; 10µg/ml) for 12h. ACS14, but not aspirin, dose-dependently inhibited IFN-γ or LPS-induced CX3CR1 expression and CX3CR1-mediated chemotaxis in macrophages. The inhibitory effect of ACS14 on CX3CR1 expression was abolished by pretreatment with GW9662, a selective peroxisome proliferator-activated receptor (PPAR)-γ antagonist, suggesting that suppression of macrophage CX3CR1 expression by ACS14 is PPAR-γ dependent. Eight-week-old male apoE(-/-) mice received intraperitoneal ACS14 (15 or 30µmol/kg/day) or aspirin (15 or 30µmol/kg/day) 4 weeks after fat feeding. Twelve weeks after ACS14 or aspirin treatment, mice were sacrificed to evaluate the extent of atherosclerosis and CX3CR1 expression in brachiocephalic artery (BCA). We found that ACS14, but not aspirin, significantly downregulated CX3CR1 expression in atherosclerotic plaques. ACS14 considerably impeded the formation and development of atherosclerosis as compared to a molar equivalent dose of aspirin. These data indicate that ACS14 may prevent the progression of atherosclerosis by downregulating macrophage CX3CR1 expression via a PPAR-γ-dependent mechanism.


Asunto(s)
Apolipoproteínas E/deficiencia , Aspirina/análogos & derivados , Aterosclerosis/tratamiento farmacológico , Tronco Braquiocefálico/efectos de los fármacos , Disulfuros/farmacología , Sulfuro de Hidrógeno/metabolismo , Macrófagos/efectos de los fármacos , Animales , Apolipoproteínas E/genética , Aspirina/administración & dosificación , Aspirina/metabolismo , Aspirina/farmacología , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/inmunología , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Receptor 1 de Quimiocinas CX3C , Línea Celular , Quimiotaxis/efectos de los fármacos , Modelos Animales de Enfermedad , Disulfuros/administración & dosificación , Disulfuros/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Inmunohistoquímica , Inyecciones Intraperitoneales , Interferón gamma/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/efectos de los fármacos , PPAR gamma/metabolismo , Placa Aterosclerótica , Receptores de Quimiocina/efectos de los fármacos , Receptores de Quimiocina/metabolismo , Factores de Tiempo
14.
Cardiovasc Drugs Ther ; 26(1): 23-30, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22038096

RESUMEN

PURPOSE: Simvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, has antioxidant and anti-inflammatory properties that are independent of lipid-lowering abilities. This experiment was carried out to explore the effects of simvastatin on apoptosis in vulnerable atherosclerotic plaques of apoE-deficient mice. METHODS AND RESULTS: Eight weeks-old apoE(-/-) mice were fed a Western-type diet. Vulnerable atherosclerotic lesions were formed in the branchiocephalic artery at the age of 30-weeks, before simvastatin administration for 8 weeks. Simvastatin did neither affect the levels of plasma glucose and lipids, nor the size of atherosclerotic lesions. Analysis of plaque composition showed that simvastatin decreased the area of lipid core and increased the amounts of macrophages and smooth muscle cells in atherosclerotic plaques of apoE(-/-) mice. In addition, simvastatin down-regulated the expression of vascular cell adhesion molecule-1 (VCAM-1) by both inhibition of nuclear factor kappa B (NF-кB) activation and suppression of the expression of the receptor for advanced glycation end products (RAGE). Moreover, we found that simvastatin administration led to reduced TUNEL-positive cells in the aortic root lesions, accompanied by up-regulation of Bcl-2 and Bcl-xL expression, and decreased P(53) expression as shown by Western blot. CONCLUSION: In the present study, we show novel data to suggest that simvastatin could suppress apoptosis in vulnerable atherosclerotic plaques of apoE(-/-) mice by regulating the expression of apoptosis-related proteins, such as p(53), Bcl-2 and Bcl-xL.


Asunto(s)
Apoptosis/efectos de los fármacos , Aterosclerosis/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Placa Aterosclerótica/metabolismo , Simvastatina/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Glucemia/análisis , Tronco Braquiocefálico/efectos de los fármacos , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Lípidos/sangre , Masculino , Ratones , Ratones Noqueados , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Simvastatina/uso terapéutico , Proteína p53 Supresora de Tumor/metabolismo , Proteína bcl-X/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 31(11): 2473-82, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21868708

RESUMEN

OBJECTIVE: Glutamate-cysteine ligase (GCL) is the rate-limiting step in glutathione synthesis. The enzyme is a heterodimer composed of a catalytic subunit, GCLC, and a modifier subunit, GCLM. We generated apolipoprotein E (apoE)-/- mice deficient in GCLM (apoE-/-/Gclm-/-) and transgenic mice that overexpress GCLC specifically in macrophages (apoE-/-/Gclc-Tg) to test the hypothesis that significantly altering the availability of glutathione has a measurable impact on both the initiation and progression of atherosclerosis. METHODS AND RESULTS: Atherosclerotic plaque size and composition were measured in the innominate artery in chow-fed male and female mice at 20, 30, 40, and 50 weeks of age and in the aortic sinus at 40 and 50 weeks of age. The apoE-/-/Gclm-/- mice more rapidly developed complex lesions, whereas the apoE-/-/Gclc-Tg mice had reduced lesion development compared with the littermate apoE-/- control mice. Transplantation of bone marrow from the apoE-/-/Gclm-/- and apoE-/-/Gclc-Tg mice into apoE-/- mice with established lesions also stimulated or inhibited further lesion development at 30 weeks posttransplant. CONCLUSION: Gain and loss of function in the capacity to synthesize glutathione especially in macrophages has reciprocal effects on the initiation and progression of atherosclerosis at multiple sites in apoE-/- mice.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/patología , Glutatión/metabolismo , Animales , Apolipoproteínas E/genética , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Glutamato-Cisteína Ligasa/deficiencia , Glutamato-Cisteína Ligasa/genética , Lípidos/sangre , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Seno Aórtico/metabolismo , Seno Aórtico/patología
16.
FASEB J ; 25(7): 2447-55, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21490292

RESUMEN

The complement C5a receptor, CD88, is present on many of the cells found within human atherosclerotic plaques, but little is known about the role of C5a in atherogenesis. Using real-time PCR, we determined that ApoE(-/-) mice fed a normal diet express more aortic CD88 mRNA compared with controls, and this increase coincides with atherosclerotic lesion development (P<0.001 for 3- vs. 25-wk-old animals). Conversely, mRNA expression of the alternative C5a receptor, C5L2, in aortas of ApoE(-/-) mice, was lower than controls at all time points. Using immunohistochemistry, we confirmed the presence of CD88 on macrophages, smooth muscle cells, and activated endothelial cells in plaques from brachiocephalic arteries. Treatment of ApoE(-/-) mice with a CD88 antagonist (PMX53; 3 mg/kg s.c. 3 ×/wk plus 1 mg/kg/d p.o.) for 25 wk reduced lesion size and lipid content in the plaque by ∼ 40% (P<0.05). Our study provides evidence for a proatherogenic role for C5a and identifies the CD88 antagonist PMX53 as a potential antiatherosclerotic drug.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Complemento C5a/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/prevención & control , Tronco Braquiocefálico/efectos de los fármacos , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Colesterol/sangre , Complemento C5a/antagonistas & inhibidores , Células Endoteliales/metabolismo , Femenino , Inmunohistoquímica , Lípidos/análisis , Lípidos/sangre , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Péptidos Cíclicos/farmacología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/genética , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Cardiovasc Res ; 91(1): 37-44, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21354994

RESUMEN

AIMS: Plasma concentrations of high-density lipoprotein (HDL)-cholesterol correlate inversely with the incidence of myocardial infarction in humans. We investigated the effect of treatment with human apolipoprotein A-I (apoA-I), the principal protein of HDL, on plaque disruption in an animal model. METHODS AND RESULTS: Seventy apolipoprotein E knockout mice were induced to develop atherosclerotic lesions in the brachiocephalic artery by feeding a high-fat diet for 9 weeks. Mice then received twice-weekly treatment with human apoA-I (8 mg/kg) or vehicle, for 2 weeks. The incidence of acute plaque disruption was reduced by 65% in mice receiving apoA-I (P < 0.01). Plaques in treated mice had a more stable phenotype, with an increase in smooth muscle cell (SMC): macrophage ratio (P = 0.05), principally the consequence of an increase in the number of SMC in plaques. In the fibrous cap, there were reductions in matrix metalloproteinase-13 (-69%, P < 0.0001) and S100A4, a marker of SMC de-differentiation (-60%, P < 0.0001). These results indicate that 2 weeks of treatment with small amounts of human apoA-I produces more stable plaques in a mouse model. CONCLUSION: Treatment with apoA-I has the potential to stabilize plaques and prevent plaque rupture in humans.


Asunto(s)
Apolipoproteína A-I/administración & dosificación , Aterosclerosis/tratamiento farmacológico , Tronco Braquiocefálico/efectos de los fármacos , Fármacos Cardiovasculares/administración & dosificación , Placa Aterosclerótica/tratamiento farmacológico , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Desdiferenciación Celular , HDL-Colesterol/sangre , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Placa Aterosclerótica/sangre , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Rotura Espontánea , Proteína de Unión al Calcio S100A4 , Proteínas S100/metabolismo
18.
Atherosclerosis ; 215(1): 52-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21251656

RESUMEN

OBJECTIVE: Studies in humans support a role for the oral pathogen Porphyromonas gingivalis in the development of inflammatory atherosclerosis. The goal of this study was to determine if P. gingivalis infection accelerates inflammation and atherosclerosis in the innominate artery of mice, an artery which has been reported to exhibit many features of human atherosclerotic disease, including plaque rupture. METHODS AND RESULTS: Apolipoprotein E-deficient (ApoE-/-) mice were orally infected with P. gingivalis, and magnetic resonance imaging (MRI) was used to monitor the progression of atherosclerosis in live mice. P. gingivalis infected mice exhibited a statistically significant increase in atherosclerotic plaque in the innominate artery as compared to uninfected mice. Polarized light microscopy and immunohistochemistry revealed that the innominate arteries of infected mice had increased lipids, macrophages and T cells as compared to uninfected mice. Increases in plaque, total cholesterol esters and cholesterol monohydrate crystals, macrophages, and T cells were prevented by immunization with heat-killed P. gingivalis prior to pathogen exposure. CONCLUSIONS: These are the first studies to demonstrate progression of inflammatory plaque accumulation in the innominate arteries by in vivo MRI analysis following pathogen exposure, and to document protection from plaque progression in the innominate artery via immunization.


Asunto(s)
Aterosclerosis/inmunología , Infecciones por Bacteroidaceae/inmunología , Tronco Braquiocefálico/patología , Inflamación/etiología , Inflamación/prevención & control , Porphyromonas gingivalis/inmunología , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/patología , Infecciones por Bacteroidaceae/patología , Tronco Braquiocefálico/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Metabolismo de los Lípidos , Macrófagos/inmunología , Angiografía por Resonancia Magnética , Imagen por Resonancia Magnética , Masculino , Ratones , Placa Aterosclerótica/etiología , Linfocitos T/inmunología
19.
Arterioscler Thromb Vasc Biol ; 29(2): 195-201, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19008530

RESUMEN

OBJECTIVE: Vascular smooth muscle cell (VSMC) apoptosis contributes to atherosclerotic plaque instability and myocardial infarction. Consequently, reducing VSMC apoptosis may be beneficial for reducing plaque instability and acute coronary events. We previously demonstrated that N-cadherin, a cell-cell adhesion molecule, reduces VSMC apoptosis in vitro. In this study, we examined whether a soluble form of N-cadherin (SNC) affected VSMC apoptosis and plaque stability. METHODS AND RESULTS: SNC significantly inhibited VSMC apoptosis in vitro by approximately 50% via activation of fibroblast growth factor receptor, phosphoinositide-3 kinase, and Akt signaling. SNC also significantly reduced macrophage and foam cell-macrophage apoptosis in vitro by >50%, without affecting monocyte invasion or macrophage proliferation. Elevation of plasma levels of SNC in male apolipoprotein E-deficient mice with existing atherosclerosis via adenoviral delivery significantly reduced VSMC and macrophage apoptosis in brachiocephalic artery plaques by approximately 60%. Additionally, SNC promoted plaques of a more stable phenotype by elevating VSMC:macrophage ratio and presence of VSMC-rich fibrous cap, as well as attenuating macrophage number and incidence of buried fibrous caps (a surrogate plaque rupture marker). CONCLUSIONS: In summary, this study demonstrates that SNC suppressed plaque instability by attenuation of apoptosis, suggesting that SNC may have a therapeutic potential for retarding plaque instability.


Asunto(s)
Apoptosis , Aterosclerosis/terapia , Tronco Braquiocefálico/patología , Cadherinas/biosíntesis , Terapia Genética/métodos , Adenoviridae/genética , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/metabolismo , Células CHO , Cadherinas/genética , Adhesión Celular , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Células Espumosas/metabolismo , Células Espumosas/patología , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fragmentos de Péptidos/biosíntesis , Transducción de Señal , Factores de Tiempo , Transducción Genética , Regulación hacia Arriba
20.
Arterioscler Thromb Vasc Biol ; 28(11): 2009-15, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18787185

RESUMEN

UNLABELLED: Background- The nuclear liver X receptor-alpha (LXR-alpha) has been implicated in the regulation of intracellular cholesterol homeostasis, inflammatory response, and atherosclerosis susceptibility. The aim of the present study was to test whether transgenic expression of LXR-alpha might affect these mechanisms and result in a reduction of atherosclerosis. METHODS AND RESULTS: We generated mice with macrophage overexpression of mouse LXR-alpha, evidenced by significantly elevated expression levels of LXR-target genes (ABCA1, ABCG1) in these cells. For atherosclerosis studies, mice were crossed onto the LDL-receptor deficient background. Plasma lipids and lipoproteins as well as liver triglycerides were not significantly different between transgenic animals and nontransgenic controls. However, lesion area at the brachiocephalic artery (BCA) was significantly reduced (-83%, P=0.02) in male LXR-alpha transgenic mice. This was associated with a significantly increased cholesterol efflux to acceptor-free media (+24%, P=0.002) and ApoA1 containing media (+20%, P<0.0001) as well as reduced lipopolysaccharide (LPS)-induced NO-release from macrophages of transgenic animals, providing a potential mechanism for the reduction of atherosclerosis. CONCLUSIONS: Our data show for the first time that transgenic overexpression of LXR-alpha in macrophages has significant antiatherogenic properties. We conclude that overexpression of LXR-alpha in macrophages might be useful as a therapeutic principle for the prevention of atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Proteínas de Unión al ADN/metabolismo , Inflamación/prevención & control , Hígado/metabolismo , Macrófagos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de LDL/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Células Cultivadas , Colesterol en la Dieta/metabolismo , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Lipoproteínas/metabolismo , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico/metabolismo , Receptores Nucleares Huérfanos , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores de LDL/deficiencia , Receptores de LDL/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA