Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Ann Biomed Eng ; 43(11): 2760-70, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25986954

RESUMEN

We previously reported differences in stiffness between macro- and micro-vessels in type 2 diabetes (T2DM). The aim of this study was to define the mechanical properties of the ECM independent of vascular cells in coronary resistance micro-vessels (CRMs) and macro-vessels (aorta) in control Db/db and T2DM db/db mice. Passive vascular remodeling and mechanics were measured in both intact and decellularized CRMs and aortas from 0 to 125 mmHg. We observed no differences in intact control and diabetic aortic diameters, wall thicknesses, or stiffnesses (p > 0.05). Aortic decellularization caused a significant increase in internal and external diameters and incremental modulus over a range of pressures that occurred to a similar degree in T2DM. Differences in aortic diameters due to decellularization occurred at lower pressures (0-75 mmHg) and converged with intact aortas at higher, physiological pressures (100-125 mmHg). In contrast, CRM decellularization caused increased internal diameter and incremental modulus only in the db/db mice, but unlike the aorta, the intact and decellularized CRM curves were more parallel. These data suggest that (1) micro-vessels may be more sensitive to early adverse consequences of diabetes than macro-vessels and (2) the ECM is a structural limit in aortas, but not CRMs.


Asunto(s)
Aorta Torácica/fisiología , Vasos Coronarios/fisiología , Diabetes Mellitus Tipo 2/fisiopatología , Microvasos/fisiología , Animales , Circulación Coronaria , Masculino , Ratones , Resistencia Vascular
2.
Can J Physiol Pharmacol ; 92(7): 598-604, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24959995

RESUMEN

Diabetes is an independent risk factor for cardiovascular disease that can eventually cause cardiomyopathy and heart failure. Cardiac fibroblasts (CF) are the critical mediators of physiological and pathological cardiac remodeling; however, the effects of hyperglycemia on cardiac fibroblast function and differentiation is not well known. Here, we performed a comprehensive investigation on the effects of hyperglycemia on cardiac fibroblasts and show that hyperglycemia enhances cardiac fibroblast function and differentiation. We found that high glucose treatment increased collagen I, III, and VI gene expression in rat adult cardiac fibroblasts. Interestingly, hyperglycemia increased CF migration and proliferation that is augmented by collagen I and III. Surprisingly, we found that short term hyperglycemia transiently inhibited ERK1/2 activation but increased AKT phosphorylation. Finally, high glucose treatment increased spontaneous differentiation of cardiac fibroblasts to myofibroblasts with increasing passage compared with low glucose. Taken together, these findings suggest that hyperglycemia induces cardiac fibrosis by modulating collagen expression, migration, proliferation, and differentiation of cardiac fibroblasts.


Asunto(s)
Diferenciación Celular , Fibroblastos/metabolismo , Hiperglucemia/metabolismo , Miocardio/metabolismo , Animales , Glucemia/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Fibroblastos/patología , Fibrosis , Hiperglucemia/patología , Masculino , Miocardio/patología , Ratas Sprague-Dawley , Transducción de Señal
3.
Circ Res ; 110(6): 851-6, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22343710

RESUMEN

RATIONALE: We previously reported that type VI collagen deposition increases in the infarcted myocardium in vivo. To date, a specific role for this nonfibrillar collagen has not been explored in the setting of myocardial infarction (MI). OBJECTIVE: To determine whether deletion of type VI collagen in an in vivo model of post-MI wound healing would alter cardiac function and remodeling in the days to weeks after injury. METHODS AND RESULTS: Wild-type and Col6a1(-/-) mice were subjected to MI, followed by serial echocardiographic and histological assessments. At 8 weeks after MI, infarct size was significantly reduced, ejection fraction was significantly preserved (43.9% ± 3.3% versus 29.1% ± 4.3% for wild-type), and left ventricular chamber dilation was attenuated in the Col6a1(-/-) MI group (25.8% ± 7.9% increase versus 62.6% ± 16.5% for wild-type). The improvement in cardiac remodeling was evident as early as 10 days after MI in the Col6a1(-/-) mice. Myocyte apoptosis within the infarcted zones was initially greater in the Col6a1(-/-) group 3 days after MI, but by day 14 this was significantly reduced. Collagen deposition also was reduced in the infarcted and remote areas of the Col6a1(-/-) hearts. The reductions in chronic myocyte apoptosis and fibrosis are critical events leading to improved long-term remodeling and functional outcomes. CONCLUSIONS: These unexpected results demonstrate for the first time that deletion of type VI collagen in this knockout model plays a critical protective role after MI by limiting infarct size, chronic apoptosis, aberrant remodeling, and fibrosis, leading to preservation of cardiac function.


Asunto(s)
Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Remodelación Ventricular/fisiología , Animales , Apoptosis/fisiología , Modelos Animales de Enfermedad , Ecocardiografía , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosis/genética , Fibrosis/patología , Fibrosis/fisiopatología , Masculino , Ratones , Ratones Noqueados , Infarto del Miocardio/diagnóstico por imagen , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología
4.
Invest New Drugs ; 29(2): 380-91, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19812893

RESUMEN

Hepatocellular carcinoma (HCC), one of the most lethal cancers, results in more than one million fatalities worldwide every year. In view of the limited therapeutic alternatives and poor prognosis of liver cancer, preventive control approaches, notably chemoprevention, have been considered to be the best strategy in lowering the present prevalence of the disease. Resveratrol, a naturally occurring antioxidant and antiinflammatory agent found in grapes and red wine, inhibits carcinogenesis with a pleiotropic mode of action. Recently, we have reported that dietary resveratrol significantly prevents chemically-induced liver tumorigenesis in rats. One of the mechanisms of resveratrol-mediated chemoprevention of hepatocarcinogenesis could be related to its antiinflammatory action through hepatic cyclooxygenase (COX-2) inhibition. Although several COX-2 inhibitors are known to exert chemopreventive efficacy, not all are considered ideal candidates for chemoprevention due to the risk of adverse cardiovascular events. Accordingly, the objective of the present study was to evaluate the role of resveratrol on cardiac performance during experimental hepatocarcinogenesis initiated with diethylnitrosamine and promoted by phenobarbital. Rats had free access to diet supplemented with resveratrol four weeks before the carcinogen injection and 14 weeks thereafter. The cardiotoxicity of resveratrol was assessed by monitoring the cardiac function using transthoracic echocardiography as well as Western blot analysis of cardiac tissue. Long-term dietary administration of resveratrol dose-dependently suppressed hepatic tumor multiplicity, the principal endpoint for evaluating the chemopreventive potential of a candidate agent. The chemopreventive effects of resveratrol were also reflected in histopathological assessment of hepatic tissues. Resveratrol did not exhibit any cardiotoxicity but rather improved the cardiac function in a dose-responsive fashion. Our results indicate that resveratrol-mediated chemoprevention of rat liver carcinogenesis is devoid of any adverse cardiovascular events. Resveratrol may be developed as a chemopreventive as well as therapeutic drug for human HCC.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Cardiotoxinas/toxicidad , Quimioprevención , Neoplasias Hepáticas/tratamiento farmacológico , Estilbenos/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Western Blotting , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ecocardiografía , Conducta Alimentaria/efectos de los fármacos , Femenino , Corazón/efectos de los fármacos , Corazón/fisiopatología , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Hígado/fisiopatología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/fisiopatología , Ratas , Ratas Sprague-Dawley , Resveratrol , Sístole/efectos de los fármacos
5.
J Mol Cell Cardiol ; 48(3): 530-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19573533

RESUMEN

Cardiac remodeling is accelerated during pathological conditions and several anabolic and catabolic regulators work in concert to repair the myocardium and maintain its functionality. The fibroblasts play a major role in this process via collagen deposition as well as supplying the degradative matrix metalloproteinases. During the more acute responses to a myocardial infarction (MI) the heart relies on a more aggressive wound healing sequence that includes the myofibroblasts, specialized secretory cells necessary for infarct scar formation and thus, rescue of the myocardium. The activated fibroblasts and myofibroblasts deposit large amounts of fibrillar collagen during the post-MI wound healing phase, type I and III collagen are the most abundant collagens in the heart and they maintain the structural integrity under normal and disease states. While collagen I and III have been the traditional focus of the myocardial matrix, recent studies have suggested that the non-fibrillar collagens (types IV and VI) are also deposited during pathological wound healing and may play key roles in myofibroblast differentiation and organization of the fibrillar collagen network. This review highlights the potential roles of the non-fibrillar collagens and how they work in concert with the fibrillar collagens in mediating myocardial remodeling.


Asunto(s)
Infarto del Miocardio/metabolismo , Colágenos no Fibrilares/metabolismo , Animales , Humanos , Metaloproteinasas de la Matriz/metabolismo , Modelos Biológicos , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Cicatrización de Heridas/fisiología
6.
Am J Physiol Endocrinol Metab ; 297(5): E1147-53, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19706787

RESUMEN

Diabetic patients are prone to developing myocardial fibrosis and suffer from decreased wound healing capabilities. The purpose of this study was to determine whether diabetes alters cardiac fibroblast activity in the myocardium in a 6-wk streptozotocin-induced type 1 diabetic model. In vivo echocardiography indicated significant dilation of the left ventricle (LV) in the diabetic animals, while cardiac function was comparable to that in the normal group. We isolated cardiac fibroblasts from diabetic and control hearts and observed increased proliferation of the diabetic fibroblasts. Microarray analysis using mRNA collected from whole LVs revealed downregulation of known inhibitors of proliferation, p53 and p21, in the diabetic group, consistent with our proliferation data. Western blot analysis confirmed a reduction in p53 protein expression in the diabetic hearts compared with control. We explored the potential signaling underlying the downregulation of these cell cycle mediators and determined that activated Akt, a signal that inhibits p53, was elevated in the diabetic group. Surprisingly, the hearts from the diabetic group contained lower levels of the myofibroblast marker α-smooth muscle actin (α-SMA) and higher levels of desmin and platelet endothelial cell adhesion molecule (PECAM). The isolated fibroblasts from the diabetic group also contained significantly less α-SMA. These data suggest that early-stage diabetic hearts contain highly proliferative fibroblasts, which predisposes the diabetic myocardium to fibrosis, but have fewer myofibroblasts, which may compromise wound healing.


Asunto(s)
Ciclo Celular/fisiología , Diabetes Mellitus Tipo 1/patología , Fibroblastos/fisiología , Miocardio/patología , Miofibroblastos/fisiología , Animales , Glucemia/metabolismo , Western Blotting , Peso Corporal/fisiología , Proteínas de Ciclo Celular/biosíntesis , Diferenciación Celular/fisiología , Proliferación Celular , Separación Celular , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/diagnóstico por imagen , Ecocardiografía , Masculino , Análisis por Micromatrices , Miocardio/citología , Fenotipo , ARN/biosíntesis , ARN/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
7.
J Mol Cell Cardiol ; 46(2): 186-92, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19041328

RESUMEN

Cardiac fibroblasts and myofibroblasts are responsible for post-MI remodeling which occurs via regulation of extracellular matrix (ECM). Accelerated post-MI remodeling leads to excessive ECM deposition and fibrosis, contributing to impaired contractile function, arrhythmias, and heart failure. We have previously reported that type VI collagen induces myofibroblast differentiation in cultured cardiac fibroblasts, and that type VI collagen and myofibroblast content were both elevated in the myocardium 20 weeks post-MI. The purpose of this study was to determine the expression patterns of type VI collagen and myofibroblast content in early post-myocardial infarction (MI) remodeling to gain insight into whether type VI collagen induces in vivo myofibroblast differentiation via specific matrix-receptor interactions. Adult male Sprague-Dawley rats were anesthetized and left coronary arteries were permanently ligated. Histological tissue sections and whole tissue protein lysates were obtained from infarcted and non-infarcted areas of MI hearts and sham operated controls. At 3 days post-MI, we observed a significant increase in alpha(3) integrin expression (2.02+/-0.18 fold); at 7 days post-infarction both type VI collagen (2.27+/-0.18 fold) and myofibroblast (4.65+/-0.6 fold) content increased. By 14 days myofibroblast content returned to sham control levels, although type VI collagen (2.42+/-0.11 fold) was still elevated. In vitro cross-linking confirmed that the alpha(3) integrin interacts with type VI collagen, and alpha(3) integrin function blocking antibodies inhibited the differentiation of isolated cardiac fibroblasts. Collectively, our in vitro results indicate that the alpha(3) integrin receptor interacts with type VI collagen to promote myofibroblast differentiation, and that this interaction may impact in vivo post-MI remodeling.


Asunto(s)
Diferenciación Celular/fisiología , Colágeno Tipo VI/metabolismo , Fibroblastos/citología , Integrina alfa3/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/citología , Animales , Colágeno Tipo VI/fisiología , Immunoblotting , Masculino , Unión Proteica , Ratas
8.
Hypertension ; 51(3): 704-11, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18195168

RESUMEN

Angiotensin II (Ang II)-induced proliferation of cardiac fibroblasts is a major contributing factor to the pathogenesis of cardiac fibrosis. Ang II activates extracellular signal-regulated kinase (ERK) 1/2 to induce cardiac fibroblast proliferation, but the signaling pathways leading to ERK 1/2 activation have not been elucidated in these cells. The goal of the current study was to identify the intracellular mediators of Ang II-induced ERK 1/2 activation in adult rat cardiac fibroblasts. We determined that 100 nmol/L of Ang II-induced ERK 1/2 phosphorylation is inhibited by simultaneous chelation of cytosolic calcium and downregulation of protein kinase C (PKC) by phorbol ester or by the specific PKCdelta inhibitor rottlerin, as well as PKCdelta small interfering RNA, but not by inhibition of 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetate, phorbol ester, rottlerin, or PKCdelta small interfering RNA alone. We also found that Ang II does not transactivate the epidermal growth factor receptor in adult cardiac fibroblasts, because pretreatment with 1 mumol/L of AG 1478 did not significantly inhibit [(3)H]-thymidine incorporation or ERK 1/2 activation. In addition, immunoprecipitation of the epidermal growth factor receptor demonstrated no significant Ang II-induced phosphorylation of tyrosine residues. Inhibition of phosphatidylinositide 3-kinase, PKCzeta, and src tyrosine kinase had no effect on Ang II-induced ERK 1/2 activation. Collectively, these data demonstrate that Ang II does not transactivate the epidermal growth factor receptor in adult rat cardiac fibroblasts to activate ERK 1/2, a common pathway described in vascular smooth muscle and other cell types, but rather occurs via activation of distinct parallel signaling pathways mechanistically controlled by intracellular Ca(2+) and PKCdelta.


Asunto(s)
Angiotensina II/fisiología , Calcio/metabolismo , Fibroblastos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocardio/citología , Proteína Quinasa C-delta/metabolismo , Acetofenonas/farmacología , Animales , Benzopiranos/farmacología , Proliferación Celular , Células Cultivadas , Activación Enzimática , Receptores ErbB/metabolismo , Masculino , Miocardio/metabolismo , Ésteres del Forbol/farmacología , Fosforilación , Proteína Quinasa C-delta/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA