Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
2.
Cardiovasc Diabetol ; 19(1): 7, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31924211

RESUMEN

BACKGROUND: Diabetes mellitus type 2 (DM2) is a risk factor for developing heart failure but there is no specific therapy for diabetic heart disease. Sodium glucose transporter 2 inhibitors (SGLT2I) are recently developed diabetic drugs that primarily work on the kidney. Clinical data describing the cardiovascular benefits of SGLT2Is highlight the potential therapeutic benefit of these drugs in the prevention of cardiovascular events and heart failure. However, the underlying mechanism of protection remains unclear. We investigated the effect of Dapagliflozin-SGLT2I, on diabetic cardiomyopathy in a mouse model of DM2. METHODS: Cardiomyopathy was induced in diabetic mice (db/db) by subcutaneous infusion of angiotensin II (ATII) for 30 days using an osmotic pump. Dapagliflozin (1.5 mg/kg/day) was administered concomitantly in drinking water. Male homozygous, 12-14 weeks old WT or db/db mice (n = 4-8/group), were used for the experiments. Isolated cardiomyocytes were exposed to glucose (17.5-33 mM) and treated with Dapagliflozin in vitro. Intracellular calcium transients were measured using a fluorescent indicator indo-1. RESULTS: Angiotensin II infusion induced cardiomyopathy in db/db mice, manifested by cardiac hypertrophy, myocardial fibrosis and inflammation (TNFα, TLR4). Dapagliflozin decreased blood glucose (874 ± 111 to 556 ± 57 mg/dl, p < 0.05). In addition it attenuated fibrosis and inflammation and increased the left ventricular fractional shortening in ATII treated db/db mice. In isolated cardiomyocytes Dapagliflozin decreased intracellular calcium transients, inflammation and ROS production. Finally, voltage-dependent L-type calcium channel (CACNA1C), the sodium-calcium exchanger (NCX) and the sodium-hydrogen exchanger 1 (NHE) membrane transporters expression was reduced following Dapagliflozin treatment. CONCLUSION: Dapagliflozin was cardioprotective in ATII-stressed diabetic mice. It reduced oxygen radicals, as well the activity of membrane channels related to calcium transport. The cardioprotective effect manifested by decreased fibrosis, reduced inflammation and improved systolic function. The clinical implication of our results suggest a novel pharmacologic approach for the treatment of diabetic cardiomyopathy through modulation of ion homeostasis.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Glucemia/efectos de los fármacos , Diabetes Mellitus/tratamiento farmacológico , Cardiomiopatías Diabéticas/prevención & control , Glucósidos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Angiotensina II , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus/sangre , Cardiomiopatías Diabéticas/inducido químicamente , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Fibrosis , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas Sprague-Dawley , Intercambiador de Sodio-Calcio/metabolismo , Intercambiador 1 de Sodio-Hidrógeno/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-29707604

RESUMEN

We have previously shown that an Epoxyeicosatrienoic Acid (EET) -agonist has pleiotropic effects and reverses cardiomyopathy by decreasing inflammatory molecules and increasing antioxidant signaling. We hypothesized that administration of an EET agonist would increase Peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α), which controls mitochondrial function and induction of HO-1 and negatively regulates the expression of the proinflammatory adipokines CCN3/NOV in cardiac and pericardial tissues. This pathway would be expected to further improve left ventricular (LV) systolic function as well as increase insulin receptor phosphorylation. Measurement of the effect of an EET agonist on oxygen consumption, fractional shortening, blood glucose levels, thermogenic and mitochondrial signaling proteins was performed. Control obese mice developed signs of metabolic syndrome including insulin resistance, hypertension, inflammation, LV dysfunction, and increased NOV expression in pericardial adipose tissue. EET agonist intervention decreased pericardial adipose tissue expression of NOV, while normalized FS, increased PGC-1α, HO-1 levels, insulin receptor phosphorylation and improved mitochondrial function, theses beneficial effect were reversed by deletion of PGC-1α. These studies demonstrate that an EET agonist increases insulin receptor phosphorylation, mitochondrial and thermogenic gene expression, decreased cardiac and pericardial tissue NOV levels, and ameliorates cardiomyopathy in an obese mouse model of the metabolic syndrome.

4.
J Ultrasound ; 18(3): 229-35, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26261465

RESUMEN

ABSTRACT: Cardiorenal syndrome type 1 (CRS-1) is the acute kidney disfunction caused by an acute worsening of cardiac function. CRS-1 is the consequence of renal vasoconstriction secondary to renin-angiotensin system (RAS) activation. No animal models of CRS-1 are described in literature. PURPOSE: To characterize a murine model of CRS-1 by using a high-resolution ultrasound echo-color Doppler system (VEVO2100). MATERIALS: Post-ischemic heart failure was induced by coronary artery ligation (LAD) in seven CD1 mice. Fifteen and thirty days after surgery, mice underwent cardiac and renal echo-color Doppler. Serum creatinine and plasma renin activity were measured after killing. Animals were compared to seven CD1 control mice. RESULTS: Heart failure with left ventricle dilatation (end diastolic area, p < 0.05 vs. controls) and significantly reduced ejection fraction (EF; p < 0.01 vs. controls) was evident 15 days after LAD. We measured a significant renal vasoconstriction in infarcted mice characterized by increased renal pulsatility index (PI; p < 0.05 vs. controls) associated to increased creatinine and renin levels (p < 0.05 vs. controls). CONCLUSIONS: The mice model of LAD is a good model of CRS-1 evaluable by Doppler sonography and characterized by renal vasoconstriction due to the activation of the renin-angiotensin system secondary to heart failure.

5.
Int J Obes (Lond) ; 38(3): 456-65, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23779049

RESUMEN

OBJECTIVE: Renin-angiotensin system (RAS) regulates adipogenic response with adipocyte hypertrophy by increasing oxidative stress. Recent studies have shown the role of peroxisome proliferator-activated receptor-δ (PPARδ) agonist in attenuation of angiotensin II-induced oxidative stress. The aim of this study was to explore a potential mechanistic link between PPARδ and the cytoprotective enzyme heme oxygenase-1 (HO-1) and to elucidate the contribution of HO-1 to the adipocyte regulatory effects of PPARδ agonism in an animal model of enhanced RAS, the Goldblatt 2 kidney 1 clip (2K1C) model. METHOD: We first established a direct stimulatory effect of the PPARδ agonist (GW 501516) on the HO-1 gene by demonstrating increased luciferase activity in COS-7 cells transfected with a luciferase-HO-1 promoter construct. Sprague-Dawley rats were divided into four groups: sham-operated animals, 2K1C rats and 2K1C rats treated with GW 501516, in the absence or presence of the HO activity inhibitor, stannous mesoporphyrin (SnMP). RESULTS: 2K1C animals had increased visceral adiposity, adipocyte hypertrophy, increased inflammatory cytokines, increased circulatory and adipose tisssue levels of renin and Ang II along with increased adipose tissue gp91 phox expression (P<0.05) when compared with sham-operated animals. Treatment with GW 501516 increased adipose tissue HO-1 and adiponectin levels (P<0.01) along with enhancement of Wnt10b and ß-catenin expression. HO-1 induction was accompanied by the decreased expression of Wnt5b, mesoderm specific transcript (mest) and C/EBPα levels and an increased number of small adipocytes (P<0.05). These effects of GW501516 were reversed in 2K1C animals exposed to SnMP (P<0.05). CONCLUSION: Taken together, our study demonstrates, for the first time, that increased levels of Ang II contribute towards adipose tissue dysregulation, which is abated by PPARδ-mediated upregulation of the heme-HO system. These findings highlight the pivotal role and symbiotic relationship of HO-1, adiponectin and PPARδ in the regulation of metabolic homeostasis in adipose tissues.


Asunto(s)
Adipocitos/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hipertensión Renovascular/metabolismo , Riñón/metabolismo , PPAR delta/metabolismo , Angiotensina II/farmacología , Animales , Activación Enzimática , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Renina/sangre
6.
Nutr Diabetes ; 2: e33, 2012 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-23169576

RESUMEN

BACKGROUND: We examined the ability of the apolipoprotein AI mimetic peptide L-4F to improve the metabolic state of female and male ob mice and the mechanisms involved. METHODS: Female and male lean and obese (ob) mice were administered L-4F or vehicle for 6 weeks. Body weight was measured weekly. Fat distribution, serum cytokines and markers of cardiovascular dysfunction were determined at the end of treatment. RESULTS: L-4F significantly decreased serum interleukin (IL)-6, tumor necrosis factor-α and IL-1ß. L-4F improved vascular function, and increased serum adiponectin levels and insulin sensitivity compared with untreated mice. In addition, L-4F treatment increased heme oxygenase (HO)-1, pAKT and pAMPK levels in kidneys of ob animals. pAKT and pAMPK levels were significantly reduced in the presence of an HO inhibitor. Interestingly, L4F did not alter body weight in female mice, but caused a significant reduction in males. CONCLUSIONS: L-4F treatments reduced cardiovascular risk factors and improved insulin sensitivity in female ob mice independent of body fat changes. Reduced inflammatory cytokine levels accompanied by increased HO activity, serum adiponectin and improved insulin sensitivity suggest that L-4F may promote the conversion of visceral fat to a healthier phenotype. Therefore, L-4F appears to be a promising therapeutic strategy for treating both cardiovascular risk factors and insulin resistance in obese patients of either gender.

7.
J Cell Biochem ; 112(9): 2616-26, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21598304

RESUMEN

Diabetes is a major health problem associated with adverse cardiovascular outcomes. The apolipoprotein A-I mimetic peptide L-4F is a putative anti-diabetic drug, has antioxidant and anti-inflammatory proprieties and improves endothelial function. In obese mice L-4F increases adiponectin levels, improving insulin sensitivity, and reducing visceral adiposity. We hypothesized that the pleiotropic actions of L-4F can prevent heart and coronary dysfunction in a mouse model of genetically induced Type II diabetes. We treated db/db mice with either L-4F or vehicle for 8 weeks. Trans-thoracic echocardiography was performed; thereafter, isolated hearts were subjected to ischemia/reperfusion (IR). Glucose, insulin, adiponectin, and pro-inflammatory cytokines (IL-1ß, TNF-α, MCP-1) were measured in plasma and HO-1, pAMPK, peNOS, iNOS, adiponectin, and superoxide in cardiac tissue. In db/db mice L-4F decreased accumulation of subcutaneous and total fat, and increased insulin sensitivity and adiponectin levels while lowering inflammatory cytokines (P < 0.05). L-4F normalized in vivo left ventricular (LV) function of db/db mice, increasing (P < 0.05) fractional shortening and decreasing (P < 0.05) LV dimensions. In I/R experiments, L-4F prevented coronary microvascular resistance from increasing and LV function from deteriorating in the db/db mice. These changes were associated with increased cardiac expression of HO-1, pAMPK, peNOS, and adiponectin and decreased levels of superoxide and iNOS (P < 0.01). In the present study we showed that L-4F prevented myocardial and coronary functional abnormalities in db/db mice. These effects were associated with stimulation of HO-1 resulting in increased levels of anti-inflammatory, anti-oxidative, and vasodilatatory action through a mechanism involving increased levels of adiponectin, pAMPK, and peNOS.


Asunto(s)
Cardiotónicos/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Péptidos/uso terapéutico , Disfunción Ventricular Izquierda/prevención & control , Adenilato Quinasa/metabolismo , Adiponectina/sangre , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Cardiotónicos/farmacología , Citocinas/sangre , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/fisiopatología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Pruebas de Función Cardíaca , Hemo-Oxigenasa 1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Miocardio/enzimología , Miocardio/metabolismo , Óxido Nítrico Sintasa/metabolismo , Péptidos/farmacología , Superóxidos/metabolismo , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/fisiopatología
8.
Curr Pharm Des ; 14(5): 412-21, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18289068

RESUMEN

Heme oxygenase-1 (HO-1) is central to the regulation of oxidative injury. The role of increased HO-1 expression and Heme oxygenase (HO) activity in mitigating the detrimental side effect of diabetes is examined. A review of the mechanism(s) of action is included. This may lead to the development of pharmacological and genetic approaches to mitigate the clinical complications associated with the progression of diabetes and obesity.


Asunto(s)
Diabetes Mellitus/prevención & control , Hemo-Oxigenasa 1/genética , Obesidad/prevención & control , Adiponectina/sangre , Animales , Citocinas/sangre , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Mejoramiento Genético , Hemo-Oxigenasa 1/biosíntesis , Humanos , Obesidad/enzimología , Obesidad/genética , Estrés Oxidativo
9.
Kidney Int ; 72(8): 945-53, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17667987

RESUMEN

Radiocontrast agents are thought to induce acute kidney injury in part through increased production of reactive oxygen species and increased cellular apoptosis. In this study we determined whether heme oxygenase-1 could prevent or reduce radiocontrast-induced acute kidney injury and, if so, what were the mechanisms by which this can occur. Sodium iothalamate was administered to uninephrectomized, salt-depleted male Sabra rats to initiate acute kidney injury. Heme oxygenase-1 was induced with cobalt protoporphyrin or inhibited with stannous mesoporphyrin. Inhibition of heme oxygenase exacerbated kidney injury as measured by an increase in plasma creatinine and in superoxide production. Heme oxygenase-1 induction prevented the increase in plasma creatinine and in superoxide in both the cortex and medulla compared to untreated rats with acute kidney injury. This protective effect of heme oxygenase-1 was associated with increased anti-apoptotic proteins Bcl-2 and Bcl-xl and a decrease of pro-apoptotic caspase-3 and caspase-9 along with increased expression of inactive BAX. Our study suggests that increased levels of heme oxygenase-1 are protective against acute kidney injury due to radiocontrast exposure.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Medios de Contraste/efectos adversos , Hemo-Oxigenasa 1/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo , Lesión Renal Aguda/metabolismo , Animales , Bilirrubina/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Creatinina/sangre , Hemo/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Humanos , Masculino , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Ratas Wistar , Superóxidos/metabolismo , Proteína X Asociada a bcl-2/metabolismo
10.
Cell Mol Biol (Noisy-le-grand) ; 53(4): 51-60, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17531161

RESUMEN

The goal of this study was to characterize the impact of induction or inhibition of the heme-HO system on renal apoptosis in clipped and non-clipped kidneys from 2K1C hypertensive rats. Male Sprague-Dawley rats had a 0.25 mm silver clip placed around the left renal artery. Four groups of rats were studied: sham operated animals, 2K1C control rats, 2K1C rats received weekly injections of CoPP (5 mg/100 g body wt, administered subcutaneously), and 2K1C rats pretreated with SnMP (5 mg/ 100g body wt, administered intraperitoneally three times a week). The animals were sacrificed three weeks after surgery. We measured systolic blood pressure, plasma renin activity, non-clipped and clipped kidney HO-1 and HO-2 protein expression, HO activity, heme content, nitrotyrosine levels, and activation of selected pro- and anti-apoptotic proteins. Systolic blood pressure and plasma renin activity were significantly higher in 2K1C rats compared to sham rats. Compared to kidneys from sham animals, clipped kidneys from 2K1C rats showed a significant increase in HO-1 expression with increases in HO activity (26%), heme content (47%) and nitrotyrosine levels (49%), accompanied by an increase in caspase-3 and caspase-9 activity. In contrast, non-clipped kidneys from 2K1C rats showed no differences in HO-1 expression, HO activity, heme content, nitrotyrosine levels and caspase activity compared to sham rats. In clipped kidneys from 2K1C rats, inhibition of HO activity by SnMP augmented caspase-3 and caspase-9 activity and decreased expression of the anti-apoptotic Bcl-2 protein, while induction of HO-1 with CoPP strongly inhibited the activity of both caspases and increased the induction of Bcl-2 and Bcl-xl proteins. These findings demonstrate that the clipped kidneys responded to decreased renal perfusion pressure and increased oxidative stress by activation of the heme-HO system, which exerts antiapoptotic action via mechanisms involving decreased caspase-3 and caspase-9 activity, and increased expression of antiapoptotic molecules.


Asunto(s)
Apoptosis/fisiología , Hemo-Oxigenasa 1/genética , Hipertensión Renovascular/genética , Animales , Caspasas/metabolismo , Regulación de la Expresión Génica , Hemo/análisis , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hipertensión Renovascular/enzimología , Hipertensión Renovascular/etiología , Hipertensión Renovascular/patología , Riñón/química , Riñón/enzimología , Riñón/patología , Riñón/cirugía , Masculino , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Tirosina/análogos & derivados , Tirosina/análisis , Proteína bcl-X/metabolismo
11.
Ann N Y Acad Sci ; 1113: 256-75, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17513461

RESUMEN

This article summarizes some aspects of stress in the metabolic syndrome at the psychosocial, tissue, and cellular levels. The metabolic syndrome is a valuable research concept for studying population health and social-biological translation. The cluster of cardiovascular risk factors labeled the metabolic syndrome is linked with low socioeconomic status. Systematic differences in diet and physical activity contribute to social patterning of the syndrome. In addition, psychosocial factors including chronic work stress are linked with its development. Psychosocial factors could lead to metabolic perturbations and increase cardiovascular risk via activation of neuroendocrine responses, for example, in the autonomic nervous system and in several hormonal pathways. High glucocorticoid levels will promote lipid storage in visceral rather than subcutaneous adipose tissue. Adipocytes secrete several proinflammatory cytokines, which considered major contributors to increase in oxidants and cell injury. Upregulation of heme oxygenase 1 (HO-1) and peroxidase in the early development of diabetes produces a decrease in oxidative-mediated injury. Increased HO activity is associated with a significant decrease in superoxide, endothelial cell shedding and blood pressure. Finally, it is proposed that overexpression of glutathione peroxidase in beta cells may protect beta cell deterioration from oxidative stress during development of diabetes and hyperglycemia and this may result in attenuation of beta cell failure. If this proves to be the case, then the scene will be set to develop glutathione peroxidase mimetics for use in preclinical and clinical trials.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Síndrome Metabólico/metabolismo , Síndrome Metabólico/psicología , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/fisiopatología , Animales , Diabetes Mellitus Tipo 2/genética , Humanos , Síndrome Metabólico/fisiopatología , Estrés Oxidativo/fisiología , Factores de Riesgo
12.
J Cell Biochem ; 100(4): 815-23, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17131376

RESUMEN

The contribution of heme oxygenase HO-2, the primary source of bilirubin and carbon monoxide (CO) under physiological conditions, to the regulation of vascular function has remained largely unexplored. Using siRNA HO-2, we examined the effect of suppressed levels of HO-2 on vascular antioxidant and survival proteins. In vivo HO-2 siRNA treatment decreased the basal levels of EC-SOD, pAKT proteins (serine-473 and threonine-308), without changing Akt protein expression. HO-2 siRNA treatment increased 3-nitrotyrosine (3-NT) and apoptotic signaling kinase-1 (ASK-1) (P < 0.01). HO activity was decreased by the use of siRNA HO-2. We extended these studies to the mitochondria, examining for the presence of HO-1 and its role in the regulation of pro- and anti-apoptotic proteins. HO activity was increased by the administration of CoPP resulting in the translocation of HO-1 into the mitochondria, mainly to the inner face of the mitochondrial inner membrane. These findings suggest that HO-2 is critical in the maintenance of heme homeostasis and also the regulation of apoptosis by controlling levels of EC-SOD, Akt, 3-NT, and ASK-1. In addition, localization of HO-1 in the mitochondrial compartment plays a critical role in mitochondria-mediated apoptosis.


Asunto(s)
Hemo Oxigenasa (Desciclizante)/genética , Mitocondrias/metabolismo , Interferencia de ARN , Superóxido Dismutasa/metabolismo , Animales , Apoptosis/fisiología , Western Blotting , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo Oxigenasa (Desciclizante)/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Transducción de Señal/fisiología , Tirosina/análogos & derivados , Tirosina/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
13.
Int J Hematol ; 86(5): 438-45, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18192113

RESUMEN

Curative therapy for diabetes mellitus mainly involves pancreas or islet transplantation to recruit insulin-producing cells. This approach is limited, however, because of both the shortage of donor organs and allograft rejection. Intra-bone marrow bone marrow transplantation (IBM-BMT) has recently been shown to be effective in inducing donor-specific tolerance in mice and rats without the use of immunosuppressants. After induction of diabetes in 15 C3H mice with streptozotocin, the mice received both allotransplants of bone marrow cells from C57BL/6 mice by IBM-BMT and injections via the portal vein of insulin-producing cells that were induced in vitro from stem cells derived from adult C57BL/6 bone marrow. We evaluated the expression of these cells by examining the expression of not only insulin but also the crucial transcription factors insulin I and insulin II. The diabetic mice were treated with IBM-BMT and precultured insulin-producing cells. Hyperglycemia was normalized by 5 days after the treatment and remained normal for more than 45 days. This strategy might be applicable to patients with type I diabetes mellitus.


Asunto(s)
Células de la Médula Ósea , Trasplante de Médula Ósea , Diabetes Mellitus Experimental/terapia , Supervivencia de Injerto , Células Secretoras de Insulina/trasplante , Animales , Células de la Médula Ósea/metabolismo , Diabetes Mellitus Experimental/metabolismo , Regulación de la Expresión Génica , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos C3H , Vena Porta , Trasplante Homólogo
14.
Cell Mol Biol (Noisy-le-grand) ; 51(4): 363-70, 2005 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-16309586

RESUMEN

Heme oxygenase (HO-1) has been implicated as an anti-inflammatory gene. HO-1 overexpression, transiently and chronically, affects heme protein expression, attenuates TNF-mediated cell death, and decreases adhesion molecules. We assessed the effect of oxidant-mediated agents such as glucose and heme on 8-epi-isoprostane PGF2alpha (8-epi-PGF2alpha) and monocyte chemoattractant protein-1 (MCP-1). Glucose and heme increased both 8-epi-PGF2alpha and MCP-1. Overexpression of HO-1 decreased both 8-epi-PGF2alpha and MCP-1. To identify target genes involved in HO-1-mediated regulation of inflammation, a serial analysis of gene expression mRNA profile was performed in endothelial cells (EC) overexpressing the human HO-1 gene by transduction of a retrovirus carrying the HO-1 gene. Gene arrays (differential displays among 2400 genes) were used to identify known and novel differentially expressed genes. The levels of expression for several genes were confirmed by real time PCR in cells overexpressing the HO-1 gene. In HO-1 overexpressing cells, VEGF and the prostaglandin transporter were greatly increased while MCP-1 levels were decreased by 2.5-fold. The data from this study are relevant to understanding the mechanisms underlying the pathophysiological effects of HO-1 deficiency on endothelial cell injury and inflammation.


Asunto(s)
Ciclo Celular/genética , Quimiocina CCL2/metabolismo , Regulación hacia Abajo , Células Endoteliales/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Regulación Enzimológica de la Expresión Génica , Glucosa/farmacología , Hemo/farmacología , Humanos , Inflamación/genética , Regulación hacia Arriba/genética
15.
Cell Mol Biol (Noisy-le-grand) ; 51(4): 371-6, 2005 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-16309587

RESUMEN

Hyperglycemia represents the main cause of complication of diabetes mellitus and oxidative stress, resulting from increased generation of reactive oxygen species (ROS), and plays a crucial role in their pathogenesis. Impairment of vascular responses in diabetic rats, as a result of an increase in superoxide (O2-), formation is a major complication in diabetes. Since heme oxygenase (HO) expression regulates the level of ROS by increasing antioxidant, such as glutathione and bilirubin, we investigated whether upregulation of HO-1 modulates the levels of iNOS and eNOS and altered vascular responses to phenylephrine (PE) and acetylcholine (Ach) in aorta and femoral arteries of diabetic (streptozotocin (STZ)-induced) rats. Our results showed that iNOS expression was increased, but HO activity was reduced, in diabetic compared to nondiabetic rats (p<0.05). Upregulation of HO-1 expression by cobalt protoporphyrin (CoPP), an inducer of HO-1 protein and activity, conferred an increase in eNOS and differentially decreased iNOS protein levels (p<0.05). Isolated aortic and femoral arteries obtained from diabetic rats exhibited contraction to PE and relaxation to Ach, which were markedly increased and decreased, respectively. However, HO-1 induction in diabetic rats normalized relaxation compared to controls. Therefore, overexpression of HO-1 may mediate an increase in eNOS and a decrease in iNOS, potentially contributing to restoration of vascular responses in diabetic rats.


Asunto(s)
Diabetes Mellitus/enzimología , Angiopatías Diabéticas/enzimología , Angiopatías Diabéticas/fisiopatología , Regulación Enzimológica de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Vasodilatación/genética , Acetilcolina/farmacología , Animales , Diabetes Mellitus/inducido químicamente , Diabetes Mellitus/genética , Angiopatías Diabéticas/genética , Células Endoteliales/enzimología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/genética , Fenilefrina/farmacología , Ratas , Ratas Sprague-Dawley , Estreptozocina/farmacología
16.
Diabetologia ; 48(2): 345-50, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15660261

RESUMEN

AIMS/HYPOTHESIS: Patients with diabetes mellitus are well known to be at high risk for vascular disease. Circulating endothelial cells (CECs) have been reported to be an ex vivo indicator of vascular injury. We investigated the presence of CECs in the peripheral blood of 25 patients with diabetes mellitus and in nine non-diabetic control donors. METHODS: Endothelial cells were isolated from peripheral blood with anti-CD-146-coated immunomagnetic Dynabeads, and were stained with acridine orange dye and counted by fluorescence microscopy. The cells were also stained for von Willebrand factor and Ulex europaeus lectin 1. RESULTS: Patients with diabetes mellitus had an elevated number of CECs (mean 69+/-30 cells/ml, range 35-126) compared with healthy controls (mean 10+/-5 cells/ml, range 3-18) (p<0.001). The increase in CECs did not correlate with the levels of HbA(1)c. Circulating endothelial cell numbers were elevated regardless of glucose levels, suggesting that, even with control of glucose levels, there is increased endothelial cell sloughing. CONCLUSIONS: Our study suggests that the higher number of CECs in patients with type 2 diabetes may reflect ongoing vascular injury that is not directly dependent on glucose control.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Endotelio Vascular/patología , Hemoglobina Glucada/metabolismo , Adulto , Anciano , Envejecimiento , Antígenos CD/sangre , Índice de Masa Corporal , Antígeno CD146 , Diabetes Mellitus Tipo 2/patología , Endotelio Vascular/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Moléculas de Adhesión de Célula Nerviosa/sangre , Valores de Referencia , Análisis de Regresión , Venas Umbilicales/fisiología
17.
J Cell Biochem ; 90(6): 1098-111, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14635185

RESUMEN

The purpose of the present study was to examine the role of human heme oxygenase (human HO-1) in cell cycle progression following exposure to heme or human HO-1 gene transfer and to identify target genes associated with human HO-1-meditated increases in cell cycle progression using cDNA microarray technology. Heme-induced robust human HO-1 expression in quiescent human microvessel endothelial cells cultured in 1% FBS and the levels of human HO-1 expression progressively declined without a change in the cell cyclin. To identify genes regulated by human HO-1 in the cell cycle, human endothelial cells were transduced with a retroviral vector encoded with human HO-1 gene or an empty vector. Transgene expression and functionality of the recombinant protein were assessed by Western blotting, enzyme activity, carbon monoxide, cGMP production, and cell cycle analysis. Human cDNA gene array and quantitative real-time RT-PCR were used to identify both known and novel differentially expressed genes in cells overexpressing human HO-1. Major findings were upregulation of several genes associated with cell cycle progression, including cyclin E and D; downregulation of cyclin-dependent kinase inhibitors p21 and p27, cyclin-dependent kinases 2, 5, and 6, and monocyte chemoattractant protein-1; and upregulation of growth factors, including vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor I (VEGFRI), endothelial growth factor (EGF) and hepatic-derived growth factor (HDGF). These findings identify an array of gene responses to overexpression of human HO-1 and elucidate new aspects of human HO-1 signaling involved in cell growth.


Asunto(s)
Ciclo Celular/fisiología , Células Endoteliales/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hemo Oxigenasa (Desciclizante)/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , GMP Cíclico/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Ciclinas/metabolismo , Dinoprostona/metabolismo , Células Endoteliales/citología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Hemo-Oxigenasa 1 , Humanos , Proteínas de la Membrana , Neovascularización Fisiológica , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción Genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Thromb Res ; 110(5-6): 305-9, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-14592553

RESUMEN

Heme oxygenase (HO) regulates cellular heme levels and catalyzes the formation of bilirubin and carbon monoxide (CO). We hypothesized that the status of the endothelial HO system influences the angiotensin (Ang) II-induced increase in the endothelial cell (EC) production of PGE2, eicosanoids which modulate the vascular actions of Ang II. In this study, we investigated the effect of interventions that suppress HO activity or induce HO-1 gene expression on Ang II-mediated increase in PGE2 in cultures of human microvessel endothelial cells (EC). Incubation of EC with Ang II (100 ng/ml) for 24 h increased the levels of PGE2 in the culture media. This effect of Ang II on prostaglandin production by EC was attenuated in cells treated with heme, but was magnified in cells treated with the HO inhibitor, Stannis mesoporphyrin (SnMP). Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene attenuated heme and Ang II-induced prostaglandin synthesis. We also investigated the physiological significance of human HO-1 overexpression on attenuation of Ang II-mediated oxidative stress. Decreases in COMET levels were found in EC transduced with the HO-1 gene. These results indicate that overexpression of the HO system in EC exerts an inhibitory influence on Ang II-induced synthesis of prostaglandins and attenuates DNA damage caused by exposure to Ang II.


Asunto(s)
Angiotensina II/farmacología , Daño del ADN/fisiología , Células Endoteliales/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Células Cultivadas , Daño del ADN/efectos de los fármacos , Activación Enzimática , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1 , Humanos , Isoprostanos , Proteínas de la Membrana
19.
Biochem Biophys Res Commun ; 308(4): 1001-8, 2003 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12927819

RESUMEN

Heme oxygenase-1 (HO-1) is a stress protein, which has been suggested to participate in defense mechanisms against agents that may induce oxidative injury, such as angiotensin II (Ang II). The purpose of the present study was to examine the role of human HO-1 in cell-cycle progression. We investigated the effect of Ang II on HO-1 gene expression in serum-deprived media to drive human endothelial cells into G(0)/G(1) (1% FBS) compared to exponentially grown cells (10% FBS). The addition of Ang II (100 ng/ml) to endothelial cells increased HO-1 protein and activity in G(0)/G(1) in a time-dependent manner, reaching a maximum HO-1 level at 16 h. Real-time RT-PCR demonstrated that Ang II increased the levels of HO-1 mRNA in G(0)/G(1) as early as 1 h. The rate of HO-1 induction in response to Ang II was several-fold higher in serum-starved cells compared to cells cultured in continuous 10% FBS. The addition of Ang II increased the generation of 8-epi-isoprostane PGF(2 alpha). Inhibition of HO-1, by Stannis mesoporphyrin (SnMP), potentiated Ang II-mediated DNA damage and generation of 8-epi-isoprostane PGF(2 alpha). These results imply that expression of HO-1 in G(0)/G(1), in the presence of Ang II, may be a key player in attenuating DNA damage during cell-cycle progression. Thus, exposure of endothelial cells to Ang II causes a complex response involving generation of superoxide anion, which may be involved in DNA damage. Upregulation of HO-1 ensures the generation of bilirubin and carbon monoxide (CO) in G(0)/G(1) phase to counteract Ang II-mediated oxidative DNA damage. Inducibility of HO-1 in G(0)/G(1) phase is essential and probably regulated by a complex system involving oxygen species to assure controlled cell growth.


Asunto(s)
Ciclo Celular , Dinoprost/análogos & derivados , Hemo Oxigenasa (Desciclizante)/biosíntesis , Aniones , Bilirrubina/metabolismo , Western Blotting , Monóxido de Carbono/metabolismo , División Celular , Células Cultivadas , Medio de Cultivo Libre de Suero/farmacología , Daño del ADN , Endotelio Vascular/metabolismo , F2-Isoprostanos/biosíntesis , Fase G1 , Hemo-Oxigenasa 1 , Humanos , Proteínas de la Membrana , Mesoporfirinas/farmacología , Microscopía Confocal , Estrés Oxidativo , Oxígeno/metabolismo , Fase de Descanso del Ciclo Celular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxidos/metabolismo , Temperatura , Factores de Tiempo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA