Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adipocyte ; 11(1): 366-378, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35734881

RESUMEN

High fat diet (HFD)-induced obesity leads to perturbation in the storage function of white adipose tissue (WAT) resulting in deposition of lipids in tissues ill-equipped to deal with this challenge. The role of insulin like growth factor-1 (IGF-1) in the systemic and organ-specific responses to HFD is unclear. Using cixutumumab, a monoclonal antibody that internalizes and degrades cell surface IGF-1 receptors (IGF-1 R), leaving insulin receptor expression unchanged we aimed to establish the role of IGF-1 R in the response to a HFD. Mice treated with cixutumumab fed standard chow developed mild hyperinsulinemia with no change in WAT. When challenged by HFD mice treated with cixutumumab had reduced weight gain, reduced WAT expansion, and reduced hepatic lipid vacuole formation. In HFD-fed mice, cixutumumab led to reduced levels of genes encoding proteins important in fatty acid metabolism in WAT and liver. Cixutumumab protected against blunting of insulin-stimulated phosphorylation of Akt in liver of HFD fed mice. These data reveal an important role for IGF-1 R in the WAT and hepatic response to short-term nutrient excess. IGF-1 R inhibition during HFD leads to a lipodystrophic phenotype with a failure of WAT lipid storage and protection from HFD-induced hepatic insulin resistance.


Asunto(s)
Resistencia a la Insulina , Receptor IGF Tipo 1 , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Anticuerpos Monoclonales Humanizados , Dieta Alta en Grasa/efectos adversos , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lípidos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores
3.
Endocrinology ; 159(8): 2917-2925, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29796592

RESUMEN

Reduced systemic insulin signaling promotes endothelial dysfunction and diminished endogenous vascular repair. We investigated whether restoration of endothelial insulin receptor expression could rescue this phenotype. Insulin receptor knockout (IRKO) mice were crossed with mice expressing a human insulin receptor endothelial cell-specific overexpression (hIRECO) to produce IRKO-hIRECO progeny. No metabolic differences were noted between IRKO and IRKO-hIRECO mice in glucose and insulin tolerance tests. In contrast with control IRKO littermates, IRKO-hIRECO mice exhibited normal blood pressure and aortic vasodilatation in response to acetylcholine, comparable to parameters noted in wild type littermates. These phenotypic changes were associated with increased basal- and insulin-stimulated nitric oxide production. IRKO-hIRECO mice also demonstrated normalized endothelial repair after denuding arterial injury, which was associated with rescued endothelial cell migration in vitro but not with changes in circulating progenitor populations or culture-derived myeloid angiogenic cells. These data show that restoration of endothelial insulin receptor expression alone is sufficient to prevent the vascular dysfunction caused by systemically reduced insulin signaling.


Asunto(s)
Aorta/metabolismo , Glucemia/metabolismo , Endotelio Vascular/metabolismo , Haploinsuficiencia/genética , Receptor de Insulina/genética , Vasodilatación/genética , Acetilcolina/farmacología , Animales , Antígenos CD/genética , Aorta/fisiopatología , Presión Sanguínea , Movimiento Celular , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Prueba de Tolerancia a la Glucosa , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
4.
Diabetes ; 66(11): 2808-2821, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28830894

RESUMEN

Shc homology 2-containing inositol 5' phosphatase-2 (SHIP2) is a lipid phosphatase that inhibits insulin signaling downstream of phosphatidylinositol 3-kinase (PI3K); its role in vascular function is poorly understood. To examine its role in endothelial cell (EC) biology, we generated mice with catalytic inactivation of one SHIP2 allele selectively in ECs (ECSHIP2Δ/+). Hyperinsulinemic-euglycemic clamping studies revealed that ECSHIP2Δ/+ was resistant to insulin-stimulated glucose uptake in adipose tissue and skeletal muscle compared with littermate controls. ECs from ECSHIP2Δ/+ mice had increased basal expression and activation of PI3K downstream targets, including Akt and endothelial nitric oxide synthase, although incremental activation by insulin and shear stress was impaired. Insulin-mediated vasodilation was blunted in ECSHIP2Δ/+ mice, as was aortic nitric oxide bioavailability. Acetylcholine-induced vasodilation was also impaired in ECSHIP2Δ/+ mice, which was exaggerated in the presence of a superoxide dismutase/catalase mimetic. Superoxide abundance was elevated in ECSHIP2Δ/+ ECs and was suppressed by PI3K and NADPH oxidase 2 inhibitors. These findings were phenocopied in healthy human ECs after SHIP2 silencing. Our data suggest that endothelial SHIP2 is required to maintain normal systemic glucose homeostasis and prevent oxidative stress-induced endothelial dysfunction.


Asunto(s)
Endotelio Vascular/metabolismo , Resistencia a la Insulina/fisiología , NADPH Oxidasa 2/metabolismo , Estrés Oxidativo/fisiología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Animales , Aorta , Células Cultivadas , Células Endoteliales , Regulación de la Expresión Génica/fisiología , Técnica de Clampeo de la Glucosa , Intolerancia a la Glucosa , Ratones , Ratones Noqueados , NADPH Oxidasa 2/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Vasoconstricción/fisiología
5.
Diabetes ; 66(2): 287-299, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28108607

RESUMEN

Low circulating levels of insulin-like growth factor binding protein 1 (IGFBP-1) are associated with insulin resistance and predict the development of type 2 diabetes. IGFBP-1 can affect cellular functions independently of IGF binding through an Arg-Gly-Asp (RGD) integrin-binding motif. Whether causal mechanisms underlie the favorable association of high IGFBP-1 levels with insulin sensitivity and whether these could be exploited therapeutically remain unexplored. We used recombinant IGFBP-1 and a synthetic RGD-containing hexapeptide in complementary in vitro signaling assays and in vivo metabolic profiling in obese mice to investigate the effects of IGFBP-1 and its RGD domain on insulin sensitivity, insulin secretion, and whole-body glucose regulation. The RGD integrin-binding domain of IGFBP-1, through integrin engagement, focal adhesion kinase, and integrin-linked kinase, enhanced insulin sensitivity and insulin secretion in C2C12 myotubes and INS-1 832/13 pancreatic ß-cells. Both acute administration and chronic infusion of an RGD synthetic peptide to obese C57BL/6 mice improved glucose clearance and insulin sensitivity. These favorable effects on metabolic homeostasis suggest that the RGD integrin-binding domain of IGFBP-1 may be a promising candidate for therapeutic development in the field of insulin resistance.


Asunto(s)
Glucemia/efectos de los fármacos , Resistencia a la Insulina , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Proteínas Recombinantes/farmacología , Animales , Glucemia/metabolismo , Línea Celular , Proliferación Celular , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Immunoblotting , Técnicas In Vitro , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Espectrometría de Masas , Ratones , Ratones Obesos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
6.
Circ Res ; 120(5): 784-798, 2017 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-27920123

RESUMEN

RATIONALE: In the endothelium, insulin stimulates endothelial NO synthase (eNOS) to generate the antiatherosclerotic signaling radical NO. Insulin-resistant type 2 diabetes mellitus is associated with reduced NO availability and accelerated atherosclerosis. The effect of enhancing endothelial insulin sensitivity on NO availability is unclear. OBJECTIVE: To answer this question, we generated a mouse with endothelial cell (EC)-specific overexpression of the human insulin receptor (hIRECO) using the Tie2 promoter-enhancer. METHODS AND RESULTS: hIRECO demonstrated significant endothelial dysfunction measured by blunted endothelium-dependent vasorelaxation to acetylcholine, which was normalized by a specific Nox2 NADPH oxidase inhibitor. Insulin-stimulated phosphorylation of protein kinase B was increased in hIRECO EC as was Nox2 NADPH oxidase-dependent generation of superoxide, whereas insulin-stimulated and shear stress-stimulated eNOS activations were blunted. Phosphorylation at the inhibitory residue Y657 of eNOS and expression of proline-rich tyrosine kinase 2 that phosphorylates this residue were significantly higher in hIRECO EC. Inhibition of proline-rich tyrosine kinase 2 improved insulin-induced and shear stress-induced eNOS activation in hIRECO EC. CONCLUSIONS: Enhancing insulin sensitivity specifically in EC leads to a paradoxical decline in endothelial function, mediated by increased tyrosine phosphorylation of eNOS and excess Nox2-derived superoxide. Increased EC insulin sensitivity leads to a proatherosclerotic imbalance between NO and superoxide. Inhibition of proline-rich tyrosine kinase 2 restores insulin-induced and shear stress-induced NO production. This study demonstrates for the first time that increased endothelial insulin sensitivity leads to a proatherosclerotic imbalance between NO and superoxide.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Resistencia a la Insulina/fisiología , Transducción de Señal/fisiología , Animales , Aterosclerosis/patología , Células Cultivadas , Células Endoteliales/patología , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos
7.
Nature ; 515(7526): 279-282, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25119035

RESUMEN

The mechanisms by which physical forces regulate endothelial cells to determine the complexities of vascular structure and function are enigmatic. Studies of sensory neurons have suggested Piezo proteins as subunits of Ca(2+)-permeable non-selective cationic channels for detection of noxious mechanical impact. Here we show Piezo1 (Fam38a) channels as sensors of frictional force (shear stress) and determinants of vascular structure in both development and adult physiology. Global or endothelial-specific disruption of mouse Piezo1 profoundly disturbed the developing vasculature and was embryonic lethal within days of the heart beating. Haploinsufficiency was not lethal but endothelial abnormality was detected in mature vessels. The importance of Piezo1 channels as sensors of blood flow was shown by Piezo1 dependence of shear-stress-evoked ionic current and calcium influx in endothelial cells and the ability of exogenous Piezo1 to confer sensitivity to shear stress on otherwise resistant cells. Downstream of this calcium influx there was protease activation and spatial reorganization of endothelial cells to the polarity of the applied force. The data suggest that Piezo1 channels function as pivotal integrators in vascular biology.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/fisiología , Fricción , Canales Iónicos/metabolismo , Estrés Mecánico , Animales , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/metabolismo , Femenino , Hemorreología , Masculino , Ratones
8.
Arterioscler Thromb Vasc Biol ; 34(9): 2051-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25012136

RESUMEN

OBJECTIVES: Defective endothelial regeneration predisposes to adverse arterial remodeling and is thought to contribute to cardiovascular disease in type 2 diabetes mellitus. We recently demonstrated that the type 1 insulin-like growth factor receptor (IGF1R) is a negative regulator of insulin sensitivity and nitric oxide bioavailability. In this report, we examined partial deletion of the IGF1R as a potential strategy to enhance endothelial repair. APPROACH AND RESULTS: We assessed endothelial regeneration after wire injury in mice and abundance and function of angiogenic progenitor cells in mice with haploinsufficiency of the IGF1R (IGF1R(+/-)). Endothelial regeneration after arterial injury was accelerated in IGF1R(+/-) mice. Although the yield of angiogenic progenitor cells was lower in IGF1R(+/-) mice, these angiogenic progenitor cells displayed enhanced adhesion, increased secretion of insulin-like growth factor-1, and enhanced angiogenic capacity. To examine the relevance of IGF1R manipulation to cell-based therapy, we transfused IGF1R(+/-) bone marrow-derived CD117(+) cells into wild-type mice. IGF1R(+/-) cells accelerated endothelial regeneration after arterial injury compared with wild-type cells and did not alter atherosclerotic lesion formation. CONCLUSIONS: Haploinsufficiency of the IGF1R is associated with accelerated endothelial regeneration in vivo and enhanced tube forming and adhesive potential of angiogenic progenitor cells in vitro. Partial deletion of IGF1R in transfused bone marrow-derived CD117(+) cells enhanced their capacity to promote endothelial regeneration without altering atherosclerosis. Our data suggest that manipulation of the IGF1R could be exploited as novel therapeutic approach to enhance repair of the arterial wall after injury.


Asunto(s)
Enfermedades de las Arterias Carótidas/prevención & control , Endotelio Vascular/fisiología , Arteria Femoral/lesiones , Células Madre Hematopoyéticas/fisiología , Neovascularización Fisiológica/fisiología , Receptor IGF Tipo 1/fisiología , Animales , Aorta Torácica/patología , Apolipoproteínas E/deficiencia , Enfermedades de las Arterias Carótidas/etiología , Enfermedades de las Arterias Carótidas/genética , Adhesión Celular , Endotelio Vascular/metabolismo , Femenino , Regulación de la Expresión Génica , Genotipo , Trasplante de Células Madre Hematopoyéticas , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenotipo , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética , Regeneración
9.
Stem Cells ; 32(10): 2714-23, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24916783

RESUMEN

Recent data suggest reduced indices of vascular repair in South Asian men, a group at increased risk of cardiovascular events. Outgrowth endothelial cells (OEC) represent an attractive tool to study vascular repair in humans and may offer potential in cell-based repair therapies. We aimed to define and manipulate potential mechanisms of impaired vascular repair in South Asian (SA) men. In vitro and in vivo assays of vascular repair and angiogenesis were performed using OEC derived from SA men and matched European controls, prior defining potentially causal molecular mechanisms. SA OEC exhibited impaired colony formation, migration, and in vitro angiogenesis, associated with decreased expression of the proangiogenic molecules Akt1 and endothelial nitric oxide synthase (eNOS). Transfusion of European OEC into immunodeficient mice after wire-induced femoral artery injury augmented re-endothelialization, in contrast with SA OEC and vehicle; SA OEC also failed to promote angiogenesis after induction of hind limb ischemia. Expression of constitutively active Akt1 (E17KAkt), but not green fluorescent protein control, in SA OEC increased in vitro angiogenesis, which was abrogated by a NOS antagonist. Moreover, E17KAkt expressing SA OEC promoted re-endothelialization of wire-injured femoral arteries, and perfusion recovery of ischemic limbs, to a magnitude comparable with nonmanipulated European OEC. Silencing Akt1 in European OEC recapitulated the functional deficits noted in SA OEC. Reduced signaling via the Akt/eNOS axis is causally linked with impaired OEC-mediated vascular repair in South Asian men. These data prove the principle of rescuing marked reparative dysfunction in OEC derived from these men.


Asunto(s)
Vasos Sanguíneos/patología , Células Endoteliales/citología , Células Endoteliales/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Cicatrización de Heridas , Adulto , Animales , Asia , Demografía , Células Endoteliales/efectos de los fármacos , Silenciador del Gen , Humanos , Insulina/farmacología , Masculino , Ratones Desnudos , Fosforilación/efectos de los fármacos , Factores de Riesgo , Población Blanca , Cicatrización de Heridas/efectos de los fármacos
10.
Atherosclerosis ; 230(1): 131-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23958265

RESUMEN

OBJECTIVE: Systemic insulin resistance is associated with a portfolio of risk factors for atherosclerosis development. We sought to determine whether insulin resistance specifically at the level of the endothelium promotes atherosclerosis and to examine the potential involvement of reactive oxygen species. METHODS: We cross-bred mice expressing a dominant negative mutant human insulin receptor specifically in the endothelium (ESMIRO) with ApoE(-/-) mice to examine the effect of endothelium-specific insulin resistance on atherosclerosis. RESULTS: ApoE(-/-)/ESMIRO mice had similar blood pressure, plasma lipids and whole-body glucose tolerance, but blunted endothelial insulin signalling, in comparison to ApoE(-/-) mice. Atherosclerosis was significantly increased in ApoE(-/-)/ESMIRO mice at the aortic sinus (226 ± 16 versus 149 ± 24 × 10(3) µm(2), P = 0.01) and lesser curvature of the aortic arch (12.4 ± 1.2% versus 9.4 ± 0.9%, P = 0.035). Relaxation to acetylcholine was blunted in aorta from ApoE(-/-)/ESMIRO mice (Emax 65 ± 41% versus 103 ± 6%, P = 0.02) and was restored by the superoxide dismutase mimetic MnTMPyP (Emax 112 ± 15% versus 65 ± 41%, P = 0.048). Basal generation of superoxide was increased 1.55 fold (P = 0.01) in endothelial cells from ApoE(-/-)/ESMIRO mice and was inhibited by the NADPH oxidase inhibitor gp91ds-tat (-12 ± 0.04%, P = 0.04), the NO synthase inhibitor L-NMMA (-8 ± 0.02%, P = 0.001) and the mitochondrial specific inhibitor rotenone (-23 ± 0.04%, P = 0.006). CONCLUSIONS: Insulin resistance specifically at the level of the endothelium leads to acceleration of atherosclerosis in areas with disturbed flow patterns such as the aortic sinus and the lesser curvature of the aorta. We have identified a potential role for increased generation of reactive oxygen species from multiple enzymatic sources in promoting atherosclerosis in this setting.


Asunto(s)
Aterosclerosis/fisiopatología , Endotelio Vascular/patología , Resistencia a la Insulina , Especies Reactivas de Oxígeno , Acetilcolina/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Presión Sanguínea , Peso Corporal , Células Endoteliales/citología , Endotelio Vascular/metabolismo , Genes Dominantes , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Receptor de Insulina/genética , Factores de Riesgo
11.
Diabetes ; 62(6): 2130-4, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23349484

RESUMEN

Insulin resistance is characterized by excessive endothelial cell generation of potentially cytotoxic concentrations of reactive oxygen species. We examined the role of NADPH oxidase (Nox) and specifically Nox2 isoform in superoxide generation in two complementary in vivo models of human insulin resistance (endothelial specific and whole body). Using three complementary methods to measure superoxide, we demonstrated higher levels of superoxide in insulin-resistant endothelial cells, which could be pharmacologically inhibited both acutely and chronically, using the Nox inhibitor gp91ds-tat. Similarly, insulin resistance-induced impairment of endothelial-mediated vasorelaxation could also be reversed using gp91ds-tat. siRNA-mediated knockdown of Nox2, which was specifically elevated in insulin-resistant endothelial cells, significantly reduced superoxide levels. Double transgenic mice with endothelial-specific insulin resistance and deletion of Nox2 showed reduced superoxide production and improved vascular function. This study identifies Nox2 as the central molecule in insulin resistance-mediated oxidative stress and vascular dysfunction. It also establishes pharmacological inhibition of Nox2 as a novel therapeutic target in insulin resistance-related vascular disease.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Resistencia a la Insulina/fisiología , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Acetilcolina/farmacología , Animales , Células Cultivadas , Cromatografía Líquida de Alta Presión , Immunoblotting , Resistencia a la Insulina/genética , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , NADPH Oxidasa 2 , NADPH Oxidasas/genética , Reacción en Cadena de la Polimerasa , Vasodilatadores/farmacología
12.
Pflugers Arch ; 465(8): 1065-74, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23338941

RESUMEN

The insulin-like growth factor-1 receptor (IGF-1R), like the insulin receptor (IR), plays a significant role in determining bioavailability of the critical signalling molecule nitric oxide (NO) and hence, modulates endothelial cell function, particularly in response to stimulation with insulin. In particular, the ability of the IGF-1R to form hybrid receptors with the IR appears to be highly significant in determining the sensitivity of the endothelial cell to insulin. This review will examine the structure of the IGF-1R and how this, with particular reference to the ability of the IGF-1R and the IR to form hybrid receptors, may have an effect both on endothelial cell function and the development of cardiovascular disease.


Asunto(s)
Endotelio/metabolismo , Insulina/metabolismo , Óxido Nítrico/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/fisiología , Animales , Disponibilidad Biológica , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Endotelio/fisiología , Humanos
13.
Diabetes ; 61(9): 2359-68, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22733797

RESUMEN

We recently demonstrated that reducing IGF-1 receptor (IGF-1R) numbers in the endothelium enhances nitric oxide (NO) bioavailability and endothelial cell insulin sensitivity. In the present report, we aimed to examine the effect of increasing IGF-1R on endothelial cell function and repair. To examine the effect of increasing IGF-1R in the endothelium, we generated mice overexpressing human IGF-1R in the endothelium (human IGF-1R endothelium-overexpressing mice [hIGFREO]) under direction of the Tie2 promoter enhancer. hIGFREO aorta had reduced basal NO bioavailability (percent constriction to N(G)-monomethyl-l-arginine [mean (SEM) wild type 106% (30%); hIGFREO 48% (10%)]; P < 0.05). Endothelial cells from hIGFREO had reduced insulin-stimulated endothelial NO synthase activation (mean [SEM] wild type 170% [25%], hIGFREO 58% [3%]; P = 0.04) and insulin-stimulated NO release (mean [SEM] wild type 4,500 AU [1,000], hIGFREO 1,500 AU [700]; P < 0.05). hIGFREO mice had enhanced endothelium regeneration after denuding arterial injury (mean [SEM] percent recovered area, wild type 57% [2%], hIGFREO 47% [5%]; P < 0.05) and enhanced endothelial cell migration in vitro. The IGF-1R, although reducing NO bioavailability, enhances in situ endothelium regeneration. Manipulating IGF-1R in the endothelium may be a useful strategy to treat disorders of vascular growth and repair.


Asunto(s)
Células Endoteliales/metabolismo , Endotelio Vascular/fisiología , Receptor IGF Tipo 1/fisiología , Animales , Aorta/fisiología , Glucemia/metabolismo , Presión Sanguínea , Femenino , Homeostasis , Humanos , Masculino , Ratones , Ratones Transgénicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética , Vasoconstricción/efectos de los fármacos
14.
Diabetes ; 61(4): 915-24, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22357965

RESUMEN

Low concentrations of insulin-like growth factor (IGF) binding protein-1 (IGFBP1) are associated with insulin resistance, diabetes, and cardiovascular disease. We investigated whether increasing IGFBP1 levels can prevent the development of these disorders. Metabolic and vascular phenotype were examined in response to human IGFBP1 overexpression in mice with diet-induced obesity, mice heterozygous for deletion of insulin receptors (IR(+/-)), and ApoE(-/-) mice. Direct effects of human (h)IGFBP1 on nitric oxide (NO) generation and cellular signaling were studied in isolated vessels and in human endothelial cells. IGFBP1 circulating levels were markedly suppressed in dietary-induced obese mice. Overexpression of hIGFBP1 in obese mice reduced blood pressure, improved insulin sensitivity, and increased insulin-stimulated NO generation. In nonobese IR(+/-) mice, overexpression of hIGFBP1 reduced blood pressure and improved insulin-stimulated NO generation. hIGFBP1 induced vasodilatation independently of IGF and increased endothelial NO synthase (eNOS) activity in arterial segments ex vivo, while in endothelial cells, hIGFBP1 increased eNOS Ser(1177) phosphorylation via phosphatidylinositol 3-kinase signaling. Finally, in ApoE(-/-) mice, overexpression of hIGFBP1 reduced atherosclerosis. These favorable effects of hIGFBP1 on insulin sensitivity, blood pressure, NO production, and atherosclerosis suggest that increasing IGFBP1 concentration may be a novel approach to prevent cardiovascular disease in the setting of insulin resistance and diabetes.


Asunto(s)
Aterosclerosis/prevención & control , Presión Sanguínea/fisiología , Diabetes Mellitus/metabolismo , Resistencia a la Insulina/fisiología , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Óxido Nítrico/biosíntesis , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Células Cultivadas , Células Endoteliales , Eliminación de Gen , Humanos , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Ratones Transgénicos , Obesidad/metabolismo , Receptor de Insulina/genética
15.
Diabetes ; 60(8): 2169-78, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21677284

RESUMEN

OBJECTIVE: In mice, haploinsufficiency of the IGF-1 receptor (IGF-1R(+/-)), at a whole-body level, increases resistance to inflammation and oxidative stress, but the underlying mechanisms are unclear. We hypothesized that by forming insulin-resistant heterodimers composed of one IGF-1Rαß and one insulin receptor (IR), IRαß complex in endothelial cells (ECs), IGF-1R reduces free IR, which reduces EC insulin sensitivity and generation of the antioxidant/anti-inflammatory signaling radical nitric oxide (NO). RESEARCH DESIGN AND METHODS: Using a number of complementary gene-modified mice with reduced IGF-1R at a whole-body level and specifically in EC, and complementary studies in EC in vitro, we examined the effect of changing IGF-1R/IR stoichiometry on EC insulin sensitivity and NO bioavailability. RESULTS: IGF-1R(+/-) mice had enhanced insulin-mediated glucose lowering. Aortas from these mice were hypocontractile to phenylephrine (PE) and had increased basal NO generation and augmented insulin-mediated NO release from EC. To dissect EC from whole-body effects we generated mice with EC-specific knockdown of IGF-1R. Aortas from these mice were also hypocontractile to PE and had increased basal NO generation. Whole-body and EC deletion of IGF-1R reduced hybrid receptor formation. By reducing IGF-1R in IR-haploinsufficient mice we reduced hybrid formation, restored insulin-mediated vasorelaxation in aorta, and insulin stimulated NO release in EC. Complementary studies in human umbilical vein EC in which IGF-1R was reduced using siRNA confirmed that reducing IGF-1R has favorable effects on NO bioavailability and EC insulin sensitivity. CONCLUSIONS: These data demonstrate that IGF-1R is a critical negative regulator of insulin sensitivity and NO bioavailability in the endothelium.


Asunto(s)
Células Endoteliales/metabolismo , Óxido Nítrico/metabolismo , Receptor IGF Tipo 1/fisiología , Receptor de Insulina/fisiología , Animales , Aorta/efectos de los fármacos , Disponibilidad Biológica , Regulación hacia Abajo , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenilefrina/farmacología , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Transducción de Señal , Venas Umbilicales/citología , Vasoconstricción/efectos de los fármacos
16.
Diabetes ; 60(4): 1295-303, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21317296

RESUMEN

OBJECTIVE: Circulating angiogenic progenitor cells (APCs) participate in endothelial repair after arterial injury. Type 2 diabetes is associated with fewer circulating APCs, APC dysfunction, and impaired endothelial repair. We set out to determine whether insulin resistance adversely affects APCs and endothelial regeneration. RESEARCH DESIGN AND METHODS: We quantified APCs and assessed APC mobilization and function in mice hemizygous for knockout of the insulin receptor (IRKO) and wild-type (WT) littermate controls. Endothelial regeneration after femoral artery wire injury was also quantified after APC transfusion. RESULTS: IRKO mice, although glucose tolerant, had fewer circulating Sca-1(+)/Flk-1(+) APCs than WT mice. Culture of mononuclear cells demonstrated that IRKO mice had fewer APCs in peripheral blood, but not in bone marrow or spleen, suggestive of a mobilization defect. Defective vascular endothelial growth factor-stimulated APC mobilization was confirmed in IRKO mice, consistent with reduced endothelial nitric oxide synthase (eNOS) expression in bone marrow and impaired vascular eNOS activity. Paracrine angiogenic activity of APCs from IRKO mice was impaired compared with those from WT animals. Endothelial regeneration of the femoral artery after denuding wire injury was delayed in IRKO mice compared with WT. Transfusion of mononuclear cells from WT mice normalized the impaired endothelial regeneration in IRKO mice. Transfusion of c-kit(+) bone marrow cells from WT mice also restored endothelial regeneration in IRKO mice. However, transfusion of c-kit(+) cells from IRKO mice was less effective at improving endothelial repair. CONCLUSIONS: Insulin resistance impairs APC function and delays endothelial regeneration after arterial injury. These findings support the hypothesis that insulin resistance per se is sufficient to jeopardize endogenous vascular repair. Defective endothelial repair may be normalized by transfusion of APCs from insulin-sensitive animals but not from insulin-resistant animals.


Asunto(s)
Resistencia a la Insulina/fisiología , Neovascularización Fisiológica/fisiología , Células Madre/metabolismo , Animales , Western Blotting , Adhesión Celular/genética , Adhesión Celular/fisiología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/metabolismo , Citometría de Flujo , Genotipo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Células Madre/citología
17.
Biochim Biophys Acta ; 1801(3): 320-6, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19818873

RESUMEN

The number of people with the insulin-resistant conditions of type 2 diabetes mellitus (T2DM) and obesity has reached epidemic proportions worldwide. Eighty percent of people with T2DM will die from the complications of cardiovascular atherosclerosis. Insulin resistance is characterised by endothelial dysfunction, which is a pivotal step in the initiation/progression of atherosclerosis. A hallmark of endothelial dysfunction is an unfavourable imbalance between the bioavailability of the antiatherosclerotic signalling molecule nitric oxide (NO) and proatherosclerotic reactive oxygen species. In this review we discuss the mechanisms linking insulin resistance to endothelial dysfunction, with a particular emphasis on a potential role for a toxic effect of free fatty acids on endothelial cell homeostasis.


Asunto(s)
Endotelio Vascular/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Resistencia a la Insulina , Enfermedades Vasculares/metabolismo , Animales , Dislipidemias/metabolismo , Humanos , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
18.
Endocrinology ; 150(10): 4575-82, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19608653

RESUMEN

Obesity and type 2 diabetes mellitus are characterized by insulin resistance, reduced bioavailability of the antiatherosclerotic signaling molecule nitric oxide (NO), and accelerated atherosclerosis. IGF-I, the principal growth-stimulating peptide, which shares many of the effects of insulin, may, like insulin, also be involved in metabolic and vascular homeostasis. We examined the effects of IGF-I on NO bioavailability and the effect of obesity/type 2 diabetes mellitus on IGF-I actions at a whole-body level and in the vasculature. In aortic rings IGF-I blunted phenylephrine-mediated vasoconstriction and relaxed rings preconstricted with phenylephrine, an effect blocked by N(G)-monomethyl L-arginine. IGF-I increased NO synthase activity to an extent similar to that seen with insulin and in-vivo IGF-I led to serine phosphorylation of endothelial NO synthase (eNOS). Mice rendered obese using a high-fat diet were less sensitive to the glucose-lowering effects of insulin and IGF-I. IGF-I increased aortic phospho-eNOS levels in lean mice, an effect that was blunted in obese mice. eNOS activity in aortae of lean mice increased 1.6-fold in response to IGF-I compared with obese mice. IGF-I-mediated vasorelaxation was blunted in obese mice. These data demonstrate that IGF-I increases eNOS phosphorylation in-vivo, increases eNOS activity, and leads to NO-dependent relaxation of conduit vessels. Obesity is associated with resistance to IGF-I at a whole-body level and in the endothelium. Vascular IGF-I resistance may represent a novel therapeutic target to prevent or slow the accelerated vasculopathy seen in humans with obesity or type 2 diabetes mellitus.


Asunto(s)
Grasas de la Dieta/efectos adversos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Óxido Nítrico/metabolismo , Obesidad/metabolismo , Vasodilatación , Animales , Aorta/fisiología , Endotelio Vascular/metabolismo , Activación Enzimática , Humanos , Técnicas In Vitro , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/etiología , Obesidad/fisiopatología , Fosforilación , Receptor de Insulina/metabolismo , Serina/metabolismo
19.
J Clin Endocrinol Metab ; 91(12): 5051-6, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16984984

RESUMEN

CONTEXT: Variation in the region of chromosome 8 including the genes steroid 11beta-hydroxylase (CYP11B1) and aldosterone synthase (CYP11B2) influences mineralocorticoid and glucocorticoid metabolism. However, the relative importance of polymorphisms in CYP11B1 and CYP11B2 in determining these phenotypes is unknown. OBJECTIVE: Our objective was to investigate genetic influences of the CYP11B1 and CYP11B2 genes on mineralocorticoid metabolism. DESIGN: We measured 24-h urinary excretion of the key metabolites of the principal mineralocorticoids, glucocorticoids and androgens secreted by the adrenal cortex. We genotyped polymorphisms spanning the CYP11B1 and CYP11B2 genes, which together capture all common variations at the locus. PARTICIPANTS: Participants included 573 members of 105 British Caucasian families ascertained on a hypertensive proband. MAIN OUTCOME MEASURES: We assessed heritability of urinary tetrahydroaldosterone (THAldo) excretion and association of THAldo excretion with genotype. RESULTS: The heritability of THAldo excretion was 52% (P < 10(-6)). There was significant association between THAldo and genotype at several of the CYP11B1/B2 polymorphisms. The strongest association was observed at the rs6387 (2803A/G) polymorphism in intron 3 of CYP11B1 (P = 0.0004). Association followed a codominant model with a 21% higher THAldo excretion per G allele. Genotype at rs6387 accounted for 2.1% of the total population variability of THAldo. We found significant association between THAldo excretion and urinary total androgen excretion, urinary tetrahydrodeoxycortisol level, and urinary cortisol metabolites (all P < 0.001). CONCLUSIONS: Aldosterone synthesis is highly heritable and is affected by genotype at CYP11B1. Our findings support the hypothesis that genetically determined differences in 11-hydroxylation efficiency can have downstream effects on mineralocorticoid synthesis. Such effects may be of relevance to the development of low-renin essential hypertension.


Asunto(s)
Aldosterona/biosíntesis , Variación Genética , Esteroide 11-beta-Hidroxilasa/genética , Aldosterona/análogos & derivados , Aldosterona/metabolismo , Aldosterona/orina , Cortodoxona/análogos & derivados , Cortodoxona/orina , Citocromo P-450 CYP11B2/genética , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Esteroide 11-beta-Hidroxilasa/metabolismo , Esteroides/orina
20.
Stroke ; 36(10): 2215-9, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16179573

RESUMEN

BACKGROUND AND PURPOSE: Studies in unrelated individuals have produced conflicting findings concerning the putative association between the interleukin-6 (IL-6) -174G/C polymorphism and carotid intimal-medial thickness (IMT). We have used a family-based genetic association design to assess the heritability of carotid IMT and to investigate the hypothesized association of carotid IMT with the IL-6 to -174G/C polymorphism. METHODS: We studied 854 members of 224 white British families. The heritability of carotid IMT was determined using Multipoint Engine for Rapid Likelihood Inference. Genetic association analyses were carried out using ANOVA and family-based tests of association implemented in Quantitative Transmission Disequilibrium Test. A meta-analysis of previous studies of the association was conducted to place our result in context. RESULTS: The heritability of carotid IMT was 24%. Under a recessive model (GG+GC versus CC), there was significant evidence of association between IL-6 to the -174G/C genotype and adjusted log(e) maximal carotid IMT (F=5.469; P=0.02). Family-based analyses using Quantitative Transmission Disequilibrium Test showed no evidence of population stratification as a cause of the observed association (chi2(1)=0.469; P=0.4934). The CC genotype was associated with a 4.8% increase in maximal carotid IMT and accounted for 0.6% of the observed variation in the trait, which is equivalent to 2.5% of the heritable component. A meta-analysis of the present and 2 previous large studies, which enrolled a total of 2930 subjects, confirmed the recessive effect of the C allele on carotid IMT (P=0.0014). CONCLUSIONS: The genotype at the IL-6 to -174G/C polymorphism is associated with common carotid artery IMT, although the size of the genetic effect is small.


Asunto(s)
Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/diagnóstico , Enfermedades de las Arterias Carótidas/genética , Predisposición Genética a la Enfermedad , Genotipo , Interleucina-6/genética , Polimorfismo Genético , Túnica Íntima/patología , Túnica Media/patología , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Análisis de Varianza , Arterias Carótidas/diagnóstico por imagen , Ecocardiografía , Salud de la Familia , Femenino , Humanos , Inflamación , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Modelos Genéticos , Modelos Estadísticos , Túnica Íntima/diagnóstico por imagen , Túnica Media/diagnóstico por imagen , Ultrasonografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...