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

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
2.
Nature ; 515(7526): 279-282, 2014 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25119035

RESUMO

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.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/fisiologia , Fricção , Canais Iônicos/metabolismo , Estresse Mecânico , Animais , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/metabolismo , Feminino , Hemorreologia , Masculino , Camundongos
3.
Circ Res ; 120(5): 784-798, 2017 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-27920123

RESUMO

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.


Assuntos
Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Resistência à Insulina/fisiologia , Transdução de Sinais/fisiologia , Animais , Aterosclerose/patologia , Células Cultivadas , Células Endoteliais/patologia , Endotélio Vascular/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos
4.
Stem Cells ; 32(10): 2714-23, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24916783

RESUMO

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.


Assuntos
Vasos Sanguíneos/patologia , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Cicatrização , Adulto , Animais , Ásia , Demografia , Células Endoteliais/efeitos dos fármacos , Inativação Gênica , Humanos , Insulina/farmacologia , Masculino , Camundongos Nus , Fosforilação/efeitos dos fármacos , Fatores de Risco , População Branca , Cicatrização/efeitos dos fármacos
5.
Arterioscler Thromb Vasc Biol ; 34(9): 2051-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25012136

RESUMO

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.


Assuntos
Doenças das Artérias Carótidas/prevenção & controle , Endotélio Vascular/fisiologia , Artéria Femoral/lesões , Células-Tronco Hematopoéticas/fisiologia , Neovascularização Fisiológica/fisiologia , Receptor IGF Tipo 1/fisiologia , Animais , Aorta Torácica/patologia , Apolipoproteínas E/deficiência , Doenças das Artérias Carótidas/etiologia , Doenças das Artérias Carótidas/genética , Adesão Celular , Endotélio Vascular/metabolismo , Feminino , Regulação da Expressão Gênica , Genótipo , Transplante de Células-Tronco Hematopoéticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/metabolismo , Fenótipo , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor IGF Tipo 1/deficiência , Receptor IGF Tipo 1/genética , Regeneração
6.
Pflugers Arch ; 465(8): 1065-74, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23338941

RESUMO

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.


Assuntos
Endotélio/metabolismo , Insulina/metabolismo , Óxido Nítrico/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/fisiologia , Animais , Disponibilidade Biológica , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Endotélio/fisiologia , Humanos
7.
Adipocyte ; 11(1): 366-378, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35734881

RESUMO

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.


Assuntos
Resistência à Insulina , Receptor IGF Tipo 1 , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Anticorpos Monoclonais Humanizados , Dieta Hiperlipídica/efeitos adversos , Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Lipídeos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/metabolismo , Receptor IGF Tipo 1/antagonistas & inibidores
8.
Biochim Biophys Acta ; 1801(3): 320-6, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19818873

RESUMO

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.


Assuntos
Endotélio Vascular/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Resistência à Insulina , Doenças Vasculares/metabolismo , Animais , Dislipidemias/metabolismo , Humanos , NADPH Oxidases/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
9.
Endocrinology ; 159(8): 2917-2925, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29796592

RESUMO

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.


Assuntos
Aorta/metabolismo , Glicemia/metabolismo , Endotélio Vascular/metabolismo , Haploinsuficiência/genética , Receptor de Insulina/genética , Vasodilatação/genética , Acetilcolina/farmacologia , Animais , Antígenos CD/genética , Aorta/fisiopatologia , Pressão Sanguínea , Movimento Celular , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Teste de Tolerância a Glucose , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
10.
Diabetes ; 66(2): 287-299, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28108607

RESUMO

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.


Assuntos
Glicemia/efeitos dos fármacos , Resistência à Insulina , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Animais , Glicemia/metabolismo , Linhagem Celular , Proliferação de Células , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Immunoblotting , Técnicas In Vitro , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Obesos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
11.
Diabetes ; 66(11): 2808-2821, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28830894

RESUMO

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.


Assuntos
Endotélio Vascular/metabolismo , Resistência à Insulina/fisiologia , NADPH Oxidase 2/metabolismo , Estresse Oxidativo/fisiologia , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/metabolismo , Animais , Aorta , Células Cultivadas , Células Endoteliais , Regulação da Expressão Gênica/fisiologia , Técnica Clamp de Glucose , Intolerância à Glucose , Camundongos , Camundongos Knockout , NADPH Oxidase 2/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/genética , Vasoconstrição/fisiologia
12.
J Clin Endocrinol Metab ; 91(12): 5051-6, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16984984

RESUMO

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.


Assuntos
Aldosterona/biossíntese , Variação Genética , Esteroide 11-beta-Hidroxilase/genética , Aldosterona/análogos & derivados , Aldosterona/metabolismo , Aldosterona/urina , Cortodoxona/análogos & derivados , Cortodoxona/urina , Citocromo P-450 CYP11B2/genética , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Esteroide 11-beta-Hidroxilase/metabolismo , Esteroides/urina
13.
Diabetes ; 54(8): 2492-6, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16046320

RESUMO

Rare mutations in the proopiomelanocortin (POMC) gene cause severe early-onset childhood obesity. However, it is unknown whether common variants in POMC are responsible for variation in body weight or fat distribution within the commonly observed range in the population. We tested for association between three polymorphisms spanning the POMC gene and obesity phenotypes in 1,428 members of 248 families. There was significant association between genotypes at the C8246T (P < 0.0001) and C1032G (P = 0.003) polymorphisms and waist-to-hip ratio (WHR) corrected for age, sex, smoking, exercise, and alcohol consumption. Each T allele at C8246T (or G allele at C1032G) was associated with a 0.2-SD-higher WHR in a codominant fashion. When WHR was additionally corrected for BMI, thus providing a measure of body fat distribution throughout the range of BMI, there remained significant evidence for association with both markers that was of similar magnitude and statistical significance. There was no association between genotype at any polymorphism and BMI or plasma leptin level. These data show that genetic variants at the POMC locus influence body fat distribution within the normal range, suggesting a novel role for POMC in metabolic regulation.


Assuntos
Tecido Adiposo , Composição Corporal/genética , Polimorfismo Genético/genética , Pró-Opiomelanocortina/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Índice de Massa Corporal , Genótipo , Humanos , Leptina/sangue , Pessoa de Meia-Idade , Relação Cintura-Quadril
14.
Stroke ; 36(10): 2215-9, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16179573

RESUMO

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.


Assuntos
Artérias Carótidas/patologia , Doenças das Artérias Carótidas/diagnóstico , Doenças das Artérias Carótidas/genética , Predisposição Genética para Doença , Genótipo , Interleucina-6/genética , Polimorfismo Genético , Túnica Íntima/patologia , Túnica Média/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Análise de Variância , Artérias Carótidas/diagnóstico por imagem , Ecocardiografia , Saúde da Família , Feminino , Humanos , Inflamação , Desequilíbrio de Ligação , Masculino , Pessoa de Meia-Idade , Modelos Genéticos , Modelos Estatísticos , Túnica Íntima/diagnóstico por imagem , Túnica Média/diagnóstico por imagem , Ultrassonografia
15.
J Clin Endocrinol Metab ; 90(2): 1072-7, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15522937

RESUMO

Genetic variation in the gene encoding aldosterone synthase (CYP11B2) has previously been shown to be associated with hypertension and left ventricular hypertrophy. The intermediate phenotype most consistently associated with variation at this locus is that of elevated plasma 11-deoxycortisol (S). However, in normal subjects, aldosterone synthase does not metabolize S, which is converted to cortisol (F) by the enzyme 11 beta hydroxylase, encoded by the gene CYP11B1, which lies adjacent to CYP11B2 on chromosome 8. It is possible that the quantitative trait locus for the phenotype is within CYP11B1 and that linkage disequilibrium across the extended locus could account for these observations. However, variation across the whole CYP11B1/B2 locus had not been extensively characterized with respect to these phenotypes. We genotyped six polymorphisms in the CYP11B2 gene and three polymorphisms in the CYP11B1 gene in 248 Caucasian nuclear families comprising 1428 individuals. We measured plasma levels of S and F in 460 individuals from 86 families and urinary excretion rates of tetrahydrodeoxycortisol (THS) and tetrahydrodeoxycorticosterone in 573 individuals from 105 families. We examined heritability of the phenotypes and their association with genotypes and haplotypes at this locus. All steroid phenotypes except urinary tetrahydrodeoxycorticosterone were highly heritable (P < 0.00001). There was strong linkage disequilibrium across the CYP11B1/B2 locus. There was modest evidence for association between polymorphisms of CYP11B2 and plasma levels of S (P = 0.02 for T4986C polymorphism) and the plasma S to F ratio, reflecting the activity of 11-beta hydroxylase (P = 0.01 for T4986C polymorphism). There was strong evidence for association between polymorphisms of both CYP11B1 and CYP11B2 and urinary THS, which was strongest for the CYP11B1 exon 1 polymorphism (P = 0.00002). Addition of other marker data to CYP11B1 exon 1 did not improve the fit of a log-linear model. Genotype at CYP11B1 explained approximately 5% of the variance in urinary THS excretion in the population. Thus, it is likely that linkage disequilibrium between causative CYP11B1 variants and CYP11B2 polymorphisms account for the previous observations. Further fine-mapping studies across the CYP11B1 locus are required to localize the causative variant(s) for the biochemical phenotype; this may also identify susceptibility alleles for hypertension and left ventricular hypertrophy.


Assuntos
Cortodoxona/urina , Citocromo P-450 CYP11B2/genética , Variação Genética , Desequilíbrio de Ligação/genética , Polimorfismo Genético , Esteroide 11-beta-Hidroxilase/genética , Corticosteroides/sangue , Pressão Sanguínea , Mapeamento Cromossômico , Éxons/genética , Feminino , Genótipo , Humanos , Íntrons/genética , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Caracteres Sexuais , Reino Unido , População Branca/genética
16.
J Clin Endocrinol Metab ; 88(12): 6034-9, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14671208

RESUMO

The immunodysregulation, polyendocrinopathy, enteropathy syndrome (IPEX), is a rare disorder of immune regulation resulting in multiple autoimmune disorders, which demonstrates X-linked recessive inheritance. The disease gene, FOXP3, was identified in 2001, and several mutations within this gene have since been described in patients with IPEX. We used linkage analysis, mutational screening of the FOXP3 gene, human leukocyte antigen typing, and analysis of X-chromosome inactivation to investigate 2 kindreds (21 subjects in total) with 4 male infants (3 now deceased) and 1 girl affected by IPEX. In 1 family a novel FOXP3 mutation was identified in the proband, with a single base deletion at codon 76 of exon 2, leading to a frameshift, which predicted a truncated protein product (108 residues vs. 431 in wild type). In the second family, the FOXP3 locus was excluded by recombination, and mutational analysis of the gene was negative. The affected girl from this family was shown to have human leukocyte antigen DR2 and DR6 alleles and random X-chromosome inactivation in peripheral blood mononuclear cells. Our analysis has elucidated the molecular basis of IPEX in one family and has, for the first time, provided evidence for an autosomal locus, suggesting genetic heterogeneity in this syndrome.


Assuntos
Proteínas de Ligação a DNA/genética , Heterogeneidade Genética , Doenças do Sistema Imunitário/genética , Enteropatias/genética , Poliendocrinopatias Autoimunes/genética , Sequência de Bases/genética , Estudos de Casos e Controles , DNA/genética , Análise Mutacional de DNA , Mecanismo Genético de Compensação de Dose , Feminino , Fatores de Transcrição Forkhead , Ligação Genética , Humanos , Lactente , Recém-Nascido , Escore Lod , Masculino , Mutação , Linhagem , Síndrome
17.
Atherosclerosis ; 230(1): 131-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23958265

RESUMO

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.


Assuntos
Aterosclerose/fisiopatologia , Endotélio Vascular/patologia , Resistência à Insulina , Espécies Reativas de Oxigênio , Acetilcolina/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/metabolismo , Pressão Sanguínea , Peso Corporal , Células Endoteliais/citologia , Endotélio Vascular/metabolismo , Genes Dominantes , Glucose/metabolismo , Teste de Tolerância a Glucose , Humanos , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Receptor de Insulina/genética , Fatores de Risco
18.
Diabetes ; 62(6): 2130-4, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23349484

RESUMO

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.


Assuntos
Células Endoteliais/efeitos dos fármacos , Resistência à Insulina/fisiologia , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Acetilcolina/farmacologia , Animais , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Immunoblotting , Resistência à Insulina/genética , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/genética , Reação em Cadeia da Polimerase , Vasodilatadores/farmacologia
19.
Diabetes ; 61(4): 915-24, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22357965

RESUMO

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.


Assuntos
Aterosclerose/prevenção & controle , Pressão Sanguínea/fisiologia , Diabetes Mellitus/metabolismo , Resistência à Insulina/fisiologia , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Óxido Nítrico/biossíntese , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Células Cultivadas , Células Endoteliais , Deleção de Genes , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Camundongos , Camundongos Transgênicos , Obesidade/metabolismo , Receptor de Insulina/genética
20.
Diabetes ; 61(9): 2359-68, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22733797

RESUMO

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.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/fisiologia , Receptor IGF Tipo 1/fisiologia , Animais , Aorta/fisiologia , Glicemia/metabolismo , Pressão Sanguínea , Feminino , Homeostase , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/biossíntese , Receptor IGF Tipo 1/deficiência , Receptor IGF Tipo 1/genética , Vasoconstrição/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA