Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Diabetes ; 55(12): 3529-35, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17130501

RESUMO

Impaired fasting glucose (IFG) is more prevalent in men and impaired glucose tolerance (IGT) more prevalent in women. To explore whether this sex difference is related to female sex hormones, we performed a cross-sectional analysis of data from 2,164 (1,329 women and 835 men) first-degree relatives of individuals with type 2 diabetes. Subjects were categorized based on a 75-g oral glucose tolerance test. Sex and hormone replacement therapy (HRT) effects on the distribution of glucose tolerance were assessed using multinomial logistic regression corrected for familial clustering. Compared with men, women were more likely to have isolated IGT (relative risk 1.8 [95% CI 1.3-2.5]) and less likely to have isolated IFG (0.5 [0.3-0.7]) adjusted for ethnicity, age, waist, fasting insulin, and early insulin release (DeltaI(0-30)/DeltaG(0-30)). To evaluate HRT effects, postmenopausal women using (n = 238) or not using (n = 378) HRT were compared. HRT users were more likely to have isolated IGT (2.2 [1.2-4.0]) after adjustment, but the prevalence of isolated IFG did not differ by HRT status. Based on the influence of sex and HRT on the prevalence of isolated IFG and isolated IGT, we conclude that female sex hormones may play an important role in the pathogenesis of IFG and IGT.


Assuntos
Diabetes Mellitus Tipo 2/genética , Terapia de Reposição de Estrogênios , Intolerância à Glucose/epidemiologia , Adulto , Idoso , Estudos Transversais , Família , Feminino , Teste de Tolerância a Glucose , Humanos , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Análise de Regressão , Estudos Retrospectivos , Caracteres Sexuais
2.
BMC Med Genomics ; 8: 20, 2015 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-25956355

RESUMO

BACKGROUND: Metabolic syndrome (MetS) is characterized by central obesity, insulin resistance, dysglycemia, and a pro-atherogenic plasma lipid profile. MetS creates a high risk for development of type 2 diabetes (T2DM) and cardiovascular disease (CVD), presumably by altering inflammatory responses. Presently, it is unknown how the chronic metabolic disturbances in acute hyperglycemia, MetS and T2DM affect the immune activity of peripheral blood cells. METHODS: We performed genome-wide expression analysis of peripheral blood cells obtained from patients with T2DM (n = 6) and age-, sex- , BMI- and blood pressure-matched obese individuals with MetS (n = 4) and lean healthy normoglycemic controls (n = 3), both under fasting conditions and after controlled induction of acute hyperglycemia during a 70 min hyperglycemic clamp. Differential gene expression during fasting conditions was confirmed by real-time PCR, for which we included additional age-, sex-, BMI-, and blood pressure-matched obese individuals with (n = 4) or without (n = 4) MetS. RESULTS: Pathway and Gene ontology analysis applied to baseline expression profiles of peripheral blood cells from MetS and T2DM patients revealed metabolic changes, highly similar to a reoviral infection gene signature in T2DM patients. Transcription factor binding site analysis indicated that increased HIF-1α activity, a transcription factor induced by either hypoxia or oxidative stress, is responsible for this aberrant metabolic profile in peripheral blood cells from T2DM patients. Acute hyperglycemia in healthy controls resulted in reduced expression of cytotoxicity-related genes, representing NK- and CD8(+) cells. In obese controls, MetS and especially T2DM patients, baseline expression of genes involved in cytotoxicity was already low, compared to healthy controls and did not further decrease upon acute hyperglycemia. CONCLUSIONS: The reduced activity of cytotoxic genes in T2DM is explained by chronic hyperglycemia, but its acute effects are restricted to healthy controls. Genome expression of circulating leukocytes from T2DM patients differs from MetS individuals by a specific reovirus signature. Our data thus suggest a role for suppressed anti-viral capacity in the etiology of diabetes.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Síndrome Metabólica/sangue , Reoviridae/genética , Idoso , Pressão Sanguínea , Índice de Massa Corporal , Doenças Cardiovasculares/metabolismo , Estudos de Casos e Controles , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/virologia , Feminino , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Técnica Clamp de Glucose , Humanos , Inflamação/metabolismo , Lipídeos/sangue , Masculino , Síndrome Metabólica/genética , Pessoa de Meia-Idade , Obesidade Abdominal/genética , Obesidade Abdominal/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos
3.
Vascul Pharmacol ; 75: 7-18, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26254104

RESUMO

A defect in neo-vascularization process involving circulating angiogenic mononuclear cells (CACs) dysfunction is associated with diabetes. We showed that oxidative stress was elevated in CACs cultured from blood of individuals with metabolic syndrome (MetS) and diabetes. We then assessed the action of palmitic acid (PA), a deregulated and increased NEFA in metabolic disorders, focusing on its oxidant potential. We observed that the phyto-polyphenol resveratrol normalized oxidative stress both in CACs isolated from MetS patients or treated with PA. Resveratrol further decreased the deleterious action of PA on gene expression of vascularization factors (TNFα, VEGF-A, SDF1α, PECAM-1, VEGFR2, Tie2 and CXCR4) and improved CAC motility. Particularly, resveratrol abolished the PA-induced over-expression of the pro-oxidant protein p66Shc. Neither KLF2 nor SIRT1, previously shown in resveratrol and p66Shc action, was directly involved. Silencing p66Shc normalized PA action on VEGF-A and TNFα specifically, without abolishing the PA-induced oxidative stress, which suggests a deleterious role of p66Shc independently of any major modulation of the cellular oxidative status in a high NEFA levels context. Besides showing that resveratrol reverses PA-induced harmful effects on human CAC function, certainly through profound cellular modifications, we establish p66Shc as a major therapeutic target in metabolic disorders, independent from glycemic control.


Assuntos
Estresse Oxidativo/efeitos dos fármacos , Ácido Palmítico/metabolismo , Proteínas Adaptadoras da Sinalização Shc/genética , Estilbenos/farmacologia , Antioxidantes/farmacologia , Estudos de Casos e Controles , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Tipo 2/fisiopatologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Síndrome Metabólica/fisiopatologia , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos dos fármacos , Resveratrol , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
4.
Eur J Endocrinol ; 170(3): 429-39, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24297090

RESUMO

OBJECTIVE: Anti-inflammatory glucocorticoid (GC) therapy often induces hyperglycemia due to insulin resistance and islet-cell dysfunction. Incretin-based therapies may preserve glucose tolerance and pancreatic islet-cell function. In this study, we hypothesized that concomitant administration of the dipeptidyl peptidase-4 inhibitor sitagliptin and prednisolone in men at high risk to develop type 2 diabetes could protect against the GC-induced diabetogenic effects. DESIGN AND METHODS: Men with the metabolic syndrome but without diabetes received prednisolone 30  mg once daily plus sitagliptin 100  mg once daily (n=14), prednisolone (n=12) or sitagliptin alone (n=14) or placebo (n=12) for 14 days in a double-blind 2 × 2 randomized-controlled study. Glucose, insulin, C-peptide, and glucagon were measured in the fasted state and following a standardized mixed-meal test. ß-cell function parameters were assessed both from a hyperglycemic-arginine clamp procedure and from the meal test. Insulin sensitivity (M-value) was measured by euglycemic clamp. RESULTS: Prednisolone increased postprandial area under the curve (AUC)-glucose by 17% (P<0.001 vs placebo) and postprandial AUC-glucagon by 50% (P<0.001). Prednisolone reduced 1st and 2nd phase glucose-stimulated- and combined hyperglycemia-arginine-stimulated C-peptide secretion (all P ≤ 0.001). When sitagliptin was added, both clamp-measured ß-cell function (P=NS for 1st and 2nd phase vs placebo) and postprandial hyperglucagonemia (P=NS vs placebo) remained unaffected. However, administration of sitagliptin could not prevent prednisolone-induced increment in postprandial glucose concentrations (P<0.001 vs placebo). M-value was not altered by any treatment. CONCLUSION: Fourteen-day treatment with high-dose prednisolone impaired postprandial glucose metabolism in subjects with the metabolic syndrome. Concomitant treatment with sitagliptin improved various aspects of pancreatic islet-cell function, but did not prevent deterioration of glucose tolerance by GC treatment.


Assuntos
Diabetes Mellitus Tipo 2/prevenção & controle , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Síndrome Metabólica/tratamento farmacológico , Pirazinas/uso terapêutico , Triazóis/uso terapêutico , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Método Duplo-Cego , Jejum , Glucocorticoides , Intolerância à Glucose/prevenção & controle , Humanos , Resistência à Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Pessoa de Meia-Idade , Período Pós-Prandial/fisiologia , Prednisolona/uso terapêutico , Fosfato de Sitagliptina
5.
Eur J Endocrinol ; 170(4): 565-74, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24421302

RESUMO

OBJECTIVE: Type 2 diabetes mellitus (T2DM) management requires continuous treatment intensification due to progressive decline in ß-cell function in insulin resistant individuals. Initial combination therapy of a dipeptidyl peptidase (DPP)-4 inhibitor with a thiazolidinedione (TZD) may be rational. We assessed the effects of the DPP4 inhibitor alogliptin (ALO) combined with the TZD pioglitazone (PIO), vs ALO monotherapy or placebo (PBO), on ß-cell function and glycemic control in T2DM. MATERIAL AND METHODS: A 16-week, two-center, randomized, double-blind, PBO-controlled, parallel-arm intervention study in 71 patients with well-controlled T2DM (age 59.1±6.3 years; A1C 6.7±0.1%) treated with metformin, sulfonylurea, or glinide monotherapy was conducted. Patients were treated with combined ALO 25 mg and PIO 30 mg daily or ALO 25 mg daily monotherapy or PBO. Main outcome measures included change in A1C and fasting plasma glucose (FPG) from baseline to week 16. In addition, change in ß-cell function parameters obtained from standardized meal tests at baseline and at week 16 was measured. RESULTS: ALO/PIO and ALO decreased A1C from baseline by 0.9±0.1 and 0.4±0.2% respectively (both P<0.001 vs PBO). FPG was decreased to a greater extent by ALO/PIO compared with ALO monotherapy (P<0.01). ALO/PIO treatment improved ß-cell glucose sensitivity (vs PBO; P<0.001) and fasting secretory tone (vs PBO; P=0.001), while ALO monotherapy did not change ß-cell function parameters. All treatments were well tolerated. CONCLUSION: Short-term treatment with ALO/PIO or ALO improved glycemic control in well-controlled T2DM patients, but only combined ALO/PIO improved ß-cell function. These data support that initial combination therapy with a DPP4 inhibitor and TZD to address multiple core defects in T2DM may be a sensible approach.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Piperidinas/uso terapêutico , Tiazolidinedionas/uso terapêutico , Uracila/análogos & derivados , Idoso , Glicemia , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Hemoglobinas Glicadas , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Masculino , Pessoa de Meia-Idade , Pioglitazona , Resultado do Tratamento , Uracila/uso terapêutico
6.
Metabolism ; 62(4): 568-77, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23164480

RESUMO

AIM: Glucocorticoids impair glucose tolerance by inducing insulin resistance. We investigated the dose-dependent effects of glucocorticoid treatment on islet-cell function in healthy males and studied the role of the autonomic nervous system. DESIGN AND METHODS: A randomized, placebo-controlled, double-blind, dose-response intervention study was conducted in 32 healthy males (age: 21±2years; BMI: 21.9±1.7kg/m(2)). Participants were allocated to prednisolone 7.5mg once daily (n=12), prednisolone 30mg once daily (n=12), or placebo (n=8) for two weeks. Beta-cell function was measured by hyperglycemic clamp with arginine stimulation, glucagon levels were measured following a standardized meal test. RESULTS: We found that prednisolone treatment dose-dependently reduced C-peptide secretion following arginine stimulation on top of hyperglycemia (ASI-iAUCCP): -2.8 (-5.2;0.2) and -3.1 (-8.8; -1.0) nmolL(-1)min(-1) for prednisolone 7.5mg and prednisolone 30mg, respectively (P=0.035 vs. placebo). Fasting glucagon levels increased dose-dependently (vs. placebo; P=0.001), whereas postprandial glucagon levels were only increased by prednisolone 30mg. Changes in parasympathetic activity related with changes in fasting glucose levels (r=-0.407; P=0.03) and showed a trend towards correlation with fasting glucagon concentrations (r=-0.337; P=0.07). The change in sympathovagal balance was inversely related to ASI-iAUCCP (r=-0.365; P=0.05). CONCLUSION: We conclude that in addition to inducing insulin resistance, prednisolone treatment dose-dependently impaired islet-cell function. Altered sympathovagal balance may be related to these effects.


Assuntos
Glucocorticoides/toxicidade , Ilhotas Pancreáticas/efeitos dos fármacos , Pancreatopatias/induzido quimicamente , Sistema Nervoso Simpático/fisiopatologia , Nervo Vago/fisiopatologia , Adolescente , Adulto , Antropometria , Arginina/farmacologia , Glicemia/metabolismo , Relação Dose-Resposta a Droga , Método Duplo-Cego , Glucagon/sangue , Técnica Clamp de Glucose , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Humanos , Hiperglicemia/induzido quimicamente , Incretinas/metabolismo , Masculino , Pancreatopatias/fisiopatologia , Testes de Função Pancreática , Prednisolona/farmacologia , Estimulação Química , Adulto Jovem
7.
Diabetes Care ; 34(2): 412-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21216851

RESUMO

OBJECTIVE: Glucocorticoids (GCs) are regarded as diabetogenic because they impair insulin sensitivity and islet-cell function. This study assessed whether treatment with the glucagon-like peptide receptor agonist (GLP-1 RA) exenatide (EXE) could prevent GC-induced glucose intolerance. RESEARCH DESIGN AND METHODS: A randomized, placebo-controlled, double-blind, crossover study in eight healthy men (age: 23.5 [20.0-28.3] years; BMI: 26.4 [24.3-28.0] kg/m(2)) was conducted. Participants received three therapeutic regimens for 2 consecutive days: 1) 80 mg of oral prednisolone (PRED) every day (q.d.) and intravenous (IV) EXE infusion (PRED+EXE); 2) 80 mg of oral PRED q.d. and IV saline infusion (PRED+SAL); and 3) oral placebo-PRED q.d. and intravenous saline infusion (PLB+SAL). On day 1, glucose tolerance was assessed during a meal challenge test. On day 2, participants underwent a clamp procedure to measure insulin secretion and insulin sensitivity. RESULTS: PRED+SAL treatment increased postprandial glucose levels (vs. PLB+SAL, P = 0.012), which was prevented by concomitant EXE (vs. PLB+SAL, P = NS). EXE reduced PRED-induced hyperglucagonemia during the meal challenge (P = 0.018) and decreased gastric emptying (vs. PRED+SAL, P = 0.028; vs. PLB+SAL, P = 0.046). PRED+SAL decreased first-phase glucose- and arginine-stimulated C-peptide secretion (vs. PLB+SAL, P = 0.017 and P = 0.05, respectively), whereas PRED+EXE improved first- and second-phase glucose- and arginine-stimulated C-peptide secretion (vs. PLB+SAL; P = 0.017, 0.012, and 0.093, respectively). CONCLUSIONS: The GLP-1 RA EXE prevented PRED-induced glucose intolerance and islet-cell dysfunction in healthy humans. Incretin-based therapies should be explored as a potential strategy to prevent steroid diabetes.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/agonistas , Intolerância à Glucose/prevenção & controle , Hipoglicemiantes/administração & dosagem , Peptídeos/administração & dosagem , Prednisona/efeitos adversos , Peçonhas/administração & dosagem , Adolescente , Adulto , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Peptídeo C/sangue , Estudos Cross-Over , Exenatida , Glucocorticoides/efeitos adversos , Técnica Clamp de Glucose , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/tratamento farmacológico , Humanos , Hiperglicemia/tratamento farmacológico , Hiperinsulinismo/tratamento farmacológico , Hipoglicemiantes/efeitos adversos , Resistência à Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Masculino , Peptídeos/efeitos adversos , Peçonhas/efeitos adversos , Adulto Jovem
8.
Diabetes Res Clin Pract ; 86 Suppl 1: S26-34, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20115929

RESUMO

Type 2 diabetes (T2DM) is a heterogeneous syndrome, characterized by beta-cell failure in the setting of obesity-related insulin resistance. T2DM has a progressive course and is associated with a high cardiovascular disease (CVD) risk, regardless of the treatment used. The incretin hormones glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic polypeptide (GIP) are secreted in the gut upon meal ingestion and lower blood glucose by glucose-dependent stimulation of insulin secretion and production. Exogenously administered GLP-1 lowers postprandial glucose excursions by inhibiting glucagon secretion and delaying gastric emptying, improves beta-cell function, and promotes satiety and weight loss. Native GLP-1 is degraded rapidly by the ubiquitous enzyme dipeptidyl-peptidase (DPP)-4. Thus, injectable DPP-4-resistant GLP-1 receptor agonists (GLP-1RA) and oral DPP-4 inhibitors have been developed. Exenatide is the first GLP-1RA that became available for the treatment of T2DM patients. Exenatide has unique characteristics, as to date it is the only agent that addresses the multiple defects of the T2DM phenotype, including hyperglycaemia, islet-cell dysfunction, alimentary obesity, insulin resistance, hypertension and dyslipidaemia. In animals, exenatide also increased beta-cell mass. Long-term prospective studies in high-risk populations should address the potentially disease modifying effect of exenatide and its effect on CVD risk, in addition to its safety and tolerability.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Peptídeos/uso terapêutico , Receptores de Glucagon/agonistas , Peçonhas/uso terapêutico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatologia , Exenatida , Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Incretinas/fisiologia , Incretinas/uso terapêutico , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA