Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.203
Filtrar
2.
Nature ; 627(8003): 347-357, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38374256

RESUMO

Type 2 diabetes (T2D) is a heterogeneous disease that develops through diverse pathophysiological processes1,2 and molecular mechanisms that are often specific to cell type3,4. Here, to characterize the genetic contribution to these processes across ancestry groups, we aggregate genome-wide association study data from 2,535,601 individuals (39.7% not of European ancestry), including 428,452 cases of T2D. We identify 1,289 independent association signals at genome-wide significance (P < 5 × 10-8) that map to 611 loci, of which 145 loci are, to our knowledge, previously unreported. We define eight non-overlapping clusters of T2D signals that are characterized by distinct profiles of cardiometabolic trait associations. These clusters are differentially enriched for cell-type-specific regions of open chromatin, including pancreatic islets, adipocytes, endothelial cells and enteroendocrine cells. We build cluster-specific partitioned polygenic scores5 in a further 279,552 individuals of diverse ancestry, including 30,288 cases of T2D, and test their association with T2D-related vascular outcomes. Cluster-specific partitioned polygenic scores are associated with coronary artery disease, peripheral artery disease and end-stage diabetic nephropathy across ancestry groups, highlighting the importance of obesity-related processes in the development of vascular outcomes. Our findings show the value of integrating multi-ancestry genome-wide association study data with single-cell epigenomics to disentangle the aetiological heterogeneity that drives the development and progression of T2D. This might offer a route to optimize global access to genetically informed diabetes care.


Assuntos
Diabetes Mellitus Tipo 2 , Progressão da Doença , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Adipócitos/metabolismo , Cromatina/genética , Cromatina/metabolismo , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/genética , Diabetes Mellitus Tipo 2/classificação , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Nefropatias Diabéticas/complicações , Nefropatias Diabéticas/genética , Células Endoteliais/metabolismo , Células Enteroendócrinas , Epigenômica , Predisposição Genética para Doença/genética , Ilhotas Pancreáticas/metabolismo , Herança Multifatorial/genética , Doença Arterial Periférica/complicações , Doença Arterial Periférica/genética , Análise de Célula Única
5.
Nature ; 624(7992): 621-629, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38049589

RESUMO

Type 2 diabetes mellitus (T2D), a major cause of worldwide morbidity and mortality, is characterized by dysfunction of insulin-producing pancreatic islet ß cells1,2. T2D genome-wide association studies (GWAS) have identified hundreds of signals in non-coding and ß cell regulatory genomic regions, but deciphering their biological mechanisms remains challenging3-5. Here, to identify early disease-driving events, we performed traditional and multiplexed pancreatic tissue imaging, sorted-islet cell transcriptomics and islet functional analysis of early-stage T2D and control donors. By integrating diverse modalities, we show that early-stage T2D is characterized by ß cell-intrinsic defects that can be proportioned into gene regulatory modules with enrichment in signals of genetic risk. After identifying the ß cell hub gene and transcription factor RFX6 within one such module, we demonstrated multiple layers of genetic risk that converge on an RFX6-mediated network to reduce insulin secretion by ß cells. RFX6 perturbation in primary human islet cells alters ß cell chromatin architecture at regions enriched for T2D GWAS signals, and population-scale genetic analyses causally link genetically predicted reduced RFX6 expression with increased T2D risk. Understanding the molecular mechanisms of complex, systemic diseases necessitates integration of signals from multiple molecules, cells, organs and individuals, and thus we anticipate that this approach will be a useful template to identify and validate key regulatory networks and master hub genes for other diseases or traits using GWAS data.


Assuntos
Diabetes Mellitus Tipo 2 , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Predisposição Genética para Doença , Ilhotas Pancreáticas , Humanos , Estudos de Casos e Controles , Separação Celular , Cromatina/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Redes Reguladoras de Genes/genética , Estudo de Associação Genômica Ampla , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Reprodutibilidade dos Testes
7.
Front Endocrinol (Lausanne) ; 14: 1261298, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37854186

RESUMO

Experimental studies in animal models of aging such as nematodes, fruit flies or mice have observed that decreased levels of insulin or insulin signaling promotes longevity. In humans, hyperinsulinemia and concomitant insulin resistance are associated with an elevated risk of age-related diseases suggestive of a shortened healthspan. Age-related disorders include neurodegenerative diseases, hypertension, cardiovascular disease, and type 2 diabetes. High ambient insulin concentrations promote increased lipogenesis and fat storage, heightened protein synthesis and accumulation of non-functional polypeptides due to limited turnover capacity. Moreover, there is impaired autophagy activity, and less endothelial NO synthase activity. These changes are associated with mitochondrial dysfunction and oxidative stress. The cellular stress induced by anabolic activity of insulin initiates an adaptive response aiming at maintaining homeostasis, characterized by activation of the transcription factor Nrf2, of AMP activated kinase, and an unfolded protein response. This protective response is more potent in the long-lived human species than in short-lived models of aging research resulting in a stronger pro-aging impact of insulin in nematodes and fruit flies. In humans, resistance to insulin-induced cell stress decreases with age, because of an increase of insulin and insulin resistance levels but less Nrf2 activation. These detrimental changes might be contained by adopting a lifestyle that promotes low insulin/insulin resistance levels and enhances an adaptive response to cellular stress, as observed with dietary restriction or exercise.


Assuntos
Envelhecimento , Hiperinsulinismo , Resistência à Insulina , Insulina , Animais , Humanos , Camundongos , Envelhecimento/fisiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Hiperinsulinismo/fisiopatologia , Insulina/análise , Insulina/fisiologia , Resistência à Insulina/fisiologia , Fator 2 Relacionado a NF-E2/metabolismo
8.
Diabetes Res Clin Pract ; 205: 110954, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37839755

RESUMO

AIMS: We compared beta-cell function in Chinese with type 2 diabetes diagnosed at age < 40 years (young-onset diabetes, YOD) and ≥ 40 years (late-onset diabetes, LOD). METHODS: In this cross-sectional study, we selected participants from two cohorts of people with type 2 diabetes recruited in 1996-2012 (n = 4,376) and 2020-2021 (n = 794). Multivariable linear regression models were applied to compare homeostasis model assessment of beta-cell function (HOMA2-%B) and fasting plasma C-peptide across diabetes duration at enrolment between YOD and LOD. RESULTS: The YOD group (n = 1,876, mean [SD] age: 39.9 [7.5] years, median [IQR] diabetes duration: 6 [2-12] years) was more likely to have family history of diabetes (61.6 % vs 43.6 %), obesity (41.9 % vs 26.8 %), dyslipidaemia (61.7 % vs 54.4 %), and worse glycaemic control (mean HbA1c 7.7 % vs 7.4 %) than those with LOD (n = 3,294, age: 60.8 [10.6] years, diabetes duration: 5 [1-10] years). When compared to people with LOD, HOMA2-%B and fasting plasma C-peptide were lower in the YOD group, consistently among those with BMI < 27.5 kg/m2 and HOMA2-IR ≤ 1.6 (median value), adjusted for year at enrolment, sex, diabetes duration, family history of diabetes, HbA1c, weight and lipid indices (p < 0.01). Cross-sectionally, the slopes of decline in HOMA2-%B by diabetes duration were greater in YOD than LOD among individuals with BMI < 27.5 kg/m2 (p-interaction = 0.015). CONCLUSIONS: Chinese with YOD had accelerated loss of beta-cell function than those with LOD especially in non-obese individuals.


Assuntos
Diabetes Mellitus Tipo 2 , Adulto , Humanos , Pessoa de Meia-Idade , Peptídeo C , Estudos Transversais , Diabetes Mellitus Tipo 2/fisiopatologia , População do Leste Asiático , Hemoglobinas Glicadas , Hong Kong/epidemiologia , Obesidade , Idade de Início , Idoso , Células Secretoras de Insulina/fisiologia
10.
Science ; 381(6665): eabl7398, 2023 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-37769095

RESUMO

Systemic metabolism has to be constantly adjusted to the variance of food intake and even be prepared for anticipated changes in nutrient availability. Therefore, the brain integrates multiple homeostatic signals with numerous cues that predict future deviations in energy supply. Recently, our understanding of the neural pathways underlying these regulatory principles-as well as their convergence in the hypothalamus as the key coordinator of food intake, energy expenditure, and glucose metabolism-have been revealed. These advances have changed our view of brain-dependent control of metabolic physiology. In this Review, we discuss new concepts about how alterations in these pathways contribute to the development of prevalent metabolic diseases such as obesity and type 2 diabetes mellitus and how this emerging knowledge may provide new targets for their treatment.


Assuntos
Eixo Encéfalo-Intestino , Diabetes Mellitus Tipo 2 , Ingestão de Alimentos , Metabolismo Energético , Hipotálamo , Vias Neurais , Obesidade , Humanos , Diabetes Mellitus Tipo 2/fisiopatologia , Homeostase , Hipotálamo/fisiologia , Obesidade/fisiopatologia , Vias Neurais/fisiopatologia
14.
J Racial Ethn Health Disparities ; 10(4): 1933-1946, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-35913542

RESUMO

OBJECTIVE: Using the 2013/2014 New York City (NYC) Health and Nutrition Examination Survey (NYCHANES) data, this exploratory study examined whether (a) type 2 diabetes (diabetes) prevalence differed between NYC Afro-Caribbeans and African Americans; (b) anthropometric, biochemical, and sociodemographic diabetes profiles differed between and within groups; and (c) diabetes odds differed between and within groups. METHODS: Diabetes was defined as prior diagnosis, HbA1c ≥ 6.5% (7.8 mmol/L), or fasting glucose ≥ 126 mg/dL. Weighted logistic regression estimated diabetes odds by nativity and either waist circumference (WC) (cm) or BMI (kg/m2). All regression models controlled for age, hypertension, gender, education, income, marital status, physical activity, and smoking. RESULTS: Among Afro-Caribbeans (n = 81, 65% female, age (mean ± SE) 49 ± 2 years, BMI 29.2 ± 0.7 kg/m2) and African Americans (n = 118, 50% female, age 47 ± 2 years, BMI 30.3 ± 0.9 kg/m2), Afro-Caribbeans with diabetes had lower BMI (29.9 ± 0.8 kg/m2 vs. 34.6 ± 1.7 kg/m2, P = 0.01) and lower WC (102 ± 2 cm vs. 114 ± 3 cm, P = 0.002) than African Americans with diabetes. Afro-Caribbeans with diabetes had lower prevalence of obesity (33.2% vs. 74.7%) and higher prevalence of overweight (57.2% vs. 13.5%) (P = 0.02) than African Americans with diabetes. Odds of diabetes did not differ between Afro-Caribbeans and African Americans. In models predicting the effect of WC, diabetes odds increased with WC (OR = 1.07 (95% CI 1.02, 1.11), P = 0.003) and age (OR = 1.09 (95% CI 1.03-1.15), P = 0.003) for African Americans only. In models predicting the effect of BMI, diabetes odds increased for Afro-Caribbeans with age (OR = 1.06 (1.01, 1.11)*, P = 0.04) and hypertension (OR = 5.62 (95% CI 1.04, 30.42), P = 0.045), whereas for African Americans, only age predicted higher diabetes odds (OR = 1.08 (95% CI 1.03, 1.14), P = 0.003). CONCLUSIONS: In NYC, Afro-Caribbeans with diabetes have lower BMI and lower WC than African Americans with diabetes, but odds of diabetes do not differ. Combining African-descent populations into one group obscures clinical differences and generalizes diabetes risk.


Assuntos
Negro ou Afro-Americano , Índice de Massa Corporal , População do Caribe , Diabetes Mellitus Tipo 2 , Circunferência da Cintura , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Negro ou Afro-Americano/estatística & dados numéricos , Região do Caribe/etnologia , Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Emigrantes e Imigrantes/estatística & dados numéricos , Hipertensão/epidemiologia , Fatores de Risco , Circunferência da Cintura/etnologia , Cidade de Nova Iorque/epidemiologia , População do Caribe/estatística & dados numéricos , População Negra/etnologia , População Negra/estatística & dados numéricos
15.
Diabetologia ; 66(1): 44-56, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36224274

RESUMO

AIMS/HYPOTHESIS: South Asians have a two- to fivefold higher risk of developing type 2 diabetes than those of white European descent. Greater central adiposity and storage of fat in deeper or ectopic depots are potential contributing mechanisms. We collated existing and new data on the amount of subcutaneous (SAT), visceral (VAT) and liver fat in adults of South Asian and white European descent to provide a robust assessment of potential ethnic differences in these factors. METHODS: We performed a systematic review of the Embase and PubMed databases from inception to August 2021. Unpublished imaging data were also included. The weighted standardised mean difference (SMD) for each adiposity measure was estimated using random-effects models. The quality of the studies was assessed using the ROBINS-E tool for risk of bias and overall certainty of the evidence was assessed using the GRADE approach. The study was pre-registered with the OSF Registries ( https://osf.io/w5bf9 ). RESULTS: We summarised imaging data on SAT, VAT and liver fat from eight published and three previously unpublished datasets, including a total of 1156 South Asian and 2891 white European men, and 697 South Asian and 2271 white European women. Despite South Asian men having a mean BMI approximately 0.5-0.7 kg/m2 lower than white European men (depending on the comparison), nine studies showed 0.34 SMD (95% CI 0.12, 0.55; I2=83%) more SAT and seven studies showed 0.56 SMD (95% CI 0.14, 0.98; I2=93%) more liver fat, but nine studies had similar VAT (-0.03 SMD; 95% CI -0.24, 0.19; I2=85%) compared with their white European counterparts. South Asian women had an approximately 0.9 kg/m2 lower BMI but 0.31 SMD (95% CI 0.14, 0.48; I2=53%) more liver fat than their white European counterparts in five studies. Subcutaneous fat levels (0.03 SMD; 95% CI -0.17, 0.23; I2=72%) and VAT levels (0.04 SMD; 95% CI -0.16, 0.24; I2=71%) did not differ significantly between ethnic groups in eight studies of women. CONCLUSIONS/INTERPRETATION: South Asian men and women appear to store more ectopic fat in the liver compared with their white European counterparts with similar BMI levels. Given the emerging understanding of the importance of liver fat in diabetes pathogenesis, these findings help explain the greater diabetes risks in South Asians. FUNDING: There was no primary direct funding for undertaking the systematic review and meta-analysis.


Assuntos
Diabetes Mellitus Tipo 2 , Feminino , Humanos , Diabetes Mellitus Tipo 2/etnologia , Diabetes Mellitus Tipo 2/fisiopatologia , Fígado , Gordura Subcutânea , População Branca , População do Sul da Ásia
16.
J Therm Biol ; 110: 103344, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36462879

RESUMO

Current labor demographics are changing, with the number of older adults increasingly engaged in physically demanding occupations expected to continually rise, which are often performed in the heat. Given an age-related decline in whole-body heat loss, older adults are at an elevated risk of developing heat injuries that may be exacerbated by hypertension (HTN) and type 2 diabetes (T2D). Elevated irisin production may play a role in mitigating the excess oxidative stress and acute inflammation associated with physically demanding work in the heat. However, the effects of HTN and T2D on this response remain unclear. Therefore, we evaluated serum irisin before and after 3-h of moderate intensity exercise (metabolic rate: 200 W/m2) and at the end of 60-min of post-exercise recovery in a temperate (wet-bulb globe temperature (WBGT) 16 °C) and high-heat stress (WBGT 32 °C) environment in 12 healthy older men (mean ± SD; 59 ± 4 years), 10 men with HTN (60 ± 4 years), and 9 men with T2D (60 ± 5 years). Core temperature (Tco) was measured continuously. In the heat, total exercise duration was significantly lower in older men with HTN and T2D (both, p ≤ 0.049). Despite Tco not being different between groups, Tco was higher in the hot compared to the temperate condition for all groups (p < 0.001). Similarly, serum irisin concentrations did not differ between groups under either condition but were elevated relative to the temperate condition during post-exercise and end-recovery in the heat (+93.9 pg/mL SEM 26 and + 70.5 pg/mL SEM 38 respectively; both p ≤ 0.014). Thus, our findings indicate similar irisin responses in HTN and T2D compared to healthy, age-matched controls, despite reduced exercise tolerance during prolonged exercise in the heat. Therefore, older workers with HTN and T2D may exhibit greater cellular stress during prolonged exercise in the heat, underlying greater vulnerability to heat-induced cellular injury.


Assuntos
Diabetes Mellitus Tipo 2 , Fibronectinas , Transtornos de Estresse por Calor , Hipertensão , Idoso , Humanos , Masculino , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/fisiopatologia , Exercício Físico/efeitos adversos , Exercício Físico/fisiologia , Tolerância ao Exercício/fisiologia , Fibronectinas/sangue , Fibronectinas/fisiologia , Hipertensão/sangue , Hipertensão/complicações , Hipertensão/fisiopatologia , Transtornos de Estresse por Calor/sangue , Transtornos de Estresse por Calor/complicações , Transtornos de Estresse por Calor/fisiopatologia , Pessoa de Meia-Idade , Envelhecimento/fisiologia
19.
J. physiol. biochem ; 78(2): 307-322, May. 2022.
Artigo em Inglês | IBECS | ID: ibc-215960

RESUMO

Insulin resistance, the most important characteristic of the type 2 diabetes mellitus (T2DM), is mostly caused by impairment in the insulin receptor (IR) signal transduction pathway. Protein tyrosine phosphatase 1B (PTP1B), one of the main negative regulators of the IR signaling pathway, is broadly expressed in various cells and tissues. PTP1B decreases the phosphorylation of the IR resulting in insulin resistance in various tissues. The evidence for the physiological role of PTP1B in regulation of metabolic pathways came from whole-body PTP1B-knockout mice. Whole-body and tissue-specific PTP1B-knockout mice showed improvement in adiposity, insulin resistance, and glucose tolerance. In addition, the key role of PTP1B in the pathogenesis of T2DM and its complications was further investigated in mice models of PTP1B deficient/overexpression. In recent years, targeting PTP1B using PTP1B inhibitors is being considered an attractive target to treat T2DM. PTP1B inhibitors improve the sensitivity of the insulin receptor and have the ability to cure insulin resistance-related diseases. We herein summarized the biological functions of PTP1B in different tissues in vivo and in vitro. We also describe the effectiveness of potent PTP1B inhibitors as pharmaceutical agents to treat T2DM. (AU)


Assuntos
Animais , Camundongos , Diabetes Mellitus Tipo 2/fisiopatologia , Resistência à Insulina , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Inibidores Enzimáticos/farmacologia , Insulina , Camundongos Knockout , Receptor de Insulina
20.
Int J Mol Sci ; 23(5)2022 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-35269594

RESUMO

The abnormal accumulation of methylglyoxal (MG) leading to increased glycation of protein and DNA has emerged as an important metabolic stress, dicarbonyl stress, linked to aging, and disease. Increased MG glycation produces inactivation and misfolding of proteins, cell dysfunction, activation of the unfolded protein response, and related low-grade inflammation. Glycation of DNA and the spliceosome contribute to an antiproliferative and apoptotic response of high, cytotoxic levels of MG. Glyoxalase 1 (Glo1) of the glyoxalase system has a major role in the metabolism of MG. Small molecule inducers of Glo1, Glo1 inducers, have been developed to alleviate dicarbonyl stress as a prospective treatment for the prevention and early-stage reversal of type 2 diabetes and prevention of vascular complications of diabetes. The first clinical trial with the Glo1 inducer, trans-resveratrol and hesperetin combination (tRES-HESP)-a randomized, double-blind, placebo-controlled crossover phase 2A study for correction of insulin resistance in overweight and obese subjects, was completed successfully. tRES-HESP corrected insulin resistance, improved dysglycemia, and low-grade inflammation. Cell permeable Glo1 inhibitor prodrugs have been developed to induce severe dicarbonyl stress as a prospective treatment for cancer-particularly for high Glo1 expressing-related multidrug-resistant tumors. The prototype Glo1 inhibitor is prodrug S-p-bromobenzylglutathione cyclopentyl diester (BBGD). It has antitumor activity in vitro and in tumor-bearing mice in vivo. In the National Cancer Institute human tumor cell line screen, BBGD was most active against the glioblastoma SNB-19 cell line. Recently, potent antitumor activity was found in glioblastoma multiforme tumor-bearing mice. High Glo1 expression is a negative survival factor in chemotherapy of breast cancer where adjunct therapy with a Glo1 inhibitor may improve treatment outcomes. BBGD has not yet been evaluated clinically. Glycation by MG now appears to be a pathogenic process that may be pharmacologically manipulated for therapeutic outcomes of potentially important clinical impact.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Glutationa/análogos & derivados , Hesperidina/uso terapêutico , Lactoilglutationa Liase/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Resveratrol/uso terapêutico , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Quimioterapia Combinada , Indução Enzimática/efeitos dos fármacos , Glutationa/química , Glutationa/uso terapêutico , Glicosilação/efeitos dos fármacos , Hesperidina/química , Humanos , Resistência à Insulina/fisiologia , Lactoilglutationa Liase/antagonistas & inibidores , Camundongos , Estrutura Molecular , Neoplasias Experimentais/metabolismo , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Obesidade/fisiopatologia , Aldeído Pirúvico/química , Aldeído Pirúvico/metabolismo , Resveratrol/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...