Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 535(7612): 430-4, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27398620

RESUMO

Insulin-dependent diabetes is a complex multifactorial disorder characterized by loss or dysfunction of ß-cells. Pancreatic ß-cells differ in size, glucose responsiveness, insulin secretion and precursor cell potential; understanding the mechanisms that underlie this functional heterogeneity might make it possible to develop new regenerative approaches. Here we show that Fltp (also known as Flattop and Cfap126), a Wnt/planar cell polarity (PCP) effector and reporter gene acts as a marker gene that subdivides endocrine cells into two subpopulations and distinguishes proliferation-competent from mature ß-cells with distinct molecular, physiological and ultrastructural features. Genetic lineage tracing revealed that endocrine subpopulations from Fltp-negative and -positive lineages react differently to physiological and pathological changes. The expression of Fltp increases when endocrine cells cluster together to form polarized and mature 3D islet mini-organs. We show that 3D architecture and Wnt/PCP ligands are sufficient to trigger ß-cell maturation. By contrast, the Wnt/PCP effector Fltp is not necessary for ß-cell development, proliferation or maturation. We conclude that 3D architecture and Wnt/PCP signalling underlie functional ß-cell heterogeneity and induce ß-cell maturation. The identification of Fltp as a marker for endocrine subpopulations sheds light on the molecular underpinnings of islet cell heterogeneity and plasticity and might enable targeting of endocrine subpopulations for the regeneration of functional ß-cell mass in diabetic patients.


Assuntos
Ilhotas Pancreáticas/citologia , Animais , Biomarcadores/análise , Diferenciação Celular , Linhagem da Célula/genética , Polaridade Celular , Proliferação de Células , Humanos , Resistência à Insulina , Ilhotas Pancreáticas/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Via de Sinalização Wnt
2.
Bioinformatics ; 35(22): 4834-4836, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31228198

RESUMO

SUMMARY: Despite their fundamental role in various biological processes, the analysis of small RNA sequencing data remains a challenging task. Major obstacles arise when short RNA sequences map to multiple locations in the genome, align to regions that are not annotated or underwent post-transcriptional changes which hamper accurate mapping. In order to tackle these issues, we present a novel profiling strategy that circumvents the need for read mapping to a reference genome by utilizing the actual read sequences to determine expression intensities. After differential expression analysis of individual sequence counts, significant sequences are annotated against user defined feature databases and clustered by sequence similarity. This strategy enables a more comprehensive and concise representation of small RNA populations without any data loss or data distortion. AVAILABILITY AND IMPLEMENTATION: Code and documentation of our R package at http://ibis.helmholtz-muenchen.de/deus/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Software , Perfilação da Expressão Gênica , Genoma , RNA , Análise de Sequência de RNA
3.
Diabetologia ; 61(5): 1167-1179, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29478099

RESUMO

AIMS/HYPOTHESIS: Obesity results from a constant and complex interplay between environmental stimuli and predisposing genes. Recently, we identified the IFN-activated gene Ifi202b as the most likely gene responsible for the obesity quantitative trait locus Nob3 (New Zealand Obese [NZO] obesity 3). The aim of this study was to evaluate the effects of Ifi202b on body weight and adipose tissue biology, and to clarify the functional role of its human orthologue IFI16. METHODS: The impact of Ifi202b and its human orthologue IFI16 on adipogenesis was investigated by modulating their respective expression in murine 3T3-L1 and human Simpson-Golabi-Behmel syndrome (SGBS) pre-adipocytes. Furthermore, transgenic mice overexpressing IFI202b were generated and characterised with respect to metabolic traits. In humans, expression levels of IFI16 in adipose tissue were correlated with several variables of adipocyte function. RESULTS: In mice, IFI202b overexpression caused obesity (Δ body weight at the age of 30 weeks: 10.2 ± 1.9 g vs wild-type mice) marked by hypertrophic fat mass expansion, increased expression of Zfp423 (encoding the transcription factor zinc finger protein [ZFP] 423) and white-selective genes (Tcf21, Tle3), and decreased expression of thermogenic genes (e.g. Cidea, Ucp1). Compared with their wild-type littermates, Ifi202b transgenic mice displayed lower body temperature, hepatosteatosis and systemic insulin resistance. Suppression of IFI202b/IFI16 in pre-adipocytes impaired adipocyte differentiation and triacylglycerol storage. Humans with high levels of IFI16 exhibited larger adipocytes, an enhanced inflammatory state and impaired insulin-stimulated glucose uptake in white adipose tissue. CONCLUSIONS/INTERPRETATION: Our findings reveal novel functions of Ifi202b and IFI16, demonstrating their role as obesity genes. These genes promote white adipogenesis and fat storage, thereby facilitating the development of obesity-associated insulin resistance.


Assuntos
Adipogenia , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Nucleares/fisiologia , Obesidade/genética , Fosfoproteínas/fisiologia , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Peso Corporal , Feminino , Humanos , Inflamação , Resistência à Insulina , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Nucleares/genética , Obesidade/metabolismo , Fenótipo , Fosfoproteínas/genética , Locos de Características Quantitativas , RNA Interferente Pequeno/metabolismo , Termogênese
4.
Am J Physiol Endocrinol Metab ; 314(3): E266-E273, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29138227

RESUMO

The liver is a central regulator of whole body glucose, and lipid homeostasis and hepatokines, like fetuin-A, have been identified as markers and mediators of fatty liver-induced cardiometabolic risk. The closely related protein fetuin-B was shown to be upregulated in the fatty liver and to impact on glucose homeostasis in mice. In the present study we aimed to test the relevance of these findings in humans. In 55 subjects, hepatic mRNA expression of both hepatokines, fetuin-A and fetuin-B, associated positively with liver triglyceride content, whereas only fetuin-A expression associated with the homeostatic model assessment of insulin resistance. In 220 subjects who underwent precise metabolic phenotyping, circulating fetuin-A, but not fetuin-B, associated positively with liver fat content, and negatively with insulin sensitivity, measured during the oral glucose tolerance test (OGTT) and during the euglycemic, hyperinsulinemic clamp. Both circulating fetuin-A and fetuin-B correlated positively with the glucose area under the curve during the OGTT, but after additional adjustment for insulin sensitivity this relationship remained significant only for fetuin-B. In conclusion, despite the fact that the two hepatokines, fetuin-A and fetuin-B, are upregulated in the state of hepatic steatosis in humans, it appears that they differently impact on glucose homeostasis. Our data are in agreement with observations that fetuin-A can alter insulin signaling and that fetuin-B may regulate glucose homeostasis via so far unknown effects, possibly on glucose effectiveness.


Assuntos
Fígado Gorduroso/sangue , Fígado Gorduroso/genética , Fetuína-B , alfa-2-Glicoproteína-HS , Idoso , Estudos de Coortes , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Feminino , Fetuína-B/análise , Fetuína-B/genética , Fetuína-B/metabolismo , Teste de Tolerância a Glucose , Humanos , Resistência à Insulina/fisiologia , Fígado/metabolismo , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Regulação para Cima/genética , alfa-2-Glicoproteína-HS/análise , alfa-2-Glicoproteína-HS/genética , alfa-2-Glicoproteína-HS/metabolismo
5.
Hum Mol Genet ; 25(12): 2588-2599, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27126637

RESUMO

Obesity and ectopic fat disposition are risk factors for metabolic disease. Recent data indicate that IGFBP2 expression in liver is epigenetically inhibited during hepatic steatosis. The aim of this study was to investigate if epigenetic de-regulation of hepatic Igfbp2 occurs already early in life and is associated with increased risk for diet-induced obesity (DIO) during adolescence. Male C57BL/6J mice received a high-fat diet. After 3 weeks on this diet (age of 6 weeks), DIO-susceptible (responder, Resp) and DIO-resistant (non-responder, nResp) mice were identified by early weight gain. At the age of 6 weeks, Resp mice exhibited elevated blood glucose (p < 0.05), plasma insulin (p < 0.01), HOMA-IR and leptin/adiponectin ratio, whereas liver triglycerides were identical but significantly increased (p < 0.01) in Resp mice at 20 weeks of age. Igfbp2 expression was reduced in young Resp compared with nResp mice (p < 0.01), an effect that correlated with elevated DNA methylation of intronic CpG2605 (p < 0.01). The epigenetic inhibition of Igfbp2 was stable over time and preceded DIO and hepatosteatosis in adult mice. In vitro studies demonstrated that selective methylation of CpG2605 significantly reduced reporter activity by ∼85%, indicating that Igfbp2 expression is modulated by methylation. In human whole blood cells, methylation of IGFBP2 at the homologous CpG site was increased in obese men with impaired glucose tolerance. In conclusion, our data show that increased methylation of hepatic Igfbp2 during infancy predicts the development of fatty liver later in life and is linked to deterioration of glucose metabolism.


Assuntos
Metilação de DNA/genética , Fígado Gorduroso/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Obesidade/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Glicemia , Dieta Hiperlipídica , Fígado Gorduroso/sangue , Fígado Gorduroso/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Insulina/sangue , Resistência à Insulina/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Obesidade/sangue , Obesidade/patologia
6.
Diabetes Obes Metab ; 20(7): 1793-1797, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29484808

RESUMO

We performed the largest randomized, placebo-controlled clinical trial to date (N = 112, 12-week intervention) to investigate the effects and safety of resveratrol supplementation on liver fat content and cardiometabolic risk parameters in overweight and obese and insulin-resistant subjects. At baseline the variability in liver fat content was very large, ranging from 0.09% to 37.55% (median, 7.12%; interquartile range, 3.85%-12.94%). Mean (SD) liver fat content was 9.22 (6.85) % in the placebo group and 9.91 (7.76) % in the resveratrol group. During the study liver fat content decreased in the placebo group (-0.7%) but not in the resveratrol group (-0.03%) (differences between groups: P = .018 for the intention-to-treat [ITT] population; N = 54, resveratrol, N = 54, placebo and P = .0077 for the per protocol [PP] population). No effects of resveratrol supplementation on cardiometabolic risk parameters were observed. Resveratrol supplementation was well tolerated and safe. In conclusion, these data suggest that resveratrol supplementation is safe and that it does not considerably impact liver fat content or cardiometabolic risk parameters in humans.


Assuntos
Antioxidantes/uso terapêutico , Suplementos Nutricionais , Resistência à Insulina , Gordura Intra-Abdominal/diagnóstico por imagem , Fígado/diagnóstico por imagem , Hepatopatia Gordurosa não Alcoólica/diagnóstico por imagem , Sobrepeso/metabolismo , Resveratrol/uso terapêutico , Adulto , Idoso , Método Duplo-Cego , Feminino , Hemoglobinas Glicadas/metabolismo , Humanos , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/metabolismo , Espectroscopia de Prótons por Ressonância Magnética
7.
PLoS Genet ; 11(9): e1005506, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26348837

RESUMO

Beta-cell apoptosis and failure to induce beta-cell regeneration are hallmarks of type 2-like diabetes in mouse models. Here we show that islets from obese, diabetes-susceptible New Zealand Obese (NZO) mice, in contrast to diabetes-resistant C57BL/6J (B6)-ob/ob mice, do not proliferate in response to an in-vivo glucose challenge but lose their beta-cells. Genome-wide RNAseq based transcriptomics indicated an induction of 22 cell cycle-associated genes in B6-ob/ob islets that did not respond in NZO islets. Of all genes differentially expressed in islets of the two strains, seven mapped to the diabesity QTL Nob3, and were hypomorphic in either NZO (Lefty1, Apoa2, Pcp4l1, Mndal, Slamf7, Pydc3) or B6 (Ifi202b). Adenoviral overexpression of Lefty1, Apoa2, and Pcp4l1 in primary islet cells increased proliferation, whereas overexpression of Ifi202b suppressed it. We conclude that the identified genes in synergy with obesity and insulin resistance participate in adaptive islet hyperplasia and prevention from severe diabetes in B6-ob/ob mice.


Assuntos
Proliferação de Células/genética , Diabetes Mellitus Experimental/genética , Ilhotas Pancreáticas/citologia , Animais , Humanos , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Fatores de Determinação Direita-Esquerda/genética , Camundongos , Camundongos Endogâmicos C57BL , Locos de Características Quantitativas
8.
Pharmacogenet Genomics ; 27(4): 135-142, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28134748

RESUMO

OBJECTIVE: Inhibition of the renal sodium-glucose cotransporter 2 (SGLT2) is a novel concept in the therapy of diabetes mellitus. In this study, we first assessed whether common single nucleotide polymorphisms (SNPs) in the SGLT2-encoding gene SLC5A2 affect diabetes-related metabolic traits in subjects at risk for type 2 diabetes and, second, whether these have pharmacogenetic relevance by interfering with the response to empagliflozin treatment in patients with type 2 diabetes. PATIENTS AND METHODS: Samples from a metabolically well-phenotyped cross-sectional study population (total N=2600) at increased risk for type 2 diabetes and pooled pharmacogenetic samples from patients from four phase III trials of empagliflozin (in total: 603 receiving empagliflozin, 305 receiving placebo) were genotyped for five common SNPs (minor allele frequencies ≥5%) present in the SLC5A2 gene locus. RESULTS: In the cross-sectional study, none of the SLC5A2 SNPs significantly influenced metabolic traits such as body fat, insulin sensitivity/resistance, insulin release, HbA1c, plasma glucose, or systolic blood pressure when multiple testing was taken into account (all P≥0.0083). Further, no relevant effect on response to treatment with empagliflozin on HbA1c, fasting glucose, weight, or systolic blood pressure was observed for the SNPs tested in the pharmacogenetic study. CONCLUSION: Common genetic variants in the SLC5A2 gene neither affects diabetes-related metabolic traits nor have a clinically relevant impact on response to treatment with the SGLT2 inhibitor empagliflozin.


Assuntos
Compostos Benzidrílicos/administração & dosagem , Diabetes Mellitus Tipo 2/tratamento farmacológico , Glucosídeos/administração & dosagem , Hipoglicemiantes/administração & dosagem , Transportador 2 de Glucose-Sódio/genética , Adulto , Idoso , Compostos Benzidrílicos/farmacocinética , Glicemia/análise , Pressão Sanguínea , Peso Corporal , Estudos Transversais , Diabetes Mellitus Tipo 2/genética , Feminino , Predisposição Genética para Doença , Glucosídeos/farmacocinética , Humanos , Hipoglicemiantes/farmacocinética , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único
9.
Diabetologia ; 59(12): 2622-2631, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27600277

RESUMO

AIMS/HYPOTHESIS: Variations in FTO are the strongest common genetic determinants of adiposity, and may partly act by influencing dopaminergic signalling in the brain leading to altered reward processing that promotes increased food intake. Therefore, we investigated the impact of such an interaction on body composition, and peripheral and brain insulin sensitivity. METHODS: Participants from the Tübingen Family study (n = 2245) and the Malmö Diet and Cancer study (n = 2921) were genotyped for FTO SNP rs8050136 and ANKK1 SNP rs1800497. Insulin sensitivity in the caudate nucleus, an important reward area in the brain, was assessed by fMRI in 45 participants combined with intranasal insulin administration. RESULTS: We found evidence of an interaction between variations in FTO and an ANKK1 polymorphism that associates with dopamine (D2) receptor density. In cases of reduced D2 receptor availability, as indicated by the ANKK1 polymorphism, FTO variation was associated with increased body fat and waist circumference and reduced peripheral insulin sensitivity. Similarly, altered central insulin sensitivity was observed in the caudate nucleus in individuals with the FTO obesity-risk allele and diminished D2 receptors. CONCLUSIONS/INTERPRETATION: The effects of variations in FTO are dependent on dopamine D2 receptor density (determined by the ANKK1 polymorphism). Carriers of both risk alleles might, therefore, be at increased risk of obesity and diabetes.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Proteínas Serina-Treonina Quinases/genética , Receptores de Dopamina D2/genética , Tecido Adiposo/metabolismo , Administração Intranasal , Adulto , Feminino , Predisposição Genética para Doença , Genótipo , Teste de Tolerância a Glucose , Humanos , Insulina/administração & dosagem , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Espectroscopia de Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Obesidade/genética , Polimorfismo de Nucleotídeo Único/genética
10.
Diabetologia ; 58(12): 2877-84, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26404062

RESUMO

AIMS/HYPOTHESIS: Lack of reversal of prediabetes (impaired glucose tolerance and/or impaired fasting glucose) to normal glucose regulation (NGR) during a lifestyle intervention is strongly associated with a higher incidence of diabetes later in life. In the Tübingen Lifestyle Intervention Program (TULIP) we hypothesised that an at-risk phenotype may exist at baseline that associates with this nonresponse to the intervention. METHODS: A total of 120 participants of TULIP with prediabetes at baseline were studied. Participants underwent 9 months of lifestyle intervention and had measurements of insulin secretion and insulin sensitivity during a 75 g OGTT, and measurements of liver fat content by proton magnetic resonance spectroscopy. RESULTS: During the lifestyle intervention, 55% of the participants did not revert to NGR. Even among participants with the largest body fat loss (upper quartile: -6.9 ± 3.3%, mean ± SD), 40% did not revert to NGR. In this regard, we identified at baseline a high-risk phenotype (n = 72) consisting of low disposition index or low insulin sensitivity + nonalcoholic fatty liver disease (NAFLD) and a low-risk phenotype (n = 48, all other traits). While the adjusted decrease in body fat was almost identical between these phenotypes (-5.7 ± 15.3% vs -7.7 ± 15.2%, p = 0.49), the high-risk phenotype had a smaller decrease in adjusted 2 h blood glucose levels (-3.7 ± 20.3% vs -18.5 ± 20.0%, p = 0.0009). In addition, only 31% of the participants with the high-risk phenotype, but 67% with the low-risk phenotype, reverted to NGR (p < 0.0001). The odds ratio for reaching the status NGR was 4.54 (95% CI 2.08, 9.94) for participants having the low-risk phenotype. CONCLUSIONS/INTERPRETATION: Stratification of individuals with prediabetes at baseline into a high-risk and a low-risk phenotype, based on corrected insulin secretion and insulin-resistant NAFLD, may help to determine the effectiveness of a lifestyle intervention to revert individuals to NGR.


Assuntos
Glicemia/metabolismo , Estilo de Vida , Estado Pré-Diabético/terapia , Adulto , Composição Corporal , Estudos Transversais , Gorduras/metabolismo , Feminino , Teste de Tolerância a Glucose , Humanos , Insulina/metabolismo , Resistência à Insulina , Fígado/metabolismo , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Atividade Motora , Hepatopatia Gordurosa não Alcoólica/metabolismo , Fenótipo , Estado Pré-Diabético/sangue , Risco
11.
Biochim Biophys Acta ; 1833(12): 2803-2811, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23886629

RESUMO

Akt kinases are important mediators of the insulin signal, and some Akt substrates are directly involved in glucose homeostasis. Recently, Girdin has been described as an Akt substrate that is expressed ubiquitously in mammals. Cells overexpressing Girdin show an enhanced Akt activity. However, not much is known about Girdin's role in insulin signaling. We therefore analyzed the role of Girdin in primary human myotubes and found a correlation between Girdin expression and insulin sensitivity of the muscle biopsy donors, as measured by a hyperinsulinemic-euglycemic clamp. To understand this finding on a cellular level, we then investigated the function of Girdin in C2C12 mouse myoblasts. Girdin knock-down reduced Akt and insulin receptor substrate-1 phosphorylation. In contrast, stable overexpression of Girdin in C2C12 cells strikingly increased insulin sensitivity through a massive upregulation of the insulin receptor and enhanced tyrosine phosphorylation of insulin receptor substrate-1. Furthermore, Akt and c-Abl kinases were constitutively activated. To investigate medium-term insulin responses we measured glucose incorporation into glycogen. The Girdin overexpressing cells showed a high basal glycogen synthesis that peaked already at 1nM insulin. Taken together, we characterized Girdin as a new and major regulator of the insulin signal in myoblasts and skeletal muscle.


Assuntos
Insulina/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mioblastos/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas de Transporte Vesicular/metabolismo , Adulto , Animais , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Técnicas de Silenciamento de Genes , Glicogênio/biossíntese , Humanos , Insulina/farmacologia , Masculino , Camundongos , Mioblastos/efeitos dos fármacos , Mioblastos/enzimologia , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos
12.
Hum Brain Mapp ; 35(3): 918-28, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23307469

RESUMO

Eating behavior is crucial in the development of obesity and Type 2 diabetes. To further investigate its regulation, we studied the effects of glucose versus water ingestion on the neural processing of visual high and low caloric food cues in 12 lean and 12 overweight subjects by functional magnetic resonance imaging. We found body weight to substantially impact the brain's response to visual food cues after glucose versus water ingestion. Specifically, there was a significant interaction between body weight, condition (water versus glucose), and caloric content of food cues. Although overweight subjects showed a generalized reduced response to food objects in the fusiform gyrus and precuneus, the lean group showed a differential pattern to high versus low caloric foods depending on glucose versus water ingestion. Furthermore, we observed plasma insulin and glucose associated effects. The hypothalamic response to high caloric food cues negatively correlated with changes in blood glucose 30 min after glucose ingestion, while especially brain regions in the prefrontal cortex showed a significant negative relationship with increases in plasma insulin 120 min after glucose ingestion. We conclude that the postprandial neural processing of food cues is highly influenced by body weight especially in visual areas, potentially altering visual attention to food. Furthermore, our results underline that insulin markedly influences prefrontal activity to high caloric food cues after a meal, indicating that postprandial hormones may be potential players in modulating executive control.


Assuntos
Mapeamento Encefálico/métodos , Encéfalo/fisiologia , Comportamento Alimentar/fisiologia , Alimentos , Sobrepeso/fisiopatologia , Glicemia/fisiologia , Peso Corporal , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Mapeamento Encefálico/instrumentação , Feminino , Alimentos/classificação , Glucose/administração & dosagem , Glucose/farmacologia , Humanos , Insulina/sangue , Masculino , Sobrepeso/sangue , Sobrepeso/metabolismo , Fatores de Tempo , Água/administração & dosagem , Água/farmacologia , Adulto Jovem
13.
Am J Physiol Cell Physiol ; 305(8): C877-86, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23926130

RESUMO

Muscle contraction during exercise is a major stimulus for the release of peptides and proteins (myokines) that are supposed to take part in the beneficial adaptation to exercise. We hypothesize that application of an in vitro exercise stimulus as electric pulse stimulation (EPS) to human myotubes enables the investigation of the molecular response to exercise in a clearly defined model. We applied EPS for 24 h to primary human myotubes and studied the whole genome-wide transcriptional response as well as the release of candidate myokines. We observed 183 differentially regulated transcripts with fold changes >1.3. The transcriptional response resembles several properties of the in vivo situation in the skeletal muscle after endurance exercise, namely significant enrichment of pathways associated with interleukin and chemokine signaling, lipid metabolism, and antioxidant defense. Multiplex immunoassays verified the translation of the transcriptional response of several cytokines into high-secretion levels (IL-6, IL-8, CXCL1, LIF, CSF3, IL-1B, and TNF) and the increased secretion of further myokines such as angiopoietin-like 4. Notably, EPS did not induce the release of creatine kinase. Inhibitor studies and immunoblotting revealed the participation of ERK1/2-, JNK-, and NF-κB-dependent pathways in the upregulation of myokines. To conclude, our data highlight the importance of skeletal muscle cells as endocrine cells. This in vitro exercise model is not only suitable to identify exercise-regulated myokines, but it might be applied to primary human myotubes obtained from different muscle biopsy donors to study the molecular mechanisms of the individual response to exercise.


Assuntos
Citocinas/metabolismo , Exercício Físico/fisiologia , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Adulto , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/metabolismo , Proliferação de Células , Células Cultivadas , Quimiocina CXCL1/metabolismo , Fatores Estimuladores de Colônias/metabolismo , Estimulação Elétrica , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fator Inibidor de Leucemia/metabolismo , Masculino , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
14.
Diabetologia ; 56(10): 2176-80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23979484

RESUMO

AIMS/HYPOTHESIS: Prediabetes is a collective term for different subphenotypes (impaired glucose tolerance [IGT] and/or impaired fasting glucose [IFG]) with different pathophysiologies. A positive family history for type 2 diabetes (FHD) is associated with increased risk for type 2 diabetes. We assumed that it would also associate with prediabetes, but wondered whether all subphenotypes are related to a positive family history. METHODS: In a study population of 8,106 non-diabetic individuals of European origin collected from four study centres (normal glucose tolerance, NGT n = 5,482, IFG and/or IGT n = 2,624), we analysed whether having at least one first degree relative with diabetes is associated with prediabetes. The analyses were performed using the same models in each population separately. Afterwards, a meta-analysis was performed. RESULTS: FHD was significantly associated with the risk for prediabetes (IFG and/or IGT, OR 1.40; 95% CI 1.27, 1.54). This association remained significant in multivariable logistic regression models including sex, age and BMI (OR 1.26; 95% CI 1.14, 1.40). When different prediabetic outcomes were considered separately, the association was found for isolated IFG (OR 1.37; 95% CI 1.20, 1.57), isolated IGT (OR 1.25; 95% CI 1.07, 1.46) as well as for the combination IFG+IGT (OR 1.64; 95% CI 1.40, 1.93). After stratification on BMI, association between FHD and prediabetes was seen only in non-obese individuals (BMI < 30 kg/m(2)). CONCLUSIONS/INTERPRETATION: We found that FHD is an important risk factor for prediabetes, especially for combined IGT and IFG. Its relevance seems to be more evident in the non-obese.


Assuntos
Anamnese , Estado Pré-Diabético/epidemiologia , Adulto , Diabetes Mellitus Tipo 2/epidemiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco
15.
Cell Physiol Biochem ; 30(1): 238-46, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22759970

RESUMO

BACKGROUND/AIMS: Impaired insulin action is an early event in the pathogenesis of obesity and type 2-diabetes, and among the metabolic confounders in obese, hyperleptinaemia is constantly present; however its impact on insulin action in the brain and locomotor activity is unknown. METHODS: We examined insulin action by Western Blot analysis and glycogen synthesis in primary astrocytes and brain tissue and detected locomotion in C57BL/6 mice. The insulin-mediated desire to move was evaluated in healthy volunteers and correlated to leptin levels. RESULTS: Leptin treatment led to a significant decrease in insulin-mediated phosphorylation of the insulin receptor and Akt473 which was accompanied by a decline in glycogen synthesis in primary astrocytes and significantly decreased insulin-induced phosphorylation of the insulin receptor and insulin receptor substrate-2 in brain tissues of mice. Intracerebroventricular insulin failed to promote locomotion in the presence of elevated leptin levels. Lean human subjects reported an increase in the desire to move following insulin which failed in obese and there was an inverse correlation between the insulin-mediated desire to move and leptin levels. CONCLUSIONS: Our data suggest a crosstalk of leptin and insulin in the brain which leads to a decline in locomotor activity. This might represent a molecular mechanism in obese to inhibit physical activity.


Assuntos
Astrócitos/metabolismo , Insulina/fisiologia , Leptina/fisiologia , Atividade Motora , Adiponectina/metabolismo , Adulto , Animais , Encéfalo/metabolismo , Proteína C-Reativa/metabolismo , Células Cultivadas , Feminino , Glicogênio/biossíntese , Humanos , Insulina/farmacologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Leptina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais
16.
Microvasc Res ; 81(1): 52-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21112343

RESUMO

Saturated long-chain fatty acids (SFAs) exert unfavourable metabolic effects (lipotoxicity) and induce apoptotic cell death (lipoapoptosis) in certain cell-types. Their contribution to inflammatory cell responses is unclear. We studied the expression of 113 inflammatory genes in human coronary artery endothelial cells (hCAECs) and their regulation by SFAs and unsaturated long-chain fatty acids (UFAs). Gene regulation in hCAECs was assessed with macroarrays, real-time RT-PCR and immunoblotting. Participation of the transcription factor NFκB and the stress kinases JNK and p38 MAPK in gene-regulatory events was examined with pharmacological inhibitors. Based on macroarray data, 59 inflammatory genes were expressed in hCAECs, 14 were regulated by the SFA palmitate. SFA-triggered induction of IL1A, IL6, IL8, CXCL2, CXCL3, CCL20, SPP1 and CEBPB was confirmed by RT-PCR or immunoblotting. All gene inductions were SFA-specific. Using inhibitor SN50, palmitate-induced expression of IL8, CXCL3 and CCL20 was NFκB-dependent (all p<0.05). Furthermore, JNK was involved in palmitate-induced expression of IL1A, IL8, CXCL3, SPP1 and CEBPB as determined with inhibitor SP600125 (all p<0.05). Finally, the effectiveness of the tested fatty acids to induce inflammatory genes was closely reflected by their effectiveness to trigger endoplasmic reticulum stress. In conclusion, hCAECs express a large panel of inflammatory genes with a series of genes being regulated by palmitate and stearate, but not by UFAs. Thus, SFAs represent potential contributors to vascular inflammation.


Assuntos
Vasos Coronários/citologia , Células Endoteliais/metabolismo , Ácidos Graxos/farmacologia , Inflamação/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células Cultivadas , Quimiocina CCL20/genética , Quimiocinas CXC/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células Endoteliais/efeitos dos fármacos , Ácidos Graxos Insaturados/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Perfilação da Expressão Gênica , Humanos , Inflamação/induzido quimicamente , Interleucina-1alfa/genética , Interleucina-1beta/farmacologia , Interleucina-8/genética , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Ácido Palmítico/farmacologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ácidos Esteáricos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
17.
Eur J Clin Invest ; 41(12): 1323-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21615395

RESUMO

BACKGROUND: Peroxisome proliferator-activated receptor δ (PPARδ) activation enhances muscular fatty acid oxidation and oxidative phosphorylation, and muscle's oxidative capacity positively associates with whole-body insulin sensitivity. Therefore, we asked here whether human muscle cell PPARD expression is a determinant of donors' insulin sensitivity. MATERIALS AND METHODS: Skeletal muscle cells derived from 38 nondiabetic donors were differentiated in vitro to myotubes, and gene (mRNA) expression was quantified by real-time RT-PCR. Donors' insulin sensitivity was calculated from plasma insulin and glucose levels during oral glucose tolerance test (OGTT) and hyperinsulinemic-euglycemic clamp. RESULTS: Basal myotube PPARD expression was closely related to the expression of its target genes PDK4 and ANGPTL4 (P = 0·0312 and P = 0·0003, respectively). Basal PPARD, PDK4 and ANGPTL4 expression levels were not associated with donors' insulin sensitivity (P > 0·2, all). Treatment of myotubes with a selective high-affinity PPARδ agonist (GW501516) did not change mean PPARD, but enhanced mean PDK4 and ANGPTL4 expression 13- and 16-fold, respectively (P < 0·0001, both). The individual PDK4 and ANGPTL4 expression levels reached upon GW501516 treatment were associated with donors' insulin sensitivity neither (P > 0·2, both). However, GW501516-mediated fold increments in PDK4 and ANGPTL4 expression, reflecting PPARδ responsiveness, were positively associated with donors' insulin sensitivity derived from OGTT (P = 0·0182 and P = 0·0231, respectively) and hyperinsulinemic-euglycemic clamp (P = 0·0046 and P = 0·0258, respectively). CONCLUSIONS: Using a highly selective pharmacological tool, we show here that the individual responsiveness of human muscle cell PPARδ, rather than the absolute PPARD expression level, represents a major determinant of insulin sensitivity.


Assuntos
Resistência à Insulina/fisiologia , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , PPAR delta/metabolismo , Tiazóis/metabolismo , Glicemia/metabolismo , Índice de Massa Corporal , Células Cultivadas/metabolismo , Feminino , Regulação da Expressão Gênica , Teste de Tolerância a Glucose , Humanos , Resistência à Insulina/genética , Metabolismo dos Lipídeos/genética , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Regressão
18.
J Clin Endocrinol Metab ; 106(2): e982-e989, 2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33277657

RESUMO

OBJECTIVE: Elevated plasma glutamate levels are associated with an increased risk of cardiovascular disease (CVD). Because plasma glutamate levels are also strongly associated with visceral adiposity, nonalcoholic fatty liver disease, insulin resistance, and high circulating levels of branched-chain amino acids (BCAAs), it is unknown to what extent elevated circulating glutamate is an independent marker of an increased risk of atherosclerosis. METHODS: Plasma levels of glutamate and BCAAs were measured in 102 individuals who were precisely phenotyped for body fat mass and distribution (magnetic resonance [MR] tomography), liver fat content (1H-MR spectroscopy), insulin sensitivity (oral glucose tolerance test and hyperinsulinemic, euglycemic clamp [N = 57]), and carotid intima media thickness (cIMT). RESULTS: Plasma glutamate levels, adjusted for age, sex, body fat mass, and visceral fat mass, correlated positively with liver fat content and cIMT (all std ß ≥ .22, all P ≤ .023) and negatively with insulin sensitivity (std ß ≤ -.31, P ≤ .002). Glutamate levels also were associated with cIMT, independently of additional adjustment for liver fat content, insulin sensitivity and BCAAs levels (std ß ≥ .24, P ≤ .02). Furthermore, an independent positive association of glutamate and interleukin-6 (IL-6) levels was observed (N = 50; std ß = .39, P = .03). Although glutamate, adjusted for age, sex, body fat mass, and visceral fat mass, also correlated positively with cIMT in this subgroup (std ß = .31, P = .02), after additional adjustment for the parameters liver fat content, insulin sensitivity, BCAAs, or IL-6 levels, adjustment for IL-6 most strongly attenuated this relationship (std ß = .28, P = .05). CONCLUSIONS: Elevated plasma glutamate levels are associated with increased cIMT, independently of established CVD risk factors, and this relationship may in part be explained by IL-6-associated subclinical inflammation.


Assuntos
Adiposidade , Aterosclerose/diagnóstico , Biomarcadores/sangue , Espessura Intima-Media Carotídea , Ácido Glutâmico/sangue , Resistência à Insulina , Gordura Intra-Abdominal , Aterosclerose/sangue , Estudos Transversais , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico
19.
Nat Commun ; 12(1): 6486, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34759311

RESUMO

The hepatokine follistatin is elevated in patients with type 2 diabetes (T2D) and promotes hyperglycemia in mice. Here we explore the relationship of plasma follistatin levels with incident T2D and mechanisms involved. Adjusted hazard ratio (HR) per standard deviation (SD) increase in follistatin levels for T2D is 1.24 (CI: 1.04-1.47, p < 0.05) during 19-year follow-up (n = 4060, Sweden); and 1.31 (CI: 1.09-1.58, p < 0.01) during 4-year follow-up (n = 883, Finland). High circulating follistatin associates with adipose tissue insulin resistance and non-alcoholic fatty liver disease (n = 210, Germany). In human adipocytes, follistatin dose-dependently increases free fatty acid release. In genome-wide association study (GWAS), variation in the glucokinase regulatory protein gene (GCKR) associates with plasma follistatin levels (n = 4239, Sweden; n = 885, UK, Italy and Sweden) and GCKR regulates follistatin secretion in hepatocytes in vitro. Our findings suggest that GCKR regulates follistatin secretion and that elevated circulating follistatin associates with an increased risk of T2D by inducing adipose tissue insulin resistance.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Folistatina/sangue , Proteínas Adaptadoras de Transdução de Sinal/sangue , Tecido Adiposo/metabolismo , Estudo de Associação Genômica Ampla , Hepatócitos/metabolismo , Humanos , Resistência à Insulina/fisiologia , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/sangue
20.
J Cell Mol Med ; 14(4): 903-13, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20569275

RESUMO

Among the multitude of dysregulated signalling mechanisms that comprise insulin resistance in divergent organs, the primary events in the development of type 2 diabetes are not well established. As protein kinase C (PKC) activation is consistently present in skeletal muscle of obese and insulin resistant subjects, we generated a transgenic mouse model that overexpresses constitutively active PKC-beta(2) in skeletal muscle to test whether activation of PKC is sufficient to cause an aversive whole-body phenotype. Upon this genetic modification, increased serine phosphorylation in Irs1 was observed and followed by impaired (3)H-deoxy-glucose uptake and muscle glycogen content, and transgenic mice exhibited insulin and glucose intolerance as they age. Muscle histochemistry revealed an increase in lipid deposition (intramyocellular lipids), and transgenic mice displayed impaired expression of transcriptional regulators of genes involved in fatty acid oxidation (peroxisome proliferator-activated receptor-gamma, PGC-1beta, acyl-CoA oxidase) and lipolysis (hormone-sensitive lipase). In this regard, muscle of transgenic mice exhibited a reduced capacity to oxidize palmitate and contained less mitochondria as determined by citrate synthase activity. Moreover, the phenotype included a profound decrease in the daily running distance, intra-abdominal and hepatic fat accumulation and impaired insulin action in the brain. Together, our data suggest that activation of a classical PKC in skeletal muscle as present in the pre-diabetic state is sufficient to cause disturbances in whole-body glucose and lipid metabolism followed by profound alterations in oxidative capacity, ectopic fat deposition and physical activity.


Assuntos
Encéfalo/patologia , Fígado Gorduroso/enzimologia , Resistência à Insulina , Atividade Motora , Músculo Esquelético/enzimologia , Proteína Quinase C/metabolismo , Animais , Encéfalo/metabolismo , Fígado Gorduroso/patologia , Insulina/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/patologia , Especificidade de Órgãos , Oxirredução , Condicionamento Físico Animal , Proteína Quinase C beta , Transdução de Sinais , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA