Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Diabetes ; 70(9): 1985-1999, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34226282

RESUMO

Adipose tissue (AT) is a key metabolic organ which functions are rhythmically regulated by an endogenous circadian clock. Feeding is a "zeitgeber" aligning the clock in AT with the external time, but mechanisms of this regulation remain largely unclear. We tested the hypothesis that postprandial changes of the hormone insulin directly entrain circadian clocks in AT and investigated a transcriptional-dependent mechanism of this regulation. We analyzed gene expression in subcutaneous AT (SAT) of obese subjects collected before and after the hyperinsulinemic-euglycemic clamp or control saline infusion (SC). The expressions of core clock genes PER2, PER3, and NR1D1 in SAT were differentially changed upon insulin and saline infusion, suggesting insulin-dependent clock regulation. In human stem cell-derived adipocytes, mouse 3T3-L1 cells, and AT explants from mPer2Luc knockin mice, insulin induced a transient increase of the Per2 mRNA and protein expression, leading to the phase shift of circadian oscillations, with similar effects for Per1 Insulin effects were dependent on the region between -64 and -43 in the Per2 promoter but not on CRE and E-box elements. Our results demonstrate that insulin directly regulates circadian clocks in AT and isolated adipocytes, thus representing a primary mechanism of feeding-induced AT clock entrainment.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Relógios Circadianos/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Insulina/farmacologia , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos
2.
Cells ; 10(5)2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946738

RESUMO

Liver fibrosis is a critical complication of obesity-induced fatty liver disease. Wnt1 inducible signaling pathway protein 1 (WISP1/CCN4), a novel adipokine associated with visceral obesity and insulin resistance, also contributes to lung and kidney fibrosis. The aim of the present study was to investigate the role of CCN4 in liver fibrosis in severe obesity. For this, human liver biopsies were collected from 35 severely obese humans (BMI 42.5 ± 0.7 kg/m2, age 46.7 ± 1.8 y, 25.7% males) during bariatric surgery and examined for the expression of CCN4, fibrosis, and inflammation markers. Hepatic stellate LX-2 cells were treated with human recombinant CCN4 alone or in combination with LPS or transforming growth factor beta (TGF-ß) and examined for fibrosis and inflammation markers. CCN4 mRNA expression in the liver positively correlated with BMI and expression of fibrosis markers COL1A1, COL3A1, COL6A1, αSMA, TGFB1, extracellular matrix turnover enzymes TIMP1 and MMP9, and the inflammatory marker ITGAX/CD11c. In LX-2 cells, the exposure to recombinant CCN4 caused dose-dependent induction of MMP9 and MCP1. CCN4 potentiated the TGF-ß-mediated induction of COL3A1, TIMP1, and MCP1 but showed no interaction with LPS treatment. Our results suggest a potential contribution of CCN4 to the early pathogenesis of obesity-associated liver fibrosis.


Assuntos
Proteínas de Sinalização Intercelular CCN/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Obesidade Mórbida/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Adulto , Proteínas de Sinalização Intercelular CCN/genética , Antígenos CD11/genética , Antígenos CD11/metabolismo , Linhagem Celular , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Colágeno/genética , Colágeno/metabolismo , Feminino , Humanos , Fígado/patologia , Cirrose Hepática/etiologia , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Obesidade Mórbida/complicações , Proteínas Proto-Oncogênicas/genética , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
3.
Metabol Open ; 6: 100034, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32812932

RESUMO

BACKGROUND: Galectin-1, haptoglobin, and nesfatin-1 have recently emerged as promising biomarkers implicated in immunometabolism. However, whether single blood measurements of these analytes could be suitable for large-scale human studies has not yet been evaluated. METHODS: The concentrations of galectin-1, haptoglobin, and nesfatin-1 were measured over a 4-month period in 207 healthy adults with median age of 56.7 years. Biomarker intra-individual reproducibility was assessed based on calculation of intraclass correlation coefficients (ICCs) and examining Bland-Altman plots. RESULTS: The overall ICCs were excellent for nesfatin-1 (ICC: 0.89 (95% CI: 0.86, 0.92), and good for galectin-1 and haptoglobin (ICCs: 0.70 (95% CI: 0.61, 0.77) and 0.67 (95% CI: 0.57, 0.74), respectively). Bland-Altman plots supported a high level of agreement between repeated biomarker measurements. CONCLUSIONS: Assay measurements of galectin-1, haptoglobin, and nesfatin-1 showed good to excellent within-subject reproducibility over a 4-month period, indicating that they may serve as feasible and reliable biomarkers for assessing metabolic inflammation in population research.

4.
Peptides ; 125: 170231, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31870938

RESUMO

GIP was proposed to play a key role in the development of non- alcoholic fatty liver disease (NAFLD) in response to sugar intake. Isomaltulose, is a 1,6-linked glucose-fructose dimer which improves glucose homeostasis and prevents NAFLD compared to 1,2-linked sucrose by reducing glucose-dependent insulinotropic peptide (GIP) in mice. We compared effects of sucrose vs. isomaltulose on GIP and glucagon-like peptide-1 (GLP-1) secretion, hepatic insulin clearance (HIC) and insulin sensitivity in normal (NGT), impaired glucose tolerant (IGT) and Type 2 diabetes mellitus (T2DM) participants. A randomized crossover study was performed in 15 NGT, 10 IGT and 10 T2DM subjects. In comparison to sucrose, peak glucose concentrations were reduced by 2.3, 2.1 and 2.5 mmol/l (all p < 0.05) and insulin levels were 88% (p < 0.01, NGT), 32% (p < 0.05, IGT) and 55% (T2DM) lower after the isomaltulose load. Postprandial GIPiAUC concentrations were decreased (56%, p < 0.01 in NGT; 42%, p < 0.05 in IGT and 40%,p < 0.001 in T2DM) whereas GLP-1iAUC was 77%, 85% and 85% higher compared to sucrose (p < 0.01), respectively. This resulted in ∼35 - 50% improved insulin sensitivity and reduced insulinogenic index after isomaltulose, which correlated closely with improved HIC, respectively (r = 0.62, r=-0.70; p < 0.001). HIC was inversely related to GIP (r=-0.44, p < 0.001) and positively related to GLP-1 levels (r = 0.40, p = 0.001). CONCLUSION: Endogenously released GIP correlated with reduced, and GLP-1 with increased hepatic insulin extraction. Increased peripheral insulin levels may contribute to insulin resistance and obesity. We propose that the unfavorable effects of high glycemic index Western diets are related to increased GIP-release and reduced HIC.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Polipeptídeo Inibidor Gástrico/farmacologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Incretinas/farmacologia , Resistência à Insulina , Insulina/metabolismo , Fígado/metabolismo , Adulto , Estudos de Casos e Controles , Estudos Cross-Over , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Feminino , Humanos , Isomaltose/administração & dosagem , Isomaltose/análogos & derivados , Fígado/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Sacarose/administração & dosagem
5.
Clin Nutr ; 39(3): 862-869, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-30967307

RESUMO

BACKGROUND & AIMS: Pro-inflammatory biomarkers are well-established contributors to insulin resistance and represent valid targets for diabetes management and prevention. Yet, little is known whether nutrition could play a role in modulating various aspects of immune-inflammatory responses. Our aim is to assess the effect of isocaloric animal and plant protein dietary interventions on selected biomarkers representing various immune-inflammatory pathways. METHODS: We enrolled 37 participants with type 2 diabetes (age 64 ± 6 years, body mass index 30.2 ± 3.6 kg/m2, glycated hemoglobin 7.0 ± 0.6%) who underwent an either high-animal protein (AP) or high-plant protein (PP) diet (30 E% protein, 40 E% carbohydrates, 30 E% fat) for 6-weeks. Clinical examinations were performed at beginning and end of the study. Levels of pro-inflammatory adipokines [chemerin, progranulin], cytokines [tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), soluble urokinase-type plasminogen activator receptor (suPAR), transforming growth factor beta 1 (TGF-ß1)], and proteins [calprotectin, lactoferrin and growth differentiation factor 15 (GDF-15)] were determined in blood serum using enzyme-linked immunosorbent assay. RESULTS: Chemerin and progranulin concentrations decreased following AP and PP diets. TGF-ß1 increased in AP and decreased in PP, whereas calprotectin increased in PP and decreased in AP. No statistically significant differences in the concentrations of IL-6, TNF-α, suPAR, lactoferrin and GDF-15 could be seen in either of the protein diet arms. CONCLUSIONS: These results suggest that both AP and PP diets may effectively reduce the levels of the pro-inflammatory adipokines chemerin and progranulin. The effects on the additional immune-inflammatory biomarkers seem to be more complex. CLINICAL TRIAL REGISTRY NUMBER: NCT02402985 (ww.clinicaltrials.gov).


Assuntos
Proteínas Animais da Dieta/sangue , Proteínas Animais da Dieta/imunologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/imunologia , Inflamação/sangue , Proteínas de Vegetais Comestíveis/sangue , Proteínas de Vegetais Comestíveis/imunologia , Adipocinas/sangue , Idoso , Biomarcadores/sangue , Dieta Rica em Proteínas/métodos , Feminino , Humanos , Complexo Antígeno L1 Leucocitário/sangue , Masculino , Pessoa de Meia-Idade , Fator de Crescimento Transformador beta/sangue
6.
Am J Clin Nutr ; 108(2): 279-291, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29982277

RESUMO

Background: Meal composition regulates the postprandial response of pancreatic and gastrointestinal hormones and plays an important role in patients with type 2 diabetes (T2D). Proteins have glucagon and insulinotropic effects, which may differ depending on amino acid composition, form of intake, and rate of digestibility and absorption. Objective: The aim of this study was to test effects of isolated pea protein-based (PP) compared with casein protein-based (CP) meals differing in amino acid compositions on endocrine responses to meal tolerance tests (MTTs) in patients with T2D. Design: Thirty-seven individuals with T2D [mean ± SD age: 64 ± 6 y; mean ± SD body mass index (kg/m2): 30.2 ± 3.6; mean ± SD glycated hemoglobin: 7.0% ± 0.6%] were randomly assigned to receive either high-animal-protein (∼80% of total protein) or high-plant-protein (∼72% of total protein) diets (30% of energy from protein, 40% of energy from carbohydrate, 30% of energy from fat) for 6 wk. MTTs were performed at study onset and after 6 wk. Participants received standardized high-protein (30% of energy) meals 2 times/d containing either CP-rich (∼85% wt:wt) or PP-rich (∼95% wt:wt) foods. Results: The CP and PP meals produced differences in insulin, C-peptide, glucagon, and glucose-dependent insulinotropic peptide (GIP) release. Total areas under the curve after CP were significantly lower than after the PP lunch by 40% for insulin and 23% for glucagon. Indexes of insulin sensitivity and secretion were significantly improved for the second CP MTT. This was accompanied by differential rates of appearance of amino acids. The ingestion of PP resulted in significant increases in amino acids after both meals, with a decline between meals. By contrast, CP intake resulted in increases in most amino acids after breakfast, which remained elevated but did not increase further after lunch. Conclusions: PP elicits greater postprandial increases in glucagon than does CP and consequently requires higher insulin to control glucose metabolism, which appears to be related to the rate of amino acid appearance. The metabolic impact of protein quality could be used as a strategy to lower insulin needs in patients with T2D. This trial was registered at www.clinicaltrials.gov as NCT02402985.


Assuntos
Aminoácidos/sangue , Diabetes Mellitus Tipo 2/metabolismo , Dieta Rica em Proteínas , Glucagon/metabolismo , Insulina/metabolismo , Idoso , Glicemia/análise , Feminino , Polipeptídeo Inibidor Gástrico/sangue , Peptídeo 1 Semelhante ao Glucagon/análise , Humanos , Masculino , Pessoa de Meia-Idade
7.
Diabetologia ; 61(9): 2054-2065, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29754289

RESUMO

AIMS/HYPOTHESIS: Wingless-type (Wnt) inducible signalling pathway protein-1 (WISP1) has been recently identified as a proinflammatory adipokine. We examined whether WISP1 expression and circulating levels are altered in type 2 diabetes and whether WISP1 affects insulin signalling in muscle cells and hepatocytes. METHODS: Serum and visceral adipose tissue (VAT) biopsies, for analysis of circulating WISP1 levels by ELISA and WISP1 mRNA expression by real-time quantitative RT-PCR, were collected from normal-weight men (control group, n = 33) and obese men with (n = 46) and without type 2 diabetes (n = 56) undergoing surgery. Following incubation of primary human skeletal muscle cells (hSkMCs) and murine AML12 hepatocytes with WISP1 and insulin, insulin signalling was analysed by western blotting. The effect of WISP1 on insulin-stimulated glycogen synthesis and gluconeogenesis was investigated in hSkMCs and murine hepatocytes, respectively. RESULTS: Circulating WISP1 levels were higher in obese men (independent of diabetes status) than in normal-weight men (mean [95% CI]: 70.8 [55.2, 86.4] ng/l vs 42.6 [28.5, 56.6] ng/l, respectively; p < 0.05). VAT WISP1 expression was 1.9-fold higher in obese men vs normal-weight men (p < 0.05). Circulating WISP1 levels were positively associated with blood glucose in the OGTT and circulating haem oxygenase-1 and negatively associated with adiponectin levels. In hSkMCs and AML12 hepatocytes, recombinant WISP1 impaired insulin action by inhibiting phosphorylation of insulin receptor, Akt and its substrates glycogen synthase kinase 3ß, FOXO1 and p70S6 kinase, and inhibiting insulin-stimulated glycogen synthesis and suppression of gluconeogenic genes. CONCLUSIONS/INTERPRETATION: Circulating WISP1 levels and WISP1 expression in VAT are increased in obesity independent of glycaemic status. Furthermore, WISP1 impaired insulin signalling in muscle and liver cells.


Assuntos
Proteínas de Sinalização Intercelular CCN/metabolismo , Hepatócitos/metabolismo , Resistência à Insulina/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Glicemia/metabolismo , Proteínas de Sinalização Intercelular CCN/sangue , Ensaio de Imunoadsorção Enzimática , Humanos , Gordura Intra-Abdominal/metabolismo , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas/sangue , Receptor de Insulina/metabolismo , Transdução de Sinais
8.
Horm Metab Res ; 49(5): 343-349, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28351093

RESUMO

Angiopoietin-like protein 8 (ANGPTL8)/betatrophin expression in visceral adipose tissue and associations with circulating fatty acid profile have not yet been investigated.Forty subjects were included in a cross-sectional study, 57 in a dietary weight reduction intervention. Circulating Angiopoietin-like protein 8/betatrophin was measured in all subjects. Liver and adipose tissue were sampled and plasma fatty acids and tissue Angiopoietin-like protein 8/betatrophin expression were evaluated in the cross-sectional study. In the intervention study oral glucose testing and liver magnetic resonance scanning at baseline and after 6 months were performed. Angiopoietin-like protein 8/betatrophin mRNA was increased in visceral compared to subcutaneous adipose tissue (p<0.001). Circulating ANGPTL8/betatrophin correlated with liver steatosis (r=0.42, p=0.047), triacylglycerols (r=0.34, p=0.046), saturated (r=0.43, p=0.022), monounsaturated (r=0.51, p=0.007), and polyunsaturated fatty acids (r=-0.53, p=0.004). In the intervention study, baseline Angiopoietin-like protein 8/betatrophin correlated with age (r=0.32, p=0.010) and triacylglycerols (r=0.30, p=0.02) and was increased with hepatic steatosis (p=0.033). Weight loss reduced liver fat by 45% and circulating Angiopoietin-like protein 8/betatrophin by 11% (288±17 vs. 258±17 pg/ml; p=0.015). Angiopoietin-like protein 8/betatrophin is related to liver steatosis, while visceral adipose tissue represents an additional site of expression in humans.


Assuntos
Proteínas Semelhantes a Angiopoietina/genética , Fígado Gorduroso/genética , Gordura Intra-Abdominal/metabolismo , Hormônios Peptídicos/genética , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina/metabolismo , Estudos de Coortes , Dieta , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/genética , Hormônios Peptídicos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
Minerva Endocrinol ; 41(4): 456-68, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27007919

RESUMO

BACKGROUND: Acarbose, an alpha-glucosidase inhibitor, unexpectedly reduced the incidence of hypertension and cardiovascular endpoints in the STOP-NIDDM study. Based on the growing evidence of a link between vasoregulatory peptides and metabolic traits, we hypothesized that changes of the Glycemic Index by acarbose may modulate vasoregulatory peptide levels via regulation of postprandial metabolism. METHODS: Subjects with type 2 diabetes and with metabolic syndrome were treated with acarbose (12 weeks, 300mg/d) in a double-blind, placebo-controlled, cross-over intervention. Changes in fasting and postprandial levels of midregional pro-atrial natriuretic peptide (MR-proANP), C-terminal pro-endothelin-1 (CT-proET-1) and midregional pro-adrenomedullin (MR-proADM), WNT1 Inducible Signaling Pathway Protein 1 (WISP1) as well as fasting and postprandial glucose/insulin levels in the liquid meal test were assessed. RESULTS: Acarbose strongly decreased postprandial insulin concentrations in subjects with metabolic syndrome (P=0.004), and postprandial glucose excursions in both groups. Postprandial MR-proANP and CT-proET-1 levels increased after acarbose treatment (P<0.01 and P<0.05, respectively) in subjects with metabolic syndrome only. No effect of acarbose treatment on MR-prADM was observed in both groups. All three peptides were correlated with each over, but neither with insulin sensitivity in euglycemic clamps, nor with adiponectin levels. WISP1 decreased after acarbose treatment in subjects with metabolic syndrome. CONCLUSIONS: Plasma MR- proANP and CT-proET-1 concentrations, but not MR-prADM concentrations, were affected by treatment with acarbose over 12 weeks. Our findings provide new possible mechanisms of acarbose action in diabetes and metabolic syndrome.


Assuntos
Acarbose/uso terapêutico , Cardiotônicos/uso terapêutico , Proteínas da Matriz Extracelular/sangue , Peptídeo Intestinal Vasoativo/sangue , Estudos Cross-Over , Diabetes Mellitus Tipo 2/sangue , Método Duplo-Cego , Feminino , Humanos , Insulina/sangue , Masculino , Síndrome Metabólica/sangue , Pessoa de Meia-Idade
10.
Am J Physiol Endocrinol Metab ; 309(12): E968-80, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26487005

RESUMO

In humans and rodents, risk of metabolic syndrome is sexually dimorphic, with an increased incidence in males. Additionally, the protective role of female gonadal hormones is ostensible, as prevalence of type 2 diabetes mellitus (T2DM) increases after menopause. Here, we investigated the influence of estrogen (E2) on the onset of T2DM in female New Zealand obese (NZO) mice. Diabetes prevalence (defined as blood glucose levels >16.6 mmol/l) of NZO females on high-fat diet (60 kcal% fat) in week 22 was 43%. This was markedly dependent on liver fat content in week 10, as detected by computed tomography. Only mice with a liver fat content >9% in week 10 plus glucose levels >10 mmol/l in week 9 developed hyperglycemia by week 22. In addition, at 11 wk, diacylglycerols were elevated in livers of diabetes-prone mice compared with controls. Hepatic expression profiles obtained from diabetes-prone and -resistant mice at 11 wk revealed increased abundance of two transcripts in diabetes-prone mice: Mogat1, which catalyzes the synthesis of diacylglycerols from monoacylglycerol and fatty acyl-CoA, and the fatty acid transporter Cd36. E2 treatment of diabetes-prone mice for 10 wk prevented any further increase in liver fat content and reduced diacylglycerols and the abundance of Mogat1 and Cd36, leading to a reduction of diabetes prevalence and an improved glucose tolerance compared with untreated mice. Our data indicate that early elevation of hepatic Cd36 and Mogat1 associates with increased production and accumulation of triglycerides and diacylglycerols, presumably resulting in reduced hepatic insulin sensitivity and leading to later onset of T2DM.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Estrogênios/farmacologia , Ácidos Graxos/metabolismo , Gordura Intra-Abdominal/metabolismo , Fígado/metabolismo , Obesidade/metabolismo , Animais , Feminino , Gordura Intra-Abdominal/efeitos dos fármacos , Fígado/efeitos dos fármacos , Prevalência , Ratos
11.
Cell Cycle ; 14(14): 2293-300, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25945652

RESUMO

Diabetes mellitus type 2 (T2DM), insulin therapy, and hyperinsulinemia are independent risk factors of liver cancer. Recently, the use of a novel inhibitor of insulin degrading enzyme (IDE) was proposed as a new therapeutic strategy in T2DM. However, IDE inhibition might stimulate liver cell proliferation via increased intracellular insulin concentration. The aim of this study was to characterize effects of inhibition of IDE activity in HepG2 hepatoma cells and to analyze liver specific expression of IDE in subjects with T2DM. HepG2 cells were treated with 10 nM insulin for 24 h with or without inhibition of IDE activity using IDE RNAi, and cell transcriptome and proliferation rate were analyzed. Human liver samples (n = 22) were used for the gene expression profiling by microarrays. In HepG2 cells, IDE knockdown changed expression of genes involved in cell cycle and apoptosis pathways. Proliferation rate was lower in IDE knockdown cells than in controls. Microarray analysis revealed the decrease of hepatic IDE expression in subjects with T2DM accompanied by the downregulation of the p53-dependent genes FAS and CCNG2, but not by the upregulation of proliferation markers MKI67, MCM2 and PCNA. Similar results were found in the liver microarray dataset from GEO Profiles database. In conclusion, IDE expression is decreased in liver of subjects with T2DM which is accompanied by the dysregulation of p53 pathway. Prolonged use of IDE inhibitors for T2DM treatment should be carefully tested in animal studies regarding its potential effect on hepatic tumorigenesis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Insulina/farmacologia , Insulisina/metabolismo , Fígado/metabolismo , Adulto , Idoso , Apoptose/efeitos dos fármacos , Estudos de Coortes , Ciclina G2/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Feminino , Perfilação da Expressão Gênica , Células Hep G2 , Humanos , Insulisina/antagonistas & inibidores , Insulisina/genética , Antígeno Ki-67/metabolismo , Masculino , Pessoa de Meia-Idade , Componente 2 do Complexo de Manutenção de Minicromossomo/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Interferência de RNA , Transcriptoma/efeitos dos fármacos , Receptor fas/metabolismo
12.
Diabetologia ; 58(8): 1759-68, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25994074

RESUMO

AIMS/HYPOTHESIS: Obesity is associated with elevated monocyte chemoattractant protein-1 (MCP-1), a proinflammatory chemokine related to diabetes and cardiovascular disease. Since obesity is triggered by energy dense diets, we hypothesised that nutrient induced intestinal hormones such as glucose-dependent insulinotropic peptide (GIP) may directly stimulate the release of chemokines from adipose tissue and induce low-grade inflammation. METHODS: GIP effects on gene expression and secretion of inflammatory markers were studied by microarray analysis and PCR from human subcutaneous fat biopsies of slightly obese but healthy volunteers in the metabolic ward of German Institute of Human Nutrition, Department of Clinical Nutrition, Potsdam-Rehbrücke. To allocate the participants to the study arms they were numbered in order of their recruitment and then assigned to the groups by a random number generator. In a randomised, single-blind (participants) crossover design, the participants received GIP infusions in postprandial concentrations (2 pmol kg(-1) min(-1)) or saline (154 mmol/l NaCl) infusions for 240 min either alone, in combination with hyperinsulinaemic-euglycaemic (EU) or hyperinsulinaemic-hyperglycaemic (HC) clamps. Possible mechanisms of GIP effects were investigated in single and co-cultures of macrophage and adipocyte cell lines and in primary human monocytes, macrophages and adipocytes. RESULTS: A total of 17 participants were randomised to the following groups: EU with GIP infusion (n = 9); EU with NaCl infusion (n = 9); HC with GIP infusion (n = 8); HC with NaCl infusion (n = 8); sole GIP infusion (n = 11) and sole placebo infusion (n = 11). All 17 individuals were analysed. The study is completed. In human subcutaneous adipose tissue (hSCAT), infusions of GIP significantly increased inflammatory chemokine and cytokine gene networks in transcriptomic microarray analyses. Particularly MCP-1 (180 ± 26%), MCP-2 (246 ± 58%) and IL-6 (234 ± 40%) mRNA levels in adipose tissue as well as circulating plasma concentrations of MCP-1 (165 ± 12 vs 135 ± 13 pg/ml; GIP vs saline after 240 min; p < 0.05 for all variables) in humans increased independently of circulating insulin or glucose plasma concentrations. GIP stimulation increased Mcp-1 mRNA-expression in co-cultures of differentiated 3T3L1-adipocytes and RAW 264.7 macrophages but not in the isolated cell lines. Similarly, GIP increased MCP-1 transcripts in co-cultures of primary human macrophages with human adipocytes. GIP receptor (GIPR) transcripts were present in primary monocytes and the different cell lines and induced activation of extracellular related kinase (ERK) as well as increases in cAMP, indicating functional receptors. CONCLUSIONS/INTERPRETATION: Our findings suggest that the nutrient induced gut hormone GIP may initiate adipose tissue inflammation by triggering a crosstalk of adipocytes and macrophages involving MCP-1. TRIAL REGISTRATION: ClinicalTrials.gov NCT00774488. FUNDING: This work was supported by the German Research Foundation (DFG): grant No. Pf164/021002.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Dieta , Polipeptídeo Inibidor Gástrico/farmacologia , Expressão Gênica/efeitos dos fármacos , Inflamação/metabolismo , Obesidade/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Adolescente , Adulto , Idoso , Células Cultivadas , Quimiocina CCL2/sangue , Quimiocina CCL2/genética , Estudos Cross-Over , Polipeptídeo Inibidor Gástrico/sangue , Humanos , Inflamação/sangue , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Transdução de Sinais/efeitos dos fármacos , Método Simples-Cego , Adulto Jovem
13.
Peptides ; 65: 12-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25620618

RESUMO

Obesity, type 2 diabetes and associated metabolic diseases are characterized by low-grade systemic inflammation which involves interplay of nutrition and monocyte/macrophage functions. We suggested that some factors such as nutrient components, neuropeptides involved in the control of gastrointestinal functions, and gastrointestinal hormones might influence immune cell functions and in this way contribute to the disease pathogenesis. The aim of this study was to investigate the mRNA expression of twelve nutrition-associated receptors in peripheral blood mononuclear cells (PBMC), isolated monocytes and monocyte-derived macrophages and their regulation under the switching from the high-carbohydrate low-fat diet to the low-carbohydrate high-fat (LC/HFD) isocaloric diet in healthy humans. The mRNA expression of receptors for short chain fatty acids (GPR41, GPR43), bile acids (TGR5), incretins (GIPR, GLP1R), cholecystokinin (CCKAR), neuropeptides VIP and PACAP (VIPR1, VIPR2), and neurotensin (NTSR1) was detected in PBMC and monocytes, while GPR41, GPR43, GIPR, TGR5, and VIPR1 were found in macrophages. Correlations of the receptor expression in monocytes with a range of metabolic and inflammatory markers were found. In non-obese subjects, the dietary switch to LC/HFD induced the increase of GPR43 and VIPR1 expression in monocytes. No significant differences of receptor expression between normal weight and moderately obese subjects were found. Our study characterized for the first time the expression pattern of nutrition-associated receptors in human blood monocytes and its dietary-induced changes linking metabolic responses to nutrition with immune functions in health and metabolic diseases.


Assuntos
Carboidratos da Dieta/administração & dosagem , Gorduras na Dieta/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Obesidade/genética , Adulto , Estudos de Casos e Controles , Colecistocinina/genética , Colecistocinina/metabolismo , Dieta com Restrição de Gorduras , Dieta Hiperlipídica , Feminino , Humanos , Incretinas/genética , Incretinas/metabolismo , Macrófagos/metabolismo , Masculino , Monócitos/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Obesidade/sangue , Especificidade de Órgãos , Cultura Primária de Células , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neurotensina/genética , Receptores de Neurotensina/metabolismo
15.
Diabetes ; 64(3): 856-66, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25281430

RESUMO

WISP1 (Wnt1-inducible signaling pathway protein-1, also known as CCN4) is a member of the secreted extracellular matrix-associated proteins of the CCN family and a target gene of the Wingless-type (WNT) signaling pathway. Growing evidence links the WNT signaling pathway to the regulation of adipogenesis and low-grade inflammation in obesity. We aimed to validate WISP1 as a novel adipokine. Human adipocyte differentiation was associated with increased WISP1 expression and secretion. Stimulation of human macrophages with WISP1 led to a proinflammatory response. Circulating WISP1 and WISP1 subcutaneous adipose tissue expression were regulated by weight changes in humans and mice. WISP1 expression in visceral and subcutaneous fat tissue was associated with markers of insulin resistance and inflammation in glucose-tolerant subjects. In patients with nonalcoholic fatty liver disease, we found no correlation among disease activity score, liver fat content, and WISP1 expression. Insulin regulated WISP1 expression in adipocytes in vitro but had no acute effect on WISP1 gene expression in subcutaneous fat tissue in overweight subjects who had undergone hyperinsulinemic clamp experiments. The data suggest that WISP1 may play a role in linking obesity to inflammation and insulin resistance and could be a novel therapeutic target for obesity.


Assuntos
Adipocinas/metabolismo , Proteínas de Sinalização Intercelular CCN/metabolismo , Inflamação/metabolismo , Obesidade/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Tecido Adiposo/metabolismo , Animais , Western Blotting , Proteínas de Sinalização Intercelular CCN/genética , Células Cultivadas , Humanos , Gordura Intra-Abdominal/metabolismo , Macrófagos/metabolismo , Imageamento por Ressonância Magnética , Masculino , Células-Tronco Mesenquimais , Camundongos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas Proto-Oncogênicas/genética , Reação em Cadeia da Polimerase em Tempo Real , Gordura Subcutânea/metabolismo
16.
Regul Pept ; 179(1-3): 29-32, 2012 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-22960196

RESUMO

BACKGROUND: Increased plasma levels of calcitonin gene-related peptide-I (CGRP-I) and procalcitonin (Pro-CT) (both also named calcitonin peptides (CT peptides)) are associated with obesity and systemic inflammation. Glucose-dependent insulinotropic polypeptide (GIP), a nutrient-dependent incretin hormone, was recently found to induce CGRP-I and CT expression in human adipocytes in vitro. However, a physiological relevance of a possible interaction between GIP and CT peptides has not yet been studied. METHODS: In this study, we analyzed the effect of GIP on the expression of CGRP-I and CT mRNA in human subcutaneous adipose tissue within a randomized, controlled trial. Seventeen male obese subjects were infused with GIP [2.0 pmol kg(-1) min(-1) for 240 min] or placebo, either in the fasting state, during euglycemic-hyperinsulinemic (EC) or hyperglycemic-hyperinsulinemic clamps (HC). RESULTS: The CGRP-I gene expression was detected in all investigated adipose tissue samples, whereas very low CT expression was found in only 8 out of 116 analyzed samples. No significant influence of either GIP or glucose and insulin infusions on the CGRP-I and CT expression was observed in any of the individual experiments (GIP infusion, EC and HC) or in the combined analysis of all experiments with and without GIP. Furthermore, CGRP-I expression was not correlated with plasma GIP level before or after 240 min of infusions or clamps. CONCLUSION: In contrast to in vitro data, an acute application of GIP has no effect on mRNA expression of CT peptides in subcutaneous adipose tissue of obese humans.


Assuntos
Calcitonina/sangue , Polipeptídeo Inibidor Gástrico/farmacologia , Regulação da Expressão Gênica , Obesidade/metabolismo , Precursores de Proteínas/sangue , Gordura Subcutânea/metabolismo , Adulto , Glicemia , Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina , Polipeptídeo Inibidor Gástrico/sangue , Técnica Clamp de Glucose/métodos , Humanos , Hiperinsulinismo/metabolismo , Insulina/sangue , Masculino , Pessoa de Meia-Idade , Obesidade/genética , Precursores de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Gordura Subcutânea/efeitos dos fármacos , Relação Cintura-Quadril
17.
Diabetes ; 61(2): 292-300, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22179810

RESUMO

Glucose-dependent insulinotropic polypeptide (GIP) has been suggested to have direct effects on nonislet tissues. GIP also reportedly increased glucose uptake and inhibition of lipolysis in adipocytes after inhibition of the intracellular cortisone-cortisol shuttle 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1). We here analyzed whether GIP modifies lipid metabolism and further elucidated the relation between GIP, 11ß-HSD1, and fatty acid metabolism. GIP reduced activity of 11ß-HSD1 promoter constructs and the expression and activity of 11ß-HSD1 in differentiated 3T3-L1 adipocytes in a time- and dose-dependent fashion. This was paralleled by a reduction of free fatty acid (FFA) release and a reduced expression of key enzymes regulating lipolysis in adipose tissue. Preinhibition of 11ß-HSD1 completely abolished GIP-induced effects on FFA release. To investigate the acute effects of GIP in humans, a randomized clinical trial was performed. GIP lowered circulating FFAs compared with saline control and reduced expression and ex vivo activity of 11ß-HSD1 and adipose triglyceride lipase expression in subcutaneous fat biopsies. Our data suggest that GIP reduces FFA release from adipose tissue by inhibition of lipolysis or by increased reesterification. This process appears to depend on a modification of 11ß-HSD1 activity. In general, the presented data support that GIP has direct and insulin-independent effects on adipose tissue.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/fisiologia , Ácidos Graxos não Esterificados/metabolismo , Polipeptídeo Inibidor Gástrico/farmacologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Células 3T3-L1 , Adulto , Animais , Diferenciação Celular/efeitos dos fármacos , Ácidos Graxos não Esterificados/sangue , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Lipase/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Esterol Esterase/genética
18.
Dis Markers ; 31(4): 241-6, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22045431

RESUMO

Chromosomal locus 6q23 is strongly linked to type 2 diabetes (T2DM) and related features including insulin secretion in various ethnic populations. Connective tissue growth factor (CTGF) gene is an interesting T2DM candidate gene in this chromosome region. CTGF is a key mediator of progressive pancreatic fibrosis up-regulated in type 2 diabetes. In contrast, CTGF inactivation in mice compromises islet cell proliferation during embryogenesis. The aim of our study was to investigate an impact of CTGF genetic variation on pancreatic beta-cell function and T2DM pathogenesis. We studied the effect of a common CTGF polymorphism rs9493150 on the risk of the T2DM development in three independent German cohorts. Specifically, the association between CTGF polymorphism and non-invasive markers of beta-cell area derived from oral glucose tolerance test was studied in subjects without diabetes. Neither in the Metabolic Syndrome Berlin Potsdam (MESYBEPO) study (n=1026) (OR=0.637, CI (0.387-1.050); p=0.077) nor in the European Prospective Investigation into Cancer and Nutrition-Potsdam (EPIC-Potsdam) (n=3049) cohort (RR=0.77 CI (0.49-1.20), p=0.249 for the recessive homozygote in general model), a significant association with increased diabetes risk was observed. The risk allele of rs9493150 had also no effect on markers of beta-cell area in the combined analysis of the MESYBEPO and Tübingen Family Study (n=1826). In conclusion, the polymorphism rs9493150 in the 5'-untranslated region of the CTGF gene has no association with T2DM risk and surrogate markers of beta-cell area.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/genética , Diabetes Mellitus Tipo 2/genética , Células Secretoras de Insulina/patologia , Polimorfismo de Nucleotídeo Único , Adulto , Idoso , Glicemia , Peptídeo C/sangue , Estudos de Coortes , Feminino , Frequência do Gene , Estudos de Associação Genética , Marcadores Genéticos , Genótipo , Alemanha , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco , Análise de Sequência de DNA
19.
Am J Physiol Endocrinol Metab ; 301(4): E608-17, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21586700

RESUMO

The gastric peptide ghrelin promotes energy storage, appetite, and food intake. Nutrient intake strongly suppresses circulating ghrelin via molecular mechanisms possibly involving insulin and gastrointestinal hormones. On the basis of the growing evidence that glucose-dependent insulinotropic polypeptide (GIP) is involved in the control of fuel metabolism, we hypothesized that GIP and/or insulin, directly or via changes in plasma metabolites, might affect circulating ghrelin. Fourteen obese subjects were infused with GIP (2.0 pmol·kg(-1)·min(-1)) or placebo in the fasting state during either euglycemic hyperinsulinemic (EC) or hyperglycemic hyperinsulinemic clamps (HC). Apart from analysis of plasma ghrelin and insulin levels, GC-TOF/MS analysis was applied to create a hormone-metabolite network for each experiment. The GIP and insulin effects on circulating ghrelin were analyzed within the framework of those networks. In the HC, ghrelin levels decreased in the absence (19.2% vs. baseline, P = 0.028) as well as in the presence of GIP (33.8%, P = 0.018). Ghrelin levels were significantly lower during HC with GIP than with placebo, despite insulin levels not differing significantly. In the GIP network combining data on GIP-infusion, EC+GIP and HC+GIP experiments, ghrelin was integrated into hormone-metabolite networks through a connection to a group of long-chain fatty acids. In contrast, ghrelin was excluded from the network of experiments without GIP. GIP decreased circulating ghrelin and might have affected the ghrelin system via modification of long-chain fatty acid pools. These observations were independent of insulin and offer potential mechanistic underpinnings for the involvement of GIP in systemic control of energy metabolism.


Assuntos
Glicemia/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Grelina/sangue , Insulina/sangue , Sobrepeso/metabolismo , Adulto , Polipeptídeo Inibidor Gástrico/farmacologia , Glucagon/sangue , Teste de Tolerância a Glucose , Humanos , Masculino , Metabolômica , Pessoa de Meia-Idade
20.
Results Probl Cell Differ ; 52: 183-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20865381

RESUMO

Glucose-induced insulinomimetic peptide (GIP) is a gut hormone produced by enteroendocrine K-cells in the intestinal mucosa in response to fat, glucose, and also protein. GIP releases insulin from the ß cells of the pancreatic islets of Langerhans and therefore is an incretin hormone. GIP acts on a G-protein-coupled receptor that is widely distributed in the body including adipose tissue, stomach, brain, and others. Deletion of the GIP receptor (GIPR) renders mice resistant to weight gain induced by a high fat diet.We observed that weight gain induced by ovarectomy in female mice is prevented by GIPR deletion that is linked to reduced food intake and reduced hypothalamic expression of orectic neurotransmitters. Moreover, old male GIPR(-/-) mice placed on a high glycemic index diet maintained a high insulin sensitivity and were much more active than controls, which was not seen in young animals. Thus, GIP elicits central effects in response to nutrients that protect against obesity and insulin resistance. We then investigated the acute responses of humans to treatment with GIP over 4h in a dose mimicking postprandial plasma levels of about 100pmol/L. At basal glucose, GIP does not elicit insulin release. Fat biopsies taken before and after 4h of GIP treatment were analyzed for transcriptomic responses using Agilent whole human genome assays. There was a highly significant upregulation of an inflammatory expression pattern in a pathway analysis.


Assuntos
Metabolismo Energético/fisiologia , Polipeptídeo Inibidor Gástrico/fisiologia , Animais , Ingestão de Alimentos/fisiologia , Metabolismo Energético/genética , Feminino , Polipeptídeo Inibidor Gástrico/genética , Polipeptídeo Inibidor Gástrico/metabolismo , Hormônios Gastrointestinais/metabolismo , Hormônios Gastrointestinais/fisiologia , Homeostase/genética , Homeostase/fisiologia , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Masculino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA