Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Obes (Lond) ; 39(6): 977-85, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25771927

ABSTRACT

BACKGROUND/OBJECTIVES: Lymphocytes have a critical role in visceral adipose tissue (AT) inflammation. The CD28 costimulatory molecule is required for lymphocyte activation and for the development of a functional regulatory T cells (Tregs) compartment; however, its role during obesity is unknown. METHODS: During diet-induced obesity, we investigated the effects of selective interference with CD28 signaling using knockout mice (Cd28KO) and a CTLA4-Ig fusion protein inhibiting CD28-B7 interactions. RESULTS: Cd28 deficiency decreased pathogenic T cells and Treg content within AT without changing the macrophages number. Cd28KO epididymal but not subcutaneous fat was characterized by enlarged adipocytes, reduced levels of inflammatory cytokines and increased Glut4, adiponectin and lipogenic enzyme mRNA levels. This was associated with reduced inflammation, fat accumulation and enhanced glucose metabolism in liver. Weight gain and fasting glucose tolerance were not affected. CTLA4-Ig injections reduced the number of T cells in epididymal AT (epiAT) but not the inflammatory cytokines levels and failed to improve liver fat accumulation. CONCLUSIONS: Deletion of CD28 creates a new pro/anti-inflammatory balance in epiAT and liver and exerts a protective effect against hepatic steatosis.


Subject(s)
Adipose Tissue/pathology , CD28 Antigens/genetics , Fatty Liver/pathology , Gene Deletion , Inflammation/pathology , Liver/pathology , Obesity/pathology , Animals , Disease Models, Animal , Inflammation/metabolism , Insulin Resistance , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor Receptor Superfamily, Member 7
2.
Int J Obes (Lond) ; 38(5): 737-45, 2014 May.
Article in English | MEDLINE | ID: mdl-23999197

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the expression of human adipose tissue protein 53 (p53) in subjects who varied widely in terms of obesity and insulin resistance. We also analyzed different in vivo and in vitro models to try to comprehend the associations found in humans. METHODS: p53 was analyzed in human adipose and isolated adipocytes, in high fat-fed and GLP-1R KO mice, during in vitro adipogenesis, and in adipocytes after high glucose, rosiglitazone and inflammatory conditions. The effects of surgery-induced weight loss and ex vivo metformin were also evaluated. RESULTS: Omental (OM) p53 gene expression (+27%, P=0.001) and protein (+11%, P=0.04) were increased in obese subjects and high fat diet-induced obese mice (+86%, P=0.018). Although the obesity-associated inflammatory milieu was associated with increased OM p53, this was negatively related to insulin resistance and glycated hemoglobin, and positively with biomarkers for insulin sensitivity. Multiple linear regression analyses revealed that glycated hemoglobin (P<0.0001) and body mass index (P=0.048) contributed independently to explain 13.7% (P<0.0001) of the OM p53 variance. Accordingly, the improvement of insulin sensitivity with surgery-induced weight loss (+51%, P=0.01) and metformin (+42%, P=0.02) led to increased adipose p53. While the glucose-intolerant GLP-1R KO mice showed decreased mesenteric p53 (-45.4%, P=0.017), high glucose led to decreased p53 in pre-adipocytes (-27%, P<0.0001). Inflammatory treatments led to increased p53 (+35%, P<0.0001), while Rs downregulated this expression (-40%, P=0.005) in mature adipocytes. CONCLUSION: Inflammation and insulin resistance exert dual effects on adipose p53, which seems to be the final result of these opposing forces.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Genes, p53 , Inflammation/metabolism , Insulin Resistance , Obesity/metabolism , Omentum/metabolism , Adipogenesis , Analysis of Variance , Animals , Bariatric Surgery , Diet, High-Fat , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Humans , Inflammation/genetics , Male , Metformin/pharmacology , Mice , Mice, Knockout , Obesity/genetics , Omentum/surgery , Rosiglitazone , Thiazolidinediones/pharmacology
3.
Ann Pharm Fr ; 71(1): 34-41, 2013 Jan.
Article in French | MEDLINE | ID: mdl-23348854

ABSTRACT

Personalized medicine is becoming day-after-day more urgent taking into account the great diversity characterizing patients affected by a given pathology, especially metabolic diseases. In fact, antidiabetic/obesity treatments have shown a reduced or no effect at all in some patients, representing a major challenge physicians have to face worldwide. Therefore, efforts have to be put to identify individual factors affecting our susceptibility towards a given medication. In that regard, gut microbiota may stand for the missing piece of the metabolic puzzle regulating host response, since its role in the induction of metabolic diseases has now been achieved. In fact, we firstly provided a bacterial explanation for the low-grade chronic inflammation featuring metabolic diseases, by showing the lipopolysaccharide as a trigger and risk factor of such pathologies. However, despite similar lineages of microbes characterize the gut of people, important differences still remain, which may be responsible for opposite effect of treatments such as pre- or probiotics, whose efficacy seems to be governed by the own gut microbiota of subjects. We have recently shown that gut microbiota is associated to the inclination to resist or not high-fat diet-induced type 2 diabetes in mice. In addition, the direct targeting of gut microbes by dietary fibers reversed the observed metabolic phenotype. These results, together with the literature, strongly suggest gut microbiota as a new target for the development of personalized metabolic therapy.


Subject(s)
Gastrointestinal Tract/microbiology , Metabolic Diseases/microbiology , Metabolic Diseases/therapy , Animals , Humans , Inflammation/physiopathology , Metabolic Diseases/physiopathology , Mice , Precision Medicine
4.
Diabetologia ; 55(4): 1167-78, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22237685

ABSTRACT

AIMS/HYPOTHESIS: Endoplasmic reticulum (ER) stress has been implicated in the development of type 2 diabetes, via effects on obesity, insulin resistance and pancreatic beta cell health. C/EBP homologous protein (CHOP) is induced by ER stress and has a central role in apoptotic execution pathways triggered by ER stress. The aim of this study was to characterise the role of CHOP in obesity and insulin resistance. METHODS: Metabolic studies were performed in Chop ( -/- ) and wild-type C57Bl/6 mice, and included euglycaemic-hyperinsulinaemic clamps and indirect calorimetry. The inflammatory state of liver and adipose tissue was determined by quantitative RT-PCR, immunohistology and macrophage cultures. Viability and absence of ER stress in islets of Langerhans was determined by electron microscopy, islet culture and quantitative RT-PCR. RESULTS: Systemic deletion of Chop induced abdominal obesity and hepatic steatosis. Despite marked obesity, Chop ( -/- ) mice had preserved normal glucose tolerance and insulin sensitivity. This discrepancy was accompanied by lower levels of pro-inflammatory cytokines and less infiltration of immune cells into fat and liver. CONCLUSIONS/INTERPRETATION: These observations suggest that insulin resistance is not induced by fat accumulation per se, but rather by the inflammation induced by ectopic fat. CHOP may play a key role in the crosstalk between excessive fat deposition and induction of inflammation-mediated insulin resistance.


Subject(s)
Fatty Liver/metabolism , Inflammation/metabolism , Insulin Resistance/physiology , Obesity/metabolism , Transcription Factor CHOP/metabolism , Adipose Tissue/metabolism , Animals , Fatty Liver/genetics , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Inflammation/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Liver/metabolism , Mice , Mice, Knockout , Obesity/genetics , Transcription Factor CHOP/genetics
5.
Int J Obes (Lond) ; 36(11): 1442-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22184060

ABSTRACT

OBJECTIVE: Lipopolysaccharide-binding protein (LBP) is a 65-kDa acute-phase protein present in blood at high concentrations, known to be derived from the liver. We aimed to gain insights into the association of circulating LBP with insulin resistance in humans and mice. METHODS, DESIGN AND MEASUREMENTS: We studied the cross-sectional (n=222) and weight loss-induced (n=34) associations of LBP (enzyme-linked immunosorbent assay) with inflammatory and metabolic parameters (including minimal model-measured insulin sensitivity), and the effects of high-fat diet (HFD), metformin and genetic insulin sensitization (glucagon-like peptide 1 receptor knockout model) in mice. RESULTS: Circulating LBP concentration was significantly increased in subjects with type 2 diabetes and dramatically increased in subjects with morbid obesity. LBP was significantly associated with insulin sensitivity and different inflammatory markers and decreased after weight loss (22.2 ± 5.8 vs 16.2 ± 9.3 µg ml(-1), P<0.0001) in association with changes in body mass index and insulin sensitivity. Circulating LBP concentration was increased in HFD mice, whereas decreased in glucagon-like peptide 1 receptor knockout mice (significantly more insulin sensitive than wild-type mice) and after metformin administration. CONCLUSION: LBP is an inflammatory marker associated with obesity-related insulin resistance.


Subject(s)
Carrier Proteins/blood , Inflammation/blood , Insulin Resistance , Membrane Glycoproteins/blood , Obesity/blood , Acute-Phase Proteins/metabolism , Adipose Tissue , Animals , Biomarkers/blood , Body Mass Index , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Spain , Weight Loss
6.
Diabetes Obes Metab ; 14(7): 662-5, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22268579

ABSTRACT

Dipeptidyl peptidase-4 inhibitors (DPP-4i) improve glycaemic control in type 2 diabetes, but their benefits on reverse cholesterol transport (RCT) remain unknown. We evaluated the effects of DPP-4i sitagliptin 500 mg/kg/day on RCT in obese insulin-resistant CETP-apoB100 transgenic mice. Metformin 300 mg/kg/day orally was used as a reference compound. Both metformin and sitagliptin showed the expected effects on glucose parameters. Although no significant effect was observed on total cholesterol and high-density lipoprotein (HDL) cholesterol levels, sitagliptin, but not metformin, increased faecal cholesterol mass excretion by 132% (p < 0.001 vs. vehicle), suggesting a potent effect on cholesterol metabolism. Mice were then injected i.p. with (3) H-cholesterol labelled macrophages to measure RCT over 48 h. Compared with vehicle, sitagliptin significantly increased macrophage-derived (3) H-cholesterol faecal excretion by 39%. Administration of (14) C-cholesterol labelled olive oil orally showed a significant reduction of (14) C-tracer plasma appearance over time with sitagliptin, indicating that this drug promotes RCT through reduced intestinal cholesterol absorption.


Subject(s)
Apolipoprotein B-100/pharmacology , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacology , Macrophages/metabolism , Metformin/pharmacology , Pyrazines/pharmacology , Triazoles/pharmacology , Animals , Biological Transport/drug effects , Biomarkers/metabolism , Blood Glucose/metabolism , Cholesterol/metabolism , Cholesterol Ester Transfer Proteins/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Intestinal Absorption/drug effects , Lipoproteins, HDL/metabolism , Male , Mice , Mice, Obese , Mice, Transgenic , Sitagliptin Phosphate
7.
Diabetologia ; 54(1): 180-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20953578

ABSTRACT

AIMS/HYPOTHESIS: Extracellular signal-regulated kinase (ERK) activity is increased in adipose tissue in obesity and type 2 diabetes mellitus and strong evidences suggests that it is implicated in the downregulation of insulin signalling and action in the insulin-resistant state. To determine the role of ERK1 in obesity-associated insulin resistance in vivo, we inactivated Erk1 (also known as Mapk3) in obese leptin-deficient mice (ob/ob). METHODS: Mice of genotype ob/ob-Erk1⁻(/)⁻ were obtained by crossing Erk1⁻(/)⁻ mice with ob/ob mice. Glucose tolerance and insulin sensitivity were studied in 12-week-old mice. Tissue-specific insulin sensitivity, insulin signalling, liver steatosis and adipose tissue inflammation were determined. RESULTS: While ob/ob-Erk1⁻(/)⁻ and ob/ob mice exhibited comparable body weight and adiposity, ob/ob-Erk1⁻(/)⁻ mice did not develop hyperglycaemia and their glucose tolerance was improved. Hyperinsulinaemic-euglycaemic clamp studies demonstrated an increase in whole-body insulin sensitivity in the ob/ob-Erk1⁻(/)⁻ mice associated with an increase in both insulin-stimulated glucose disposal in skeletal muscles and adipose tissue insulin sensitivity. This occurred in parallel with improved insulin signalling in both tissues. The ob/ob-Erk1⁻(/)⁻ mice were also partially protected against hepatic steatosis with a strong reduction in acetyl-CoA carboxylase level. These metabolic improvements were associated with reduced expression of mRNA encoding inflammatory cytokine and T lymphocyte markers in the adipose tissue. CONCLUSIONS/INTERPRETATION: Our results demonstrate that the targeting of ERK1 could partially protect obese mice against insulin resistance and liver steatosis by decreasing adipose tissue inflammation and by increasing muscle glucose uptake. Our results indicate that deregulation of the ERK1 pathway could be an important component in obesity-associated metabolic disorders.


Subject(s)
Insulin Resistance/physiology , Leptin/deficiency , Mitogen-Activated Protein Kinase 3/deficiency , Obesity/physiopathology , Animals , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/physiopathology , Female , Insulin Resistance/genetics , Leptin/genetics , Male , Mice , Mice, Knockout , Mice, Obese , Mitogen-Activated Protein Kinase 3/genetics , Obesity/genetics
8.
Diabetologia ; 54(12): 3055-61, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21976140

ABSTRACT

AIMS/HYPOTHESIS: Evidence suggests that bacterial components in blood could play an early role in events leading to diabetes. To test this hypothesis, we studied the capacity of a broadly specific bacterial marker (16S rDNA) to predict the onset of diabetes and obesity in a general population. METHODS: Data from an Epidemiological Study on the Insulin Resistance Syndrome (D.E.S.I.R.) is a longitudinal study with the primary aim of describing the history of the metabolic syndrome. The 16S rDNA concentration was measured in blood at baseline and its relationship with incident diabetes and obesity over 9 years of follow-up was assessed. In addition, in a nested case-control study in which participants later developed diabetes, bacterial phylotypes present in blood were identified by pyrosequencing of the overall 16S rDNA gene content. RESULTS: We analysed 3,280 participants without diabetes or obesity at baseline. The 16S rDNA concentration was higher in those destined to have diabetes. No difference was observed regarding obesity. However, the 16S rDNA concentration was higher in those who had abdominal adiposity at the end of follow-up. The adjusted OR (95% CIs) for incident diabetes and for abdominal adiposity were 1.35 (1.11, 1.60), p = 0.002 and 1.18 (1.03, 1.34), p = 0.01, respectively. Moreover, pyrosequencing analyses showed that participants destined to have diabetes and the controls shared a core blood microbiota, mostly composed of the Proteobacteria phylum (85-90%). CONCLUSIONS/INTERPRETATION: 16S rDNA was shown to be an independent marker of the risk of diabetes. These findings are evidence for the concept that tissue bacteria are involved in the onset of diabetes in humans.


Subject(s)
Biomarkers/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/microbiology , Metabolic Syndrome/blood , Metagenome , RNA, Ribosomal, 16S/blood , Adult , Aged , Bacteria/classification , Bacteria/isolation & purification , Case-Control Studies , Diabetes Mellitus, Type 2/epidemiology , Female , France , Humans , Longitudinal Studies , Male , Middle Aged , Obesity/blood , Obesity, Abdominal/blood , Obesity, Abdominal/epidemiology
9.
Nat Med ; 3(10): 1096-101, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9334720

ABSTRACT

GLUT4, the insulin-responsive glucose transporter, plays an important role in postprandial glucose disposal. Altered GLUT4 activity is suggested to be one of the factors responsible for decreased glucose uptake in muscle and adipose tissue in obesity and diabetes. To assess the effect of GLUT4 expression on whole-body glucose homeostasis, we disrupted the murine GLUT4 gene by homologous recombination. Male mice heterozygous for the mutation (GLUT4 +/-) exhibited a decrease in GLUT4 expression in adipose tissue and skeletal muscle. This decrease in GLUT4 expression did not result in obesity but led to increased serum glucose and insulin, reduced muscle glucose uptake, hypertension, and diabetic histopathologies in the heart and liver similar to those of humans with non-insulin-dependent diabetes mellitus (NIDDM). The male GLUT4 +/- mice represent a good model for studying the development of NIDDM without the complications associated with obesity.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Insulin/metabolism , Monosaccharide Transport Proteins/deficiency , Monosaccharide Transport Proteins/genetics , Muscle Proteins , Muscle, Skeletal/physiopathology , Animals , Blood Glucose/metabolism , Blood Pressure/drug effects , Deoxyglucose/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Glucose Transporter Type 4 , Heterozygote , Insulin/blood , Insulin Resistance/physiology , Insulin Secretion , Isoproterenol/pharmacology , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Myocardium/pathology , Time Factors
10.
J Physiol Biochem ; 77(1): 155-166, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32648199

ABSTRACT

Saliva plays a key role in food absorption and digestion mainly due to both its enzymes and microbiota. The main objective of this study was to compare the oral microbiota and salivary parameters between men and women in response to feeding. To answer this question, we set up a pilot study on 10 male and 10 female subjects to examine the role of saliva in glycaemia physiology. Biological parameters and the microbiotal composition of saliva were analyzed in fasted and fed states. The results show that the level of blood glucose was not different between men and women in the fasted state (88.00 mg/dL ± 6.38 vs 87.00 mg/dL ±8.07, p = 0.9149) or in the fed state (102.44 mg/dL ± 14.03 vs 116.9 mg/dL ± 25, p = 0.1362). Free fatty acids (FFA 0.15 mmol/L ± 0.15 vs 0.07 mmol/L ± 0.07, p = 0,0078), cholesterol (0.53 mmol/L ± 0.30 vs 0.15 mmol/L ± 0.14, p < 0.0001), and total saliva proteins (13.2 g/L ± 4.31 vs 9.02 g/L ± 6.98, p = 0.0168) were decreased after feeding, as well as the saliva lipase (27.89 U/L ± 25.7 vs 12.28 U/L ± 4.85, p = 0.0126). A very significant increase in the relative abundance of Streptococcaceae (24.56 ± 9.32 vs 13.53 ± 7.47, p = 0.00055) and a decrease in Prevotellaceae (34.45 ± 9.30 vs 17.43 ± 9.03, p = 0.00055) were observed in the fed condition. When investigating gender-related differences in the fasted state, men showed higher levels of cholesterol (0.71 mmol/L ± 0.26 vs 0.40 mmol/L ± 0.27, p = 0.0329), FFA (0.25 mmol/L ± 0.18 vs 0.08 mmol/L ± 0.06, p = 0.0049), and triglycerides (0.24 mmol/L ± 0.15 vs 0.09 mmol/L ± 0.04, p = 0.006) than women. Finally, differences could be observed in saliva microbiota between men and women in the fasted condition but even more in the fed condition, where Porphyromonas and Capnocytophaga were overrepresented in the male salivary samples compared with female saliva. Thus, biological parameters and microbiota in saliva could be the signatures of the feeding conditions and sex gender status.


Subject(s)
Cholesterol/metabolism , Eating , Gastrointestinal Microbiome , Saliva/metabolism , Sex Factors , Triglycerides/metabolism , Adult , Female , Humans , Male , Pilot Projects , Young Adult
11.
Diabetologia ; 52(6): 1152-63, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19183933

ABSTRACT

AIMS/HYPOTHESIS: Obesity is associated with adipose tissue inflammation. The CD40 molecule, TNF receptor superfamily member 5 (CD40)/CD40 ligand (CD40L) pathway plays a role in the onset and maintenance of the inflammatory reaction, but has not been studied in human adipose tissue. Our aim was to examine CD40 expression by human adipocytes and its participation in adipose tissue inflammation. METHODS: CD40 expression was investigated in human whole adipose tissue and during adipocyte differentiation by real-time PCR, Western blot and immunohistochemistry. The CD40/CD40L pathway was studied using recombinant CD40L (rCD40L) in adipocyte culture and neutralising antibodies in lymphocyte/adipocyte co-culture. RESULTS: CD40 mRNA levels in subcutaneous adipose tissue were higher in the adipocyte than in the stromal-vascular fraction. CD40 expression was upregulated during adipocyte differentiation. Addition of rCD40L to adipocytes induced mitogen activated protein kinase (MAPK) activation, stimulated inflammatory adipocytokine production, and decreased insulin-induced glucose transport in parallel with a downregulation of IRS1 and GLUT4 (also known as SCL2A4). rCD40L decreased the expression of lipogenic genes and increased lipolysis. CD40 mRNA levels were significantly higher in subcutaneous adipose tissue than in visceral adipose tissue of obese patients and were positively correlated with BMI, and with IL6 and leptin mRNA levels. Lymphocyte/adipocyte co-culture led to an upregulation of proinflammatory adipocytokines and a downregulation of leptin and adiponectin. Physical separation of the two cell types attenuated these effects, suggesting the involvement of a cell-cell contact. Blocking the CD40/CD40L interaction with neutralising antibodies reduced IL-6 secretion from adipocytes. CONCLUSIONS/INTERPRETATION: Adipocyte CD40 may contribute to obesity-related inflammation and insulin resistance. T lymphocytes regulate adipocytokine production through both the release of soluble factor(s) and heterotypic contact with adipocytes involving CD40.


Subject(s)
Adipocytes/metabolism , CD40 Antigens/genetics , CD40 Antigens/metabolism , Lymphocytes/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adiponectin/metabolism , Adrenomedullin/metabolism , Animals , Blotting, Western , CD40 Ligand/pharmacology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Gene Expression/drug effects , Humans , Immunohistochemistry , Lymphocytes/drug effects , Mice , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
Diabetologia ; 52(5): 941-51, 2009 May.
Article in English | MEDLINE | ID: mdl-19277604

ABSTRACT

AIMS/HYPOTHESIS: Fatty acids of marine origin, i.e. docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) act as hypolipidaemics, but they do not improve glycaemic control in obese and diabetic patients. Thiazolidinediones like rosiglitazone are specific activators of peroxisome proliferator-activated receptor gamma, which improve whole-body insulin sensitivity. We hypothesised that a combined treatment with a DHA and EPA concentrate (DHA/EPA) and rosiglitazone would correct, by complementary additive mechanisms, impairments of lipid and glucose homeostasis in obesity. METHODS: Male C57BL/6 mice were fed a corn oil-based high-fat diet. The effects of DHA/EPA (replacing 15% dietary lipids), rosiglitazone (10 mg/kg diet) or a combination of both on body weight, adiposity, metabolic markers and adiponectin in plasma, as well as on liver and muscle gene expression and metabolism were analysed. Euglycaemic-hyperinsulinaemic clamps were used to characterise the changes in insulin sensitivity. The effects of the treatments were also analysed in dietary obese mice with impaired glucose tolerance (IGT). RESULTS: DHA/EPA and rosiglitazone exerted additive effects in prevention of obesity, adipocyte hypertrophy, low-grade adipose tissue inflammation, dyslipidaemia and insulin resistance, while inducing adiponectin, suppressing hepatic lipogenesis and decreasing muscle ceramide concentration. The improvement in glucose tolerance reflected a synergistic stimulatory effect of the combined treatment on muscle glycogen synthesis and its sensitivity to insulin. The combination treatment also reversed dietary obesity, dyslipidaemia and IGT. CONCLUSIONS/INTERPRETATION: DHA/EPA and rosiglitazone can be used as complementary therapies to counteract dyslipidaemia and insulin resistance. The combination treatment may reduce dose requirements and hence the incidence of adverse side effects of thiazolidinedione therapy.


Subject(s)
Dietary Fats/pharmacology , Fatty Acids, Omega-3/pharmacology , Glycogen/biosynthesis , Insulin/physiology , Muscle, Skeletal/metabolism , Thiazolidinediones/pharmacology , Animals , Corn Oil/pharmacology , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Glucose Intolerance/metabolism , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Rosiglitazone
13.
Diabetes Metab ; 34(6 Pt 1): 627-30, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19026584

ABSTRACT

The first antidiabetic agent was a hormone--insulin--and ever since, all therapeutic strategies have been based on the synthesis of chemical compounds to bind its receptors or transcription factors, or to trigger its intracellular mechanisms. Eighty years on, new therapeutic molecules are available for the treatment of diabetes and, again, are based on a hormone--glucagon-like peptide-1 (GLP-1). Whereas the theoretical benefit of insulin is based on normalization of functional physiology, therapeutic strategies based on GLP-1 aim to increase the circulating concentration of a natural component--the hormone GLP-1. There are two strategies for increasing GLP-1 plasma concentrations: replace the hormone with a long-acting analogue or molecule with a longer half-life; and prevent its degradation by inhibiting its natural protease, dipeptidyl peptidase IV (DPPIV). Although numerous clinical trials have been carried out and vast amounts of data are available, the mechanisms through which GLP-1-based therapy reduces blood glucose in diabetic patients remain unclear. Thus, it is essential to ask the right questions and to design appropriate clinical trials and experiments to increase our understanding of the mode of action of GLP-1-based therapy. For this reason, in the spring of 2008, expert scientists and clinicians in the field of GLP-1 got together for an intensive debate on the subject at the first meeting of the European Club for the study of GLP-1, held in Marseille. The subject of the round table discussions was: what is known, new and controversial about GLP-1? During these discussions, numerous facts and controversies were reevaluated, and revealed that several long-held, dogmatic beliefs have never been fully and scientifically established. These points are detailed here in these minutes of the landmark meeting.


Subject(s)
Glucagon-Like Peptide 1/physiology , Insulin/metabolism , Animals , Cell Differentiation , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/physiopathology , France , Glucagon/physiology , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/therapeutic use , Humans , Insulin Secretion , Insulin-Secreting Cells/cytology , L Cells/physiology , Mice
14.
Diabetes Metab ; 34 Suppl 2: S49-55, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18640586

ABSTRACT

A major, yet poorly understood, feature of type 2 diabetes is the excessive hepatic glucose production and the corresponding insulin resistance leading to fasting hyperglycaemia. The tremendous amount of work done to provide the physiological and molecular mechanisms explaining this impairment has led to the emergence of several consensual hypotheses. Among these, is the increased daily and unregulated plasma glucagon concentration in type 2 diabetic patients. Therefore, studies aiming to understand the physiological regulation of glucagon secretion and the corresponding impairment during diabetes are directly relevant to the treatment of type 2 diabetes. Glucagon secretion by alpha-cells is an immediate response to glucopenia. Abnormal secretion of glucagon and other counterregulatory hormones is a hallmark of type 1 and type 2 diabetes and a major limitation to the use of strong hypoglycaemia agents. A few molecular mechanisms of glucose detection triggering counterregulation and in particular inducing glucagon secretion or suppressing it during hyperglycaemic episodes, have been identified. Such mechanisms are related to those of the insulin secreted beta-cell. The glucose transporter GLUT2 and the K-ATP dependent channel, as well as regulatory mechanisms, involved the central nervous system and the gut-brain hormone GLP-1. Over the last years, glucoincretins have provided promising results for the normalization of plasma glucagon concentration of type 2 diabetic patients, which could partly explain the therapeutic benefits of incretin-related therapy. The underlined mechanisms of GLP-1 regulated glucagon secretion are most likely related to the action of the hormone on the activation of the portal and brain glucose sensors. Certainly, strategies aiming to restore glucose-regulated glucagon secretion are important milestones for the treatment of diabetic patient and the prevention of iatrogenic hypoglycaemia.


Subject(s)
Diabetes Mellitus/physiopathology , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Hyperglycemia/physiopathology , Humans , Hyperglycemia/blood
15.
J Clin Invest ; 98(3): 629-34, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8698853

ABSTRACT

The ability of muscles from Glut 4-null mice to take up and metabolize glucose has been studied in the isolated white EDL and red soleus muscles. In EDL muscles from male or female Glut 4-null mice, basal deoxyglucose uptake was lower than in control muscles and was not stimulated by insulin. In parallel, glycogen synthesis and content were decreased. Soleus muscles from male Glut 4-null mice took up twice more deoxyglucose in the absence of insulin than control muscles, but did not respond to insulin. In females, soleus deoxyglucose uptake measured in the absence of hormone was similar in Glut 4-null mice and in control mice. This uptake was stimulated twofold in Glut 4-null mice and threefold in control mice. Basal glycogen synthesis was increased by 4- and 2.2-fold in male and female null mice, respectively, compared to controls, and insulin had no or small (20% stimulation over basal) effect. These results indicate that while EDL muscles behaved as expected, soleus muscles were able to take up a large amount of glucose in the absence (males) or the presence of insulin (females). Whether this is due to a change in Glut 1 intrinsic activity or targeting and/or to the appearance of another glucose transporter remains to be determined.


Subject(s)
Glucose/metabolism , Glycogen/biosynthesis , Monosaccharide Transport Proteins/physiology , Muscle Proteins , Muscle, Skeletal/metabolism , Animals , Female , Glucose Transporter Type 4 , Male , Mice , Monosaccharide Transport Proteins/genetics , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Receptor, Insulin/metabolism
16.
J Clin Invest ; 114(3): 339-48, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15286800

ABSTRACT

Transient neonatal diabetes mellitus (TNDM) is a rare inherited diabetic syndrome apparent in the first weeks of life and again during early adulthood. The relative contributions of reduced islet beta cell number and impaired beta cell function to the observed hypoinsulinemia are unclear. The inheritance pattern of this imprinted disorder implicates overexpression of one or both genes within the TNDM locus: ZAC, which encodes a proapoptotic zinc finger protein, and HYMAI, which encodes an untranslated mRNA. To investigate the consequences for pancreatic function, we have developed a high-copy transgenic mouse line, TNDM29, carrying the human TNDM locus. TNDM29 neonates display hyperglycemia, and older adults, impaired glucose tolerance. Neonatal hyperglycemia occurs only on paternal transmission, analogous to paternal dependence of TNDM in humans. Embryonic pancreata of TNDM29 mice showed reductions in expression of endocrine differentiation factors and numbers of insulin-staining structures. By contrast, beta cell mass was normal or elevated at all postnatal stages, whereas pancreatic insulin content in neonates and peak serum insulin levels after glucose infusion in adults were reduced. Expression of human ZAC and HYMAI in these transgenic mice thus recapitulates key features of TNDM and implicates impaired development of the endocrine pancreas and beta cell function in disease pathogenesis.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2 , Homeostasis , Aging , Animals , Animals, Newborn , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Genomic Imprinting , Humans , Hyperglycemia/genetics , Immunohistochemistry , In Situ Hybridization , Insulin/blood , Insulin/metabolism , Islets of Langerhans/physiopathology , Mice , Mice, Transgenic , Pancreas/embryology , Pancreas/pathology , Pancreas/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transcription, Genetic
17.
Swiss Med Wkly ; 137(49-50): 700-4, 2007 Dec 22.
Article in English | MEDLINE | ID: mdl-18197485

ABSTRACT

BACKGROUND: nitric oxide (NO) plays an important role in the regulation of cardiovascular and glucose homeostasis. Mice lacking the gene encoding the neuronal isoform of nitric oxide synthase (nNOS) are insulin-resistant, but the underlying mechanism is unknown. nNOS is expressed in skeletal muscle tissue where it may regulate glucose uptake. Alternatively, nNOS driven NO synthesis may facilitate skeletal muscle perfusion and substrate delivery. Finally, nNOS dependent NO in the central nervous system may facilitate glucose disposal by decreasing sympathetic nerve activity. METHODS: in nNOS null and control mice, we studied whole body glucose uptake and skeletal muscle blood flow during hyperinsulinaemic clamp studies in vivo and glucose uptake in skeletal muscle preparations in vitro. We also examined the effects of alpha-adrenergic blockade (phentolamine) on glucose uptake during the clamp studies. RESULTS: as expected, the glucose infusion rate during clamping was roughly 15 percent lower in nNOS null than in control mice (89 (17) vs 101 (12) [-22 to -2]). Insulin stimulation of muscle blood flow in vivo, and intrinsic muscle glucose uptake in vitro, were comparable in the two groups. Phentolamine, which had no effect in the wild-type mice, normalised the insulin sensitivity in the mice lacking the nNOS gene. CONCLUSIONS: insulin resistance in nNOS null mice was not related to defective insulin stimulation of skeletal muscle perfusion and substrate delivery or insulin signaling in the skeletal muscle cell, but to a sympathetic alpha-adrenergic mechanism.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Insulin Resistance/physiology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Animals , Blood Glucose/drug effects , Female , Glucose Clamp Technique , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Phentolamine/metabolism , Phentolamine/pharmacology
18.
J Physiol Biochem ; 63(4): 305-15, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18457006

ABSTRACT

The combination of vanadate plus benzylamine has been reported to stimulate glucose transport in rodent adipocytes and to mimic other insulin actions in diverse studies. However, benzylamine alone activates glucose uptake in human fat cells and increases glucose tolerance in rabbits. The aim of this work was to unravel the benzylamine antihyperglycemic action and to test whether its chronic oral administration could restore the defective glucose handling of mice rendered slightly obese and diabetic by very high-fat diet (VHFD). When VHFD mice were i.p. injected with benzylamine at 0.7 to 700 micromol/kg before glucose tolerance test, they exhibited reduced hyperglycemic response without alteration of insulin secretion. Whole body glucose turnover, as assessed by the glucose isotopic dilution technique, was unchanged in mice perfused with benzylamine (total dose of 75 micromol/kg). However, their in vivo glycogen synthesis rate was increased. Benzylamine appeared therefore to directly facilitate glucose utilisation in peripheral tissues. When given chronically at 2000 or 4000 micromol/kg/d in drinking water, benzylamine elicited a slight reduction of water consumption but did not change body weight or adiposity and did not modify oxidative stress markers. Benzylamine treatment improved glucose tolerance but failed to normalize the elevated glucose fasting plasma levels of VHFD mice. There was no influence of benzylamine ingestion on lipolytic activity, basal and insulin-stimulated glucose uptake, and on inflammatory adipokine expression in adipocytes. The improvement of glucose tolerance and the lack of adverse effects on adipocyte metabolism, reported here in VHFD mice allow to consider orally given benzylamine as a potential antidiabetic strategy which deserves to be further studied in other diabetic models.


Subject(s)
Benzylamines/administration & dosage , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Hypoglycemic Agents/administration & dosage , Adipocytes/metabolism , Animals , Benzylamines/pharmacology , Diabetes Mellitus, Experimental/complications , Dietary Fats/administration & dosage , Glucose Tolerance Test , Hyperlipidemias/metabolism , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/metabolism , Oxidative Stress
19.
Obes Rev ; 18(1): 86-98, 2017 01.
Article in English | MEDLINE | ID: mdl-27636208

ABSTRACT

Over the past 30 years, there has been a dramatic rise in global obesity prevalence, resulting in significant economic and social consequences. Attempts to develop pharmacological agents to treat obesity have met with many obstacles including the lack of long-term effectiveness and the potential for adverse effects. Historically, there have been limited treatment options for overweight and obesity; however, since 2012, a number of new drugs have become available. A number of peptides produced in the gut act as key mediators of the gut-brain axis, which is involved in appetite regulation. This review discusses the role of the gut-brain axis in appetite regulation with special focus on glucagon-like peptide-1. Liraglutide 3.0 mg, a glucagon-like peptide-1 receptor agonist that targets this pathway, is now approved for the treatment of obesity and overweight (body mass index ≥27 kg/m2 ) with comorbidities such as type 2 diabetes, high blood pressure, high cholesterol or obstructive sleep apnoea. In addition, other glucagon-like peptide-1 receptor agonists offer promise for obesity management in the future. This review examines how glucagon-like peptide-1 receptor agonists promote weight loss and summarizes the clinical data on weight loss with glucagon-like peptide-1 receptor agonists.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Liraglutide/therapeutic use , Obesity/drug therapy , Overweight/drug therapy , Appetite Regulation/drug effects , Comorbidity , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Randomized Controlled Trials as Topic , Weight Loss
20.
Pediatr Obes ; 12(4): e28-e32, 2017 08.
Article in English | MEDLINE | ID: mdl-27135640

ABSTRACT

Gut microbes are active participants of host metabolism. At birth, child physiology is committed towards healthiness or sickness depending, in part, on maternal condition (i.e. lean vs obesity) and delivery. Finally, changes from breastfeeding to solid food also account to define gut microbiota ecology in adulthood. Nowadays, alterations of gut microbiota, named dysbiosis, are acquired risk factors for multiple diseases, especially type 2 diabetes and obesity. Despite important evidence linking nutrition to dysbiosis to energetic dysmetabolism, molecular mechanisms for causality are still missing. That the status of gut microbiota of mother and child is crucial for future diseases is witnessed by adulthood overweight and obesity observed in children with dysbiosis. In this short review we highlight the importance of early life events related to the microbiota and their impact on future adult disease risk. Therefore, our effort to treat or prevent metabolic diseases should be addressed towards early or previous life steps, when microbial decisions are going to affect our metabolic fate.


Subject(s)
Dysbiosis/microbiology , Gastrointestinal Microbiome , Pediatric Obesity/microbiology , Adult , Child , Humans , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL