Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
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
2.
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
3.
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
4.
Benef Microbes ; 7(1): 11-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26565087

ABSTRACT

Changes in the gut microbiota are associated with metabolic disorders, such as overweight and elevated blood glucose. Mouse studies have shown that gut microbiota can regulate metabolism with a mechanism related to gut barrier function. An impaired gut barrier permits the translocation of bacteria and their components which, when in contact with the sub-mucosal immune system, evoke metabolic inflammation and distract signalling in metabolically active tissues. Despite thorough research of the topic in animals, the hypothesis is yet to be proven in humans. Cross-sectional studies have shown that certain bacterial populations - such as Akkermansia muciniphila, Faecalibacterium prausnitzii, Methanobrevibacter smithii and Christensenellaceae - are better represented in lean individuals compared to those who are overweight or metabolically unhealthy. Although these differences reflect those seen in mice, it is possible that they are caused by different dietary or other lifestyle habits. Diet has an indisputable influence on gut microbiota making it very difficult to draw conclusions on microbiota-host interactions from cross-sectional studies. Certain research areas do, however, indicate that gut microbiota could causally influence metabolism. Several studies show that antibiotic use in infancy increases body weight in later childhood. Also, probiotics are emerging as a potential therapy for metabolic syndrome. In fact, a handful of human studies and numerous animal studies show promise for probiotics in reducing blood glucose levels or improving insulin sensitivity. For weight management human evidence is scarcer. Nevertheless, it is becoming increasingly recognised that gut microbiota plays a part regulating metabolism, also in humans, which gives rise to novel opportunities for preventative and treatment strategies.


Subject(s)
Gastrointestinal Microbiome , Glucose/metabolism , Probiotics/therapeutic use , Weight Gain , Animals , Humans , Metabolic Diseases/microbiology
5.
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
6.
Benef Microbes ; 5(4): 437-45, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25062610

ABSTRACT

Alterations of the gut microbiota and mucosal barrier are linked with metabolic diseases. Our aim was to investigate the potential benefit of the potential probiotic Bifidobacterium animalis ssp. lactis 420 in reducing high-fat diet-induced body weight gain and diabetes in mice. In the obesity model, C57Bl/6J mice were fed a high-fat diet (60 energy %) for 12 weeks, and gavaged daily with B. lactis 420 (109 cfu) or vehicle. In the diabetes model, mice were fed a high-fat, ketogenic diet (72 energy % fat) for 4 weeks, with a 6-week subsequent treatment with B. lactis 420 (108-1010 cfu/day) or vehicle, after which they were analysed for body composition. We also analysed glucose tolerance, plasma lipopolysaccharide and target tissue inflammation using only one of the B. lactis 420 groups (109 cfu/day). Intestinal bacterial translocation and adhesion were analysed in a separate experiment using an Escherichia coli gavage. Body fat mass was increased in both obese (10.7 ± 0.8 g (mean ± standard error of mean) vs. 1.86 ± 0.21 g, P<0.001) and diabetic mice (3.01 ± 0.4 g vs. 1.14 ± 0.15 g, P<0.001) compared to healthy controls. Treatment with B. lactis 420 significantly decreased fat mass in obese (7.83 ± 0.67 g, P=0.007 compared to obese with vehicle) and diabetic mice (1.89 ± 0.16 g, P=0.02 for highest dose). This was reflected as reduced weight gain and improved glucose tolerance. Furthermore, B. lactis 420 decreased plasma lipopolysaccharide levels (P<0.001), liver inflammation (P=0.04), and E. coli adhesion in the distal gut (P<0.05). In conclusion, B. lactis 420 reduces fat mass and glucose intolerance in both obese and diabetic mice. Reduced intestinal mucosal adherence and plasma lipopolysaccharide suggest a mechanism related to reduced translocation of gut microbes.


Subject(s)
Bifidobacterium/physiology , Glucose Intolerance/prevention & control , Obesity/prevention & control , Probiotics/administration & dosage , Animals , Body Composition , Male , Mice, Inbred C57BL , Mice, Obese , Treatment Outcome
7.
Diabetes Metab ; 40(3): 186-90, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24462190

ABSTRACT

One major discovery of the last decade in the field of metabolic diseases is that the microorganisms comprising the gut microbiota are now considered a metabolic "organ", modulating multiple functions of the host, such as intestinal immune system maturation, adiposity, cardiac metabolism, liver triglyceride storage, and brain development and behaviour. The corresponding mechanisms involve increased energy harvesting through the production by microbiota of short-chain fatty acids for use by the host, and the release of pro-inflammatory compounds, such as lipopolysaccharide (LPS), flagellin and peptidoglycan. In particular, a high-fat diet (HFD) modifies gut microbiota, resulting in an increase of plasma LPS levels known as "metabolic endotoxaemia", a major driver of the onset of metabolic diseases through a CD14-dependent mechanism. The LPS-sensitive cell types can be seen within bone marrow-derived cells (BMC), which are involved in the development of inflammation in the adipose tissue of obese and type 2 diabetic mice. Furthermore, the expression of LPS receptor/cofactor CD14 cells from the stromal vascular fraction of adipose depots can also be directly targeted by LPS to initiate precursor cell development and adiposity. Moreover, data from the literature also indicate an impact of gut microbiota on intestinal stem cells. Thus, this mini review presents the experimental evidence supporting a relationship between gut microbiota and stem cells as a new axis of metabolic homoeostasis control.


Subject(s)
Adipose Tissue/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Gastrointestinal Tract/microbiology , Inflammation/metabolism , Insulin Resistance , Lipopolysaccharides/metabolism , Microbiota , Stem Cells/metabolism , Adipose Tissue/metabolism , Adipose Tissue/microbiology , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/microbiology , Diet, High-Fat , Endotoxemia/blood , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Inflammation/immunology , Inflammation/microbiology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/blood , Mice , Mice, Inbred C57BL , Mice, Obese
8.
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
9.
Diabetes Metab ; 39(2): 89-93, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23523140

ABSTRACT

Glucagon-like peptide (GLP)-1 action involves both endocrine and neural pathways to control peripheral tissues. In diabetes the impairment of either pathway may define different subsets of patients: some may be better treated with GLP-1 receptor agonists that are more likely to directly stimulate beta-cells and extrapancreatic receptors, while others may benefit from dipeptidyl peptidase (DPP)-4 inhibitor treatments that are more likely to increase the neural gut-brain-pancreas axis. Elevated plasma concentrations of GLP-1 associated with agonist treatment or bariatric surgery also appear to exert neuroprotective effects, ameliorate postprandial and fasting lipids, improve heart physiology and protect against heart failure, thereby expanding the possible positioning of GLP-1-based therapies. However, the mechanisms behind GLP-1 secretion, the role played by proximal and distal intestinal GLP-1-producing cells as well as the molecular basis of GLP-1 resistance in diabetes are still to be ascertained. The pharmacological features distinguishing GLP-1 receptor agonists from DPP-4 inhibitors are discussed here to address their respective positions in type 2 diabetes.


Subject(s)
Cardiovascular Diseases/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Glucagon-Like Peptide 1/metabolism , Incretins/metabolism , Bariatric Surgery/trends , Cardiovascular Diseases/blood , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/blood , Diabetic Angiopathies/prevention & control , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Fasting , Female , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Humans , Incretins/pharmacology , Incretins/therapeutic use , Lipids/blood , Male
10.
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
11.
Ann Cardiol Angeiol (Paris) ; 61(3): 173-7, 2012 Jun.
Article in French | MEDLINE | ID: mdl-22621847

ABSTRACT

Diabetes-driven cardiovascular diseases represent a high challenge for developed countries. Periodontal disease is strictly linked to the aforementioned diseases, due to its Gram negative-driven inflammation. Thus, we investigated the effects of periodontal disease on arterial pressure during the development of diabetes in mice. To this aim, C57BL/6 female mice were colonized with pathogens of periodontal tissue (Porphyromonas gingivalis, Prevotella intermedia and Fusobacterium nucleatum) for 1month, whereas another group of mice did not undergo the colonization. Subsequently, all mice were fed a high-fat carbohydrate-free diet for 3months. Then, arterial pressure was measured in vivo and a tomodensitometric analysis of mandibles was realized as well. Our results show increased mandibular bone-loss induced by colonization with periopathogens. In addition, periodontal infection augmented glucose-intolerance and systolic and diastolic arterial pressure, parameters already known to be affected by a fat-diet. In conclusion, we show here that periodontal disease amplifies metabolic troubles and deregulates arterial pressure, emerging as a new axis of metabolic investigation.


Subject(s)
Arterial Pressure , Diabetes Complications/microbiology , Periodontal Diseases/microbiology , Alveolar Bone Loss/microbiology , Animals , Cardiovascular Diseases/immunology , Cardiovascular Diseases/microbiology , Diabetes Complications/immunology , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Fusobacterium nucleatum/growth & development , Insulin Resistance/immunology , Mandibular Diseases/microbiology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Periodontal Index , Porphyromonas gingivalis/growth & development , Prevotella intermedia/growth & development
12.
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
13.
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
14.
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
15.
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
16.
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
18.
Diabetes Metab ; 36(6 Pt 1): 503-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21075654

ABSTRACT

Over the past 2 years, more than 1300 manuscripts have been published on glucagon-like peptide-1 (GLP-1) and yet, what do we know about it for sure? The European Club for the Study of GLP-1 (EuCSGLP-1) has debated the latest controversies concerning GLP-1, including both fundamental and clinical aspects, and concluded that the control of glucose metabolism by GLP-1 requires paracrine activation of the enteric nervous system to regulate numerous physiological functions. This involves-but is not limited to-the endocrine pancreas, liver, cardiovascular system, gastric-emptying and the brain. For this reason, the role of GLP-1 as an endocrine hormone has come under question. As systemic concentration of the peptide was not thought to be relevant to its physiological action, it was proposed that dipeptidyl peptidase-4 (DPP4) inhibitors would involve the control of enteric, rather than circulating, DPP4 activity to effectively regulate glycaemia. In any case, the concomitant insulinotropic and glucagonostatic roles of GLP-1 were believed to be of equal importance to glucose control. Another important question was related to the role of GLP-1 on beta cell apoptosis, regeneration and differentiation in type 2 diabetic patients. Although evidence in vitro showed that GLP-1 controls these functions, such effects remain elusive in humans in vivo. Nevertheless, the consensus was that GLP-1 could control glucose responsiveness, one of the first impaired physiological functions at the onset of diabetes. The therapeutic efficiency of GLP-1 would be related to the initial restoration of glucose competence, while an increase of beta cell mass has not yet been demonstrated. From a clinical and fundamental point of view, it was concluded that, at the onset of diabetes, an initial triggering of GLP-1 secretion-by metformin coupled with a DPP4 inhibitor-would help to activate the gut-peripheral axis and, hence, restore adequate regulation of glycaemia. GLP-1 analogues would certainly be helpful in association with long-acting insulin (albeit off-label) in patients with impaired beta cells and GLP-1 secretory potential. However, a reliable and routine feasible test to systematically assess dynamic insulin secretion is essential. More important, factors that influence therapeutic response, such as compliance and lifestyle, as well as pharmacokinetics and dosing, disease duration, age, gender, ethnicity, patients' clinical characteristics, autonomic nervous system integrity and genotype characteristics also need to be considered. A few innovative perspectives have been debated, such as the recently discovered cardiovascular protective effects of the native GLP-1 peptide and its degradation product GLP-1(9-36), as well as the neuroprotection offered by GLP-1. Although still considered speculative, these perspectives remain hopeful and promising.


Subject(s)
Endocrine System/drug effects , Endocrine System/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/physiology , Animals , Glucagon/metabolism , Glucagon-Like Peptide 1/analogs & derivatives , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Molecular Targeted Therapy , Peptides/pharmacology
19.
Diabetes Metab ; 36 Suppl 3: S54-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21211737

ABSTRACT

Glucose homeostasis corresponds to the overall physiological, cellular, and molecular mechanisms which tightly maintain the glycaemia between ∼4.5 and ∼6 mM. The resulting blood glucose concentration is the consequence of a balance between the mechanisms that ensure the entry and the output of glucose in the blood. A dynamic balance needs hence to be perfectly achieved in order to maintain a physiological glycaemic concentration. Specialized cells from the intestine continuously detect changes in glucose concentration and send signals to peripheral tissues and the brain through the vagus nerve. The molecular mechanisms involved in glucose detection have not been perfectly defined but could resemble those from the insulin-secreting beta cells. The brain then integrates the enteric and circulating endocrine signals to generate a new signal towards peripheral tissues such as the pancreas, liver, muscles, and blood vessels. This metabolic reflex is called anticipatory since it allows the peripheral tissues to prepare for the adequate handling of nutrients. Diabetes is associated with an impaired anticipatory reflex, which hampers the proper detection of nutrients and leads to hyperglycaemic episodes. Recently, GLP-1-based therapies have demonstrated the improvement of glucose detection and their efficacy on glycaemic control. Although not yet fully demonstrated, GLP-1-based therapies regulate glucose sensors, which leads to the glycaemic improvement. Certainly other molecular targets could be identified to further generate new therapeutic strategies.


Subject(s)
Brain/physiology , Glucose/metabolism , Intestines/physiology , Animals , Autonomic Nervous System/metabolism , Diabetes Mellitus/metabolism , Humans , Incretins/metabolism , Signal Transduction
20.
Diabetes Metab ; 35(4): 262-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19419895

ABSTRACT

Recent advances in molecular sequencing technology have allowed researchers to answer major questions regarding the relationship between a vast genomic diversity-such as found in the intestinal microflora-and host physiology. Over the past few years, it has been established that, in obesity, type 1 diabetes and Crohn's disease-to cite but a few-the intestinal microflora play a pathophysiological role and can induce, transfer or prevent the outcome of such conditions. A few of the molecular vectors responsible for this regulatory role have been determined. Some are related to control of the immune, vascular, endocrine and nervous systems located in the intestines. However, more important is the fact that the intestinal microflora-to-host relationship is bidirectional, with evidence of an impact of the host genome on the intestinal microbiome. This means that the ecology shared by the host and gut microflora should now be considered a new player that can be manipulated, using pharmacological and nutritional approaches, to control physiological functions and pathological outcomes. What now remains is to demonstrate the molecular connection between the intestinal microflora and metabolic diseases. We propose here that the proinflammatory lipopolysaccharides play a causal role in the onset of metabolic disorders.


Subject(s)
Bacterial Physiological Phenomena , Intestines/microbiology , Metabolic Diseases/physiopathology , Animals , Colony Count, Microbial , Humans , Metabolic Diseases/microbiology , Prebiotics , Probiotics/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...