Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Benef Microbes ; 13(4): 355-363, 2022 Oct 04.
Article in English | MEDLINE | ID: mdl-35866596

ABSTRACT

Results from high altitude studies in humans and controlled animal experiments suggest that hypoxia exposure induces alterations in gut microbiota composition, which may in turn affect host metabolism. However, well-controlled studies investigating the effects of normobaric hypoxia exposure on gut microbiota composition in humans are lacking. The aim of this study was to explore the impact of mild intermittent hypoxia (MIH) exposure on gut microbiota composition in men with overweight and/or obesity. We performed a randomised, single-blind crossover study, in which participants were exposed to MIH (FiO2: 15%, 3×2 h per day) and normoxia (FiO2: 21%) for seven consecutive days. Following the MIH and normoxia exposure regimens, faecal samples were collected for determination of faecal microbiota composition using 16S rRNA gene-amplicon sequencing in the morning of day 8. Paired faecal samples were available for five individuals. Furthermore, tissue-specific insulin sensitivity was determined using the gold-standard two-step hyperinsulinemic-euglycemic clamp. MIH did not affect microbial alpha and beta-diversity but reduced the relative abundance of Christensenellaceae and Clostridiaceae bacterial families. MIH significantly increased the abundances of obligate anaerobic bacterial genera including Fusicatenibacter, Butyricicoccus and Holdemania, whilst reducing Christensenellaceae R-7 group and Clostridium sensu stricto 1, although these findings were not statistically significant after correction for multiple testing. Furthermore, MIH-induced alterations in abundances of several genera were associated with changes in metabolic parameters such as adipose and peripheral insulin sensitivity, plasma levels of insulin, fatty acids, triacylglycerol and lactate, and substrate oxidation. In conclusion, we demonstrate for the first time that MIH exposure induces modest effects on faecal microbiota composition in humans, shifting several bacterial families and genera towards higher abundances of anaerobic butyrate-producing bacteria. Moreover, MIH-induced effects on faecal microbial composition were associated with parameters related to glucose and lipid homeostasis, supporting a link between MIH-induced alterations in faecal microbiota composition and host metabolism. The study was registered at the Netherlands Trial Register: NL7120/NTR7325.


Subject(s)
Gastrointestinal Microbiome , Insulin Resistance , Probiotics , Animals , Butyrates/analysis , Cross-Over Studies , Fatty Acids/analysis , Feces/microbiology , Glucose/metabolism , Humans , Hypoxia , Insulin , Lactates , Lipids/analysis , Male , Obesity/microbiology , Overweight/complications , RNA, Ribosomal, 16S/analysis , RNA, Ribosomal, 16S/genetics , Single-Blind Method , Triglycerides/analysis
2.
Benef Microbes ; 12(3): 259-266, 2021 Jun 15.
Article in English | MEDLINE | ID: mdl-33880973

ABSTRACT

The gut microbiota may affect host metabolic health through microbial metabolites. The balance between the production of microbial metabolites by saccharolytic and proteolytic fermentation may be an important determinant of metabolic health. Amongst the best-studied saccharolytic microbial metabolites are the short-chain fatty acids acetate, propionate and butyrate. However, human data on the role of other microbial fermentation by-products in metabolic health are greatly lacking. Therefore, we compared in a cross-sectional study the faecal microbial metabolites (caproate, lactate, valerate, succinate, and the branched-chain fatty acids (BCFA) (isobutyrate, isovalerate)) between insulin sensitive (homeostatic model assessment of insulin resistance (HOMA-IR), HOMA-IR<1.85, IS) and insulin resistant (HOMA-IR>1.85, IR) individuals. Additionally, we assessed the relationships between faecal metabolites and markers of metabolic health including fasting glucose, insulin, free fatty acids, insulin resistance (HOMA-IR) and fasting substrate oxidation in 86 individuals with a wide range of body mass index. Faecal metabolite concentrations did not significantly differ between IS and IR. Furthermore, there were no associations between microbial metabolites and metabolic health markers, except for a slight positive association of isovalerate with carbohydrate oxidation (E%, std ß 0.194, P=0.011) and fat oxidation (E%, std ß -0.075, P=0.047), also after adjustment for age, sex and BMI. In summary, faecal caproate, lactate, valerate, succinate, and BCFA (isobutyrate, isovalerate) were not different between IR and IS individuals, nor was there any association between these faecal metabolites and parameters of metabolic health. Further human intervention studies are warranted to investigate the role of these microbially-derived fermentation products and their kinetics in metabolic health and insulin sensitivity.


Subject(s)
Feces/chemistry , Gastrointestinal Microbiome/physiology , Insulin Resistance/physiology , Adult , Aged , Blood Glucose/analysis , Body Weights and Measures , Cross-Sectional Studies , Fatty Acids, Nonesterified/blood , Fatty Acids, Volatile/analysis , Fatty Acids, Volatile/chemistry , Feces/microbiology , Female , Fermentation , Humans , Insulin/blood , Lactic Acid/analysis , Male , Middle Aged , Succinic Acid/analysis , Young Adult
3.
Int J Obes (Lond) ; 45(4): 818-827, 2021 04.
Article in English | MEDLINE | ID: mdl-33473175

ABSTRACT

BACKGROUND/OBJECTIVES: To investigate (1) the association of four VDR polymorphisms (TaqI/rs731236, ApaI/rs7975232, FokI/rs10735810, and Bsml/rs1544410) with markers of adiposity and tissue-specific insulin resistance at baseline, after weight loss and weight maintenance; (2) the effect of the VDR polymorphisms in the SAT transcriptome in overweight/obese Caucasians of the DiOGenes cohort. METHODS: We included 553 adult obese individuals (mean BMI 34.8 kg/m2), men (n = 197) and women (n = 356) at baseline, following an 8-week weight loss intervention and 26 weeks weight maintenance. Genotyping was performed using an Illumina 660W-Quad SNP chip on the Illumina iScan Genotyping System. Tissue-specific IR was determined using Hepatic Insulin Resistance Index (HIRI), Muscle Insulin Sensitivity Index (MISI), and Adipose Tissue Insulin Resistance Index (Adipo-IR). Expression quantitative trait loci (eQTL) analysis was performed to determine the effect of SNPs on SAT gene expression. RESULTS: None of the VDR polymorphisms were associated with HIRI or MISI. Interestingly, carriers of the G allele of VDR FokI showed higher Adipo-IR (GG + GA 7.8 ± 0.4 vs. AA 5.6 ± 0.5, P = 0.010) and higher systemic FFA (GG + GA: 637.8 ± 13.4 vs. AA: 547.9 ± 24.7 µmol/L, P = 0.011), even after adjustment with age, sex, center, and FM. However, eQTL analysis showed minor to no effect of these genotypes on the transcriptional level in SAT. Also, VDR polymorphisms were not related to changes in body weight and IR as result of dietary intervention (P > 0.05 for all parameters). CONCLUSIONS: The VDR Fokl variant is associated with elevated circulating FFA and Adipo-IR at baseline. Nevertheless, minor to no effect of VDR SNPs on the transcriptional level in SAT, indicating that putative mechanisms of action remain to be determined. Finally, VDR SNPs did not affect dietary intervention outcome in the present cohort.


Subject(s)
Insulin Resistance/genetics , Obesity/genetics , Receptors, Calcitriol/genetics , Adult , Alleles , Body Composition , Female , Genotype , Haplotypes , Humans , Linkage Disequilibrium , Male , Middle Aged , Overweight/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Randomized Controlled Trials as Topic , Transcriptome , White People
4.
Benef Microbes ; 11(5): 411-455, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32865024

ABSTRACT

Evidence is accumulating that short chain fatty acids (SCFA) play an important role in the maintenance of gut and metabolic health. The SCFA acetate, propionate and butyrate are produced from the microbial fermentation of indigestible carbohydrates and appear to be key mediators of the beneficial effects elicited by the gut microbiome. Microbial SCFA production is essential for gut integrity by regulating the luminal pH, mucus production, providing fuel for epithelial cells and effects on mucosal immune function. SCFA also directly modulate host metabolic health through a range of tissue-specific mechanisms related to appetite regulation, energy expenditure, glucose homeostasis and immunomodulation. Therefore, an increased microbial SCFA production can be considered as a health benefit, but data are mainly based on animal studies, whereas well-controlled human studies are limited. In this review an expert group by ILSI Europe's Prebiotics Task Force discussed the current scientific knowledge on SCFA to consider the relationship between SCFA and gut and metabolic health with a particular focus on human evidence. Overall, the available mechanistic data and limited human data on the metabolic consequences of elevated gut-derived SCFA production strongly suggest that increasing SCFA production could be a valuable strategy in the preventing gastro-intestinal dysfunction, obesity and type 2 diabetes mellitus. Nevertheless, there is an urgent need for well controlled longer term human SCFA intervention studies, including measurement of SCFA fluxes and kinetics, the heterogeneity in response based on metabolic phenotype, the type of dietary fibre and fermentation site in fibre intervention studies and the control for factors that could shape the microbiome like diet, physical activity and use of medication.


Subject(s)
Fatty Acids, Volatile/metabolism , Gastrointestinal Diseases/prevention & control , Gastrointestinal Microbiome , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Animals , Carbohydrate Metabolism , Diabetes Mellitus, Type 2/prevention & control , Host Microbial Interactions , Humans , Obesity/prevention & control , Prebiotics
5.
Sci Rep ; 9(1): 4202, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862933

ABSTRACT

Body fat distribution is an important determinant of cardiometabolic health. Lower-body adipose tissue (AT) has protective characteristics as compared to upper-body fat, but the underlying depot-differences remain to be elucidated. Here, we compared the proteome and morphology of abdominal and femoral AT. Paired biopsies from abdominal and femoral subcutaneous AT were taken from eight overweight/obese (BMI ≥ 28 kg/m2) women with impaired glucose metabolism after an overnight fast. Proteins were isolated and quantified using liquid chromatography-mass spectrometry, and protein expression in abdominal and femoral subcutaneous AT was compared. Moreover, correlations between fat cell size and the proteome of both AT depots were determined. In total, 651 proteins were identified, of which 22 proteins tended to be differentially expressed between abdominal and femoral AT after removal of blood protein signals (p < 0.05). Proteins involved in cell structure organization and energy metabolism were differently expressed between AT depots. Fat cell size, which was higher in femoral AT, was significantly correlated with ADH1B, POSTN and LCP1. These findings suggest that there are only slight differences in protein expression between abdominal and femoral subcutaneous AT. It remains to be determined whether these differences, as well as differences in protein activity, contribute to functional and/or morphological differences between these fat depots.


Subject(s)
Abdominal Fat/metabolism , Obesity/metabolism , Proteome/metabolism , Subcutaneous Fat/metabolism , Abdominal Fat/pathology , Female , Humans , Middle Aged , Obesity/pathology , Proteomics , Subcutaneous Fat/pathology
6.
Sci Rep ; 8(1): 4677, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549282

ABSTRACT

Obesity is associated with a disturbed adipose tissue (AT) function characterized by adipocyte hypertrophy, an impaired lipolysis and pro-inflammatory phenotype, which contributes to insulin resistance (IR). We investigated whether AT phenotype in different AT depots of obese individuals with and without type 2 diabetes mellitus (T2DM) is associated with whole-body IR. Subcutaneous (SC) and visceral (V) AT biopsies from 18 lean, 17 obese and 8 obese T2DM men were collected. AT phenotype was characterized by ex vivo measurement of basal and stimulated lipolysis (mature adipocytes), adipocyte size distribution (AT tissue sections) and AT immune cells (flow cytometry). In VAT, mean adipocyte size, CD45+ leukocytes and M1 macrophages were significantly increased in both obese groups compared to lean individuals. In SCAT, despite adipocyte hypertrophy, no significant differences in immune cell populations between groups were found. In SCAT, multiple linear regression analysis showed that none of the AT phenotype markers independently contributed to HOMA-IR while in VAT, mean adipocyte size was significantly related to HOMA-IR. In conclusion, beside adipocyte hypertrophy in VAT, M1 macrophage- or B-cell-mediated inflammation, may contribute to IR, while inflammation in hypertrophic SCAT does not seem to play a major role in IR.


Subject(s)
Diabetes Mellitus, Type 2/immunology , Intra-Abdominal Fat/pathology , Obesity/immunology , Subcutaneous Fat/pathology , Adipocytes , Case-Control Studies , Diabetes Mellitus, Type 2/pathology , Humans , Insulin Resistance , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/immunology , Leukocyte Common Antigens/metabolism , Linear Models , Lipolysis , Macrophages/immunology , Male , Middle Aged , Obesity/complications , Obesity/pathology , Phenotype , Subcutaneous Fat/cytology , Subcutaneous Fat/immunology
7.
Sci Rep ; 8(1): 3933, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29500454

ABSTRACT

Increased activation of the renin-angiotensin system is involved in the onset and progression of cardiometabolic diseases, while natriuretic peptides (NP) may exert protective effects. We have recently demonstrated that sacubitril/valsartan (LCZ696), a first-in-class angiotensin receptor neprilysin inhibitor, which blocks the angiotensin II type-1 receptor and augments natriuretic peptide levels, improved peripheral insulin sensitivity in obese hypertensive patients. Here, we investigated the effects of sacubitril/valsartan (400 mg QD) treatment for 8 weeks on the abdominal subcutaneous adipose tissue (AT) phenotype compared to the metabolically neutral comparator amlodipine (10 mg QD) in 70 obese hypertensive patients. Abdominal subcutaneous AT biopsies were collected before and after intervention to determine the AT transcriptome and expression of proteins involved in lipolysis, NP signaling and mitochondrial oxidative metabolism. Both sacubitril/valsartan and amlodipine treatment did not significantly induce AT transcriptional changes in pathways related to lipolysis, NP signaling and oxidative metabolism. Furthermore, protein expression of adipose triglyceride lipase (ATGL) (Ptime*group = 0.195), hormone-sensitive lipase (HSL) (Ptime*group = 0.458), HSL-ser660 phosphorylation (Ptime*group = 0.340), NP receptor-A (NPRA) (Ptime*group = 0.829) and OXPHOS complexes (Ptime*group = 0.964) remained unchanged. In conclusion, sacubitril/valsartan treatment for 8 weeks did not alter the abdominal subcutaneous AT transcriptome and expression of proteins involved in lipolysis, NP signaling and oxidative metabolism in obese hypertensive patients.


Subject(s)
Adipose Tissue/drug effects , Aminobutyrates/therapeutic use , Angiotensin Receptor Antagonists/therapeutic use , Hypertension/drug therapy , Neprilysin/antagonists & inhibitors , Obesity/metabolism , Proteins/metabolism , Tetrazoles/therapeutic use , Transcriptome , Adipose Tissue/metabolism , Adult , Aminobutyrates/pharmacology , Amlodipine/pharmacology , Angiotensin Receptor Antagonists/pharmacology , Biphenyl Compounds , Double-Blind Method , Drug Combinations , Female , Humans , Hypertension/complications , Hypertension/metabolism , Male , Middle Aged , Obesity/complications , Subcutaneous Fat/metabolism , Tetrazoles/pharmacology , Valsartan
8.
Obes Rev ; 18(11): 1243-1259, 2017 11.
Article in English | MEDLINE | ID: mdl-28901677

ABSTRACT

Natriuretic peptides have long been known for their cardiovascular function. However, a growing body of evidence emphasizes the role of natriuretic peptides in human substrate and energy metabolism, thereby connecting the heart with several insulin-sensitive organs like adipose tissue, skeletal muscle and liver. Obesity may be associated with an impaired regulation of the natriuretic peptide system, also indicated as a natriuretic handicap. Evidence points towards a contribution of this natriuretic handicap to the development of obesity, type 2 diabetes mellitus and cardiometabolic complications, although the causal relationship is not fully understood. Nevertheless, targeting the natriuretic peptide pathway may improve metabolic health in obesity and type 2 diabetes mellitus. This review will focus on current literature regarding the metabolic roles of natriuretic peptides with emphasis on lipid metabolism and insulin sensitivity. Furthermore, it will be discussed how exercise and lifestyle intervention may modulate the natriuretic peptide-related metabolic effects.


Subject(s)
Insulin Resistance , Lipid Metabolism , Natriuretic Peptides/blood , Natriuretic Peptides/deficiency , Adipokines/blood , Diabetes Mellitus, Type 2/blood , Exercise , Humans , Life Style , Liver/metabolism , Muscle, Skeletal/metabolism , Natriuretic Peptides/metabolism , Obesity/blood , Receptors, Atrial Natriuretic Factor
9.
Benef Microbes ; 8(4): 557-562, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28618864

ABSTRACT

Gut microbiota composition may play an important role in the development of obesity-related comorbidities. However, only few studies have investigated gender-differences in microbiota composition and gender-specific associations between microbiota or microbial products and insulin sensitivity. Insulin sensitivity (hyperinsulinemic-euglycemic clamp), body composition (dual energy X-ray absorptiometry), substrate oxidation (indirect calorimetry), systemic inflammatory markers and microbiota composition (PCR) were determined in male (n=15) and female (n=14) overweight and obese subjects. Bacteroidetes/Firmicutes-ratio was higher in men than in women (P=0.001). Bacteroidetes/Firmicutes-ratio was inversely related to peripheral insulin sensitivity only in men (men: P=0.003, women: P=0.882). This association between Bacteroidetes/Firmicutes-ratio and peripheral insulin sensitivity did not change after adjustment for dietary fibre and saturated fat intake, body composition, fat oxidation and markers of inflammation. Bacteroidetes/Firmicutes-ratio was not associated with hepatic insulin sensitivity. Men and women differ in microbiota composition and its impact on insulin sensitivity, implying that women might be less sensitive to gut microbiota-induced metabolic aberrations than men. This trial was registered at clinicaltrials.gov as NCT02381145.


Subject(s)
Bacteroidetes/isolation & purification , Firmicutes/isolation & purification , Gastrointestinal Microbiome , Insulin/metabolism , Obesity/microbiology , Adult , Bacteroidetes/classification , Bacteroidetes/genetics , Feces/microbiology , Female , Firmicutes/classification , Firmicutes/genetics , Glucose/metabolism , Humans , Insulin Resistance , Male , Obesity/metabolism
10.
Eur J Clin Nutr ; 71(9): 1040-1045, 2017 09.
Article in English | MEDLINE | ID: mdl-28589947

ABSTRACT

BACKGROUND/OBJECTIVES: The intestinal microbiota may have a profound impact on host metabolism. As evidence suggests that polyphenols affect substrate utilization, the present study aimed to investigate the effects of polyphenol supplementation on intestinal microbiota composition in humans. Furthermore, we examined whether (changes in) gut microbiota composition may determine the metabolic response to polyphenol supplementation. SUBJECTS/METHODS: In this randomized, double-blind, placebo (PLA)-controlled trial, 37 overweight and obese men and women (18 males/19 females, 37.8±1.6 years, body mass index: 29.6±0.5 kg/m2) received either epigallocatechin-3-gallate and resveratrol (EGCG+RES, 282 and 80 mg/day, respectively) or PLA for 12 weeks. Before and after intervention, feces samples were collected to determine microbiota composition. Fat oxidation was assessed by indirect calorimetry during a high-fat mixed meal test (2.6 MJ, 61 energy% fat) and skeletal muscle mitochondrial oxidative capacity by means of ex vivo respirometry on isolated skeletal muscle fibers. Body composition was measured by dual-energy X-ray absorptiometry. RESULTS: Fecal abundance of Bacteroidetes was higher in men as compared with women, whereas other assessed bacterial taxa were comparable. EGCG+RES supplementation significantly decreased Bacteroidetes and tended to reduce Faecalibacterium prausnitzii in men (P=0.05 and P=0.10, respectively) but not in women (P=0.15 and P=0.77, respectively). Strikingly, baseline Bacteroidetes abundance was predictive for the EGCG+RES-induced increase in fat oxidation in men but not in women. Other bacterial genera and species were not affected by EGCG+RES supplementation. CONCLUSIONS: We demonstrated that 12-week EGCG+RES supplementation affected the gut microbiota composition in men but not in women. Baseline microbiota composition determined the increase in fat oxidation after EGCG+RES supplementation in men.


Subject(s)
Catechin/analogs & derivatives , Gastrointestinal Microbiome/drug effects , Overweight/drug therapy , Polyphenols/administration & dosage , Stilbenes/administration & dosage , Absorptiometry, Photon , Adult , Bacteroidetes/isolation & purification , Catechin/administration & dosage , Catechin/pharmacology , Double-Blind Method , Energy Metabolism , Faecalibacterium prausnitzii/isolation & purification , Feces/microbiology , Female , Humans , Male , Muscle, Skeletal/metabolism , Overweight/metabolism , Overweight/microbiology , Polyphenols/pharmacology , Resveratrol , Stilbenes/pharmacology , Treatment Outcome
11.
Chem Phys Lipids ; 207(Pt B): 239-245, 2017 10.
Article in English | MEDLINE | ID: mdl-28647339

ABSTRACT

BACKGROUND: Changes in the microbiota composition have been implicated in the development of obesity and type 2 diabetes. However, not much is known on the involvement of gut microbiota in lipid and cholesterol metabolism. In addition, the gut microbiota might also be a potential source of plasma oxyphytosterol and oxycholesterol concentrations (oxidation products of plant sterols and cholesterol). Therefore, the aim of this study was to modulate the gut microbiota by antibiotic therapy to investigate effects on parameters reflecting cholesterol metabolism and oxyphytosterol concentrations. DESIGN: A randomized, double blind, placebo-controlled trial was performed in which 55 obese, pre-diabetic men received oral amoxicillin (broad-spectrum antibiotic), vancomycin (antibiotic directed against Gram-positive bacteria) or placebo (microcrystalline cellulose) capsules for 7days (1500mg/day). Plasma lipid and lipoprotein, non-cholesterol sterol, bile acid and oxy(phyto)sterol concentrations were determined at baseline and after 1-week intervention. RESULTS: Plasma secondary bile acids correlated negatively with cholestanol (marker for cholesterol absorption, r=-0.367; P<0.05) and positively with lathosterol concentrations (marker for cholesterol synthesis, r=0.430; P<0.05). Fasting plasma secondary bile acid concentrations were reduced after vancomycin treatment as compared to placebo treatment (-0.24±0.22µmol/L vs. -0.08±0.29µmol/L; P<0.01). Vancomycin and amoxicillin treatment did not affect markers for cholesterol metabolism, plasma TAG, total cholesterol, LDL-C or HDL-C concentrations as compared to placebo. In addition, both antibiotic treatments did not affect individual isoforms or total plasma oxyphytosterol or oxycholesterol concentrations. CONCLUSION: Despite strong correlations between plasma bile acid concentrations and cholesterol metabolism (synthesis and absorption), amoxicillin and vancomycin treatment for 7days did not affect plasma lipid and lipoprotein, plasma non-cholesterol sterol and oxy(phyto)sterol concentrations in obese, pre-diabetic men.


Subject(s)
Amoxicillin/pharmacology , Anti-Bacterial Agents/pharmacology , Cholesterol/metabolism , Vancomycin/pharmacology , Administration, Oral , Amoxicillin/administration & dosage , Anti-Bacterial Agents/administration & dosage , Bile Acids and Salts/blood , Bile Acids and Salts/metabolism , Cholesterol/blood , Double-Blind Method , Humans , Male , Middle Aged , Obesity/blood , Obesity/metabolism , Prediabetic State/blood , Prediabetic State/metabolism , Vancomycin/administration & dosage
13.
Int J Obes (Lond) ; 41(5): 722-728, 2017 05.
Article in English | MEDLINE | ID: mdl-28179648

ABSTRACT

BACKGROUND/OBJECTIVES: Although adipose tissue (AT) hypoxia is present in rodent models of obesity, evidence for this in humans is limited. Here, we investigated the effects of diet-induced weight loss (WL) on abdominal subcutaneous AT oxygen tension (pO2), AT blood flow (ATBF), AT capillary density, AT morphology and transcriptome, systemic inflammatory markers and insulin sensitivity in humans. SUBJECTS/METHODS: Fifteen overweight and obese individuals underwent a dietary intervention (DI), consisting of a 5-week very-low-calorie diet (VLCD, 500 kcal day-1; WL), and a subsequent 4-week weight stable diet (WS). Body composition, AT pO2 (optochemical monitoring), ATBF (133Xe washout), and whole-body insulin sensitivity were determined, and AT biopsies were collected at baseline, end of WL (week 5) and end of WS (week 9). RESULTS: Body weight, body fat percentage and adipocyte size decreased significantly during the DI period. The DI markedly decreased AT pO2 and improved insulin sensitivity, but did not alter ATBF. Finally, the DI increased AT gene expression of pathways related to mitochondrial biogenesis and non-mitochondrial oxygen consumption. CONCLUSIONS: VLCD-induced WL markedly decreases abdominal subcutaneous AT pO2, which is paralleled by a reduction in adipocyte size, increased AT gene expression of mitochondrial biogenesis markers and non-mitochondrial oxygen consumption pathways, and improved whole-body insulin sensitivity in humans.


Subject(s)
Inflammation/physiopathology , Insulin Resistance/physiology , Insulin/metabolism , Obesity/physiopathology , Overweight/physiopathology , Oxygen/metabolism , Subcutaneous Fat, Abdominal/metabolism , Weight Loss/physiology , Adipocytes/physiology , Cell Hypoxia/physiology , Diet, Reducing , Female , Gene Expression Regulation , Humans , Inflammation/diet therapy , Inflammation/metabolism , Male , Middle Aged , Obesity/diet therapy , Obesity/metabolism , Overweight/diet therapy , Overweight/metabolism , Oxygen Consumption , Phenotype , Treatment Outcome
14.
Amino Acids ; 49(1): 161-172, 2017 01.
Article in English | MEDLINE | ID: mdl-27714515

ABSTRACT

Plasma levels of several amino acids are correlated with metabolic dysregulation in obesity and type 2 diabetes. To increase our understanding of human amino-acid metabolism, we aimed to determine splanchnic interorgan amino-acid handling. Twenty patients planned to undergo a pylorus preserving pancreatico-duodenectomy were included in this study. Blood was sampled from the portal vein, hepatic vein, superior mesenteric vein, inferior mesenteric vein, splenic vein, renal vein, and the radial artery during surgery. The difference between arterial and venous concentrations of 21 amino acids was determined using liquid chromatography as a measure of amino-acid metabolism across a given organ. Whereas glutamine was significantly taken up by the small intestine (121.0 ± 23.8 µmol/L; P < 0.0001), citrulline was released (-36.1 ± 4.6 µmol/L; P < 0.0001). This, however, was not seen for the colon. Interestingly, the liver showed a small, but a significant uptake of citrulline from the circulation (4.8 ± 1.6 µmol/L; P = 0.0138) next to many other amino acids. The kidneys showed a marked release of serine and alanine into the circulation (-58.0 ± 4.4 µmol/L and -61.8 ± 5.2 µmol/L, P < 0.0001), and a smaller, but statistically significant release of tyrosine (-12.0 ± 1.3 µmol/L, P < 0.0001). The spleen only released taurine (-9.6 ± 3.3 µmol/L; P = 0.0078). Simultaneous blood sampling in different veins provides unique qualitative and quantitative information on integrative amino-acid physiology, and reveals that the well-known intestinal glutamine-citrulline pathway appears to be functional in the small intestine but not in the colon.


Subject(s)
Amino Acids/blood , Duodenal Neoplasms/metabolism , Pancreatic Neoplasms/metabolism , Pancreaticoduodenectomy/methods , Splanchnic Circulation/physiology , Aged , Colon/blood supply , Colon/metabolism , Duodenal Neoplasms/blood supply , Duodenal Neoplasms/surgery , Female , Hepatic Veins/metabolism , Humans , Intestine, Small/blood supply , Intestine, Small/metabolism , Kidney/blood supply , Kidney/metabolism , Liver/blood supply , Liver/metabolism , Male , Mesenteric Veins/metabolism , Middle Aged , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/surgery , Portal Vein/metabolism , Radial Artery/metabolism , Renal Veins/metabolism , Spleen/blood supply , Spleen/metabolism , Splenic Vein/metabolism
15.
Clin Pharmacol Ther ; 101(2): 254-263, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27542885

ABSTRACT

Natriuretic peptide (NP) deficiency and sustained renin-angiotensin system activation are associated with impaired oxidative metabolism and predispose to type-2 diabetes. We hypothesized that sacubitril/valsartan (LCZ696), which augments NP through neprilysin inhibition while blocking angiotensin II type-1 (AT1 )-receptors, improves insulin sensitivity, lipid mobilization, and oxidation. After 8 weeks of treatment of obese patients with hypertension, sacubitril/valsartan 400 mg q.d., but not amlodipine 10 mg q.d., was associated with a significant increase from baseline in the insulin sensitivity index (hyperinsulinemic-euglycemic clamp), and tended to be higher in patients treated with sacubitril/valsartan compared to amlodipine. Abdominal adipose tissue interstitial glycerol concentrations increased with sacubitril/valsartan, but decreased with amlodipine. Whole-body lipolysis and substrate oxidation did not change with either treatment. Results confirm that sacubitril/valsartan treatment leads to a metabolic benefit in the study population and supports the relevance of neprilysin inhibition along with AT1 -receptor blockade in the regulation of human glucose and lipid metabolism.


Subject(s)
Aminobutyrates/pharmacology , Antihypertensive Agents/pharmacology , Insulin Resistance/physiology , Neprilysin/antagonists & inhibitors , Obesity/metabolism , Tetrazoles/pharmacology , Adipose Tissue/drug effects , Adult , Aminobutyrates/therapeutic use , Amlodipine/pharmacology , Angiotensin II Type 1 Receptor Blockers/metabolism , Biphenyl Compounds , Drug Combinations , Energy Metabolism/drug effects , Female , Glycerol/analysis , Humans , Hypertension/drug therapy , Lipid Metabolism/drug effects , Male , Middle Aged , Natriuretic Peptides/genetics , Natriuretic Peptides/metabolism , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Tetrazoles/therapeutic use , Valsartan
16.
Int J Obes (Lond) ; 40(3): 507-13, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26471343

ABSTRACT

BACKGROUND/OBJECTIVES: Adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) are key enzymes involved in intracellular lipid catabolism. We have previously shown decreased expression and activity of these lipases in adipose tissue of obese insulin resistant individuals. Here we hypothesized that lipase deficiency might impact on insulin sensitivity and metabolic homeostasis in adipocytes not just by enhancing lipid accumulation, but also by altering lipid and carbohydrate catabolism in a peroxisome proliferator-activated nuclear receptor (PPAR)-dependent manner. METHODS: To address our hypothesis, we performed a series of in vitro experiments in a human white adipocyte model, the human multipotent adipose-derived stem (hMADS) cells, using genetic (siRNA) and pharmacological knockdown of ATGL and/or HSL. RESULTS: We show that ATGL and HSL knockdown in hMADS adipocytes disrupted mitochondrial respiration, which was accompanied by a decreased oxidative phosphorylation (OxPhos) protein content. This lead to a reduced exogenous and endogenous palmitate oxidation following ATGL knockdown, but not in HSL deficient adipocytes. ATGL deficiency was followed by excessive triacylglycerol accumulation, and HSL deficiency further increased diacylglycerol accumulation. Both single and double lipase knockdown reduced insulin-stimulated glucose uptake, which was attributable to impaired insulin signaling. These effects were accompanied by impaired activation of the nuclear receptor PPARα, and restored on PPARα agonist treatment. CONCLUSIONS: The present study indicates that lipase deficiency in human white adipocytes contributes to mitochondrial dysfunction and insulin resistance, in a PPARα-dependent manner. Therefore, modulation of adipose tissue lipases may provide a promising strategy to reverse insulin resistance in obese and type 2 diabetic patients.


Subject(s)
Adipocytes, White/metabolism , Adiposity/physiology , Insulin Resistance/physiology , Lipase/deficiency , Mitochondria/metabolism , Obesity/metabolism , PPAR alpha/agonists , Cells, Cultured , Energy Metabolism , Humans , Lipase/physiology , Lipolysis/physiology , Obesity/complications , Sterol Esterase/metabolism
17.
Obes Rev ; 16(9): 715-57, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26179344

ABSTRACT

Disturbances in fatty acid metabolism in adipose tissue, liver, skeletal muscle, gut and pancreas play an important role in the development of insulin resistance, impaired glucose metabolism and type 2 diabetes mellitus. Alterations in diet composition may contribute to prevent and/or reverse these disturbances through modulation of fatty acid metabolism. Besides an increased fat mass, adipose tissue dysfunction, characterized by an altered capacity to store lipids and an altered secretion of adipokines, may result in lipid overflow, systemic inflammation and excessive lipid accumulation in non-adipose tissues like liver, skeletal muscle and the pancreas. These impairments together promote the development of impaired glucose metabolism, insulin resistance and type 2 diabetes mellitus. Furthermore, intrinsic functional impairments in either of these organs may contribute to lipotoxicity and insulin resistance. The present review provides an overview of fatty acid metabolism-related pathways in adipose tissue, liver, skeletal muscle, pancreas and gut, which can be targeted by diet or food components, thereby improving glucose metabolism.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Fatty Acids, Nonesterified/metabolism , Glucose/metabolism , Insulin Resistance , Muscle, Skeletal/metabolism , Obesity/metabolism , Pancreas/metabolism , Diabetes Mellitus, Type 2/etiology , Diet , Humans , Obesity/complications , Receptor Cross-Talk
18.
Int J Obes (Lond) ; 38(3): 470-3, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23958793

ABSTRACT

Polyphenolic compounds, such as resveratrol, have recently received widespread interest because of their ability to mimic effects of calorie restriction. The objective of the present study was to gain more insight into the effects of 30 days resveratrol supplementation on adipose tissue morphology and underlying processes. Eleven healthy obese men were supplemented with placebo and resveratrol for 30 days (150 mg per day), separated by a 4-week washout period in a double-blind randomized crossover design. A postprandial abdominal subcutaneous adipose tissue biopsy was collected to assess adipose tissue morphology and gene expression using microarray analysis. Resveratrol significantly decreased adipocyte size, with a shift toward a reduction in the proportion of large and very-large adipocytes and an increase in small adipocytes. Microarray analysis revealed downregulation of Wnt and Notch signaling pathways and upregulation of pathways involved in cell cycle regulation after resveratrol supplementation, suggesting enhanced adipogenesis. Furthermore, lysosomal/phagosomal pathway and transcription factor EB were upregulated reflecting an alternative pathway of lipid breakdown by autophagy. Further research is necessary to investigate whether resveratrol improves adipose tissue function.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/metabolism , Enzyme Inhibitors/therapeutic use , Obesity/drug therapy , Stilbenes/therapeutic use , Adipogenesis/drug effects , Adipose Tissue/drug effects , Adult , Aged , Cross-Over Studies , Double-Blind Method , Gene Expression Profiling , Humans , Male , Middle Aged , Obesity/genetics , Obesity/metabolism , Receptors, Notch/drug effects , Receptors, Notch/metabolism , Resveratrol , Signal Transduction/drug effects , Treatment Outcome , Wnt Signaling Pathway/drug effects
19.
Int J Obes (Lond) ; 38(5): 698-706, 2014 May.
Article in English | MEDLINE | ID: mdl-24317366

ABSTRACT

BACKGROUND AND OBJECTIVES: Impaired regulation of lipid oxidation (metabolic inflexibility) is associated with obesity and type 2 diabetes mellitus. Recent evidence has indicated that dietary polyphenols may modulate mitochondrial function, substrate metabolism and energy expenditure in humans. The present study investigated the effects of short-term supplementation of two combinations of polyphenols on energy expenditure (EE) and substrate metabolism in overweight subjects. SUBJECTS AND METHODS: Eighteen healthy overweight volunteers (9 women, 9 men; age 35±2.5 years; body mass index 28.9±0.4 kg m(-2)) participated in a randomized, double-blind cross-over trial. Combinations of epigallocatechin-gallate (E, 282 mg day(-1))+resveratrol (R, 200 mg day(-1)) and E+R+80 mg day(-1) soy isoflavones (S) or placebo capsules (PLA) were supplemented twice daily for a period of 3 days. On day 3, circulating metabolite concentrations, EE and substrate oxidation (using indirect calorimetry) were measured during fasting and postprandial conditions for 6 h (high-fat-mixed meal (2.6 MJ, 61.2 E% fat)). RESULTS: Short-term supplementation of E+R increased resting EE (E+R vs PLA: 5.45±0.24 vs 5.23±0.25 kJ min(-1), P=0.039), whereas both E+R (699±18 kJ 120 min(-1) vs 676±20 kJ 120 min(-1), P=0.028) and E+R+S (704±18 kJ 120 min(-1) vs 676±20 kJ 120 min(-1), P=0.014) increased 2-4 h-postprandial EE compared with PLA. Metabolic flexibility, calculated as the postprandial increase to the highest respiratory quotient achieved, tended to be improved by E+R compared with PLA and E+R+S only in men (E+R vs PLA: 0.11±0.02 vs 0.06±0.02, P=0.059; E+R+S: 0.03±0.02, P=0.009). E+R+S significantly increased fasting plasma free fatty acid (P=0.064) and glycerol (P=0.021) concentrations compared with PLA. CONCLUSIONS: We demonstrated for the first time that combined E+R supplementation for 3 days significantly increased fasting and postprandial EE, which was accompanied by improved metabolic flexibility in men but not in women. Addition of soy isoflavones partially reversed these effects possibly due to their higher lipolytic potential. The present findings may imply that long-term supplementation of these dosages of epigallocatechin-gallate combined with resveratrol may improve metabolic health and body weight regulation.


Subject(s)
Antioxidants/therapeutic use , Catechin/analogs & derivatives , Energy Metabolism/drug effects , Lipid Metabolism/drug effects , Overweight/diet therapy , Polyphenols/therapeutic use , Stilbenes/therapeutic use , Adult , Body Mass Index , Catechin/therapeutic use , Dietary Supplements , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Overweight/metabolism , Oxidation-Reduction , Postprandial Period , Resveratrol , Treatment Outcome
20.
Diabetes Metab Res Rev ; 30(4): 323-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24302564

ABSTRACT

BACKGROUND: Obesity is associated with insulin resistance and chronic low-grade inflammation. Insulin has been described to have anti-inflammatory effects in immune cells. Therefore, insulin resistance in immune cells can be expected to have important consequences for immune function. Here, we investigate whether freshly isolated monocytes and T cells, isolated from study subjects with a normal or disturbed glucometabolic state, respond to insulin with phosphorylation of Akt, a key molecule in the insulin signalling pathway. METHODS: A total of 25 study subjects were enrolled in the study. An oral glucose tolerance test (OGTT) was performed, and from fasting insulin and glucose, the homeostasis model assessment of insulin resistance (HOMA-IR) index was calculated. Peripheral blood mononuclear cells were isolated from heparinized blood and phenotypically characterized by flow cytometry. Basal and insulin-induced fractions of pAkt(S473)-positive monocytes and T cells were determined by Phosflow. RESULTS: On the basis of the OGTT, 11 subjects were classified as normal glucose tolerant (NGT), 9 had an impaired glucose metabolism (IGM) and 5 had type 2 diabetes (T2DM). The fraction of pAkt(S473)positive-T cells and monocytes, in the absence of insulin, was low in all subjects. Incubation with insulin did not induce Akt phosphorylation in CD4⁺ and CD8⁺ T cells in normal subjects. However, in the monocyte fraction, an insulin-dose-dependent increase of the pAkt(S473)positive-cell fraction was observed. This response did not differ between NGT, IGM and T2DM and was not correlated with HOMA-IR. CONCLUSIONS: In this study, we show that freshly isolated monocytes, but not T cells, are insulin-sensitive cells and that this insulin sensitivity of monocytes is not negatively affected by the glucometabolic state of the donor.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Monocytes/metabolism , Prediabetic State/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Aged , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 2/immunology , Female , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Resistance , Male , Middle Aged , Monocytes/drug effects , Monocytes/immunology , Phosphorylation/drug effects , Prediabetic State/immunology , Protein Processing, Post-Translational/drug effects , Serine/chemistry , Signal Transduction/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...