Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33765118

ABSTRACT

Circulating branched chain amino acid (BCAA) levels are elevated in obese humans and genetically obese rodents. However, the relationship of BCAAs to insulin resistance in diet-induced obese mice, a commonly used model to study glucose homeostasis, is still ill-defined. Here we examined how high-fat high-sucrose (HFHS) or high-fat diet (HFD) feeding, with or without BCAA supplementation in water, alters the metabolome in serum/plasma and tissues in mice and whether raising circulating BCAA levels worsens insulin resistance and glucose intolerance. Neither HFHS nor HFD feeding raised circulating BCAA levels in insulin-resistant diet-induced obese mice. BCAA supplementation raised circulating BCAA and branched-chain α-keto acid levels and C5-OH/C3-DC acylcarnitines (AC) in muscle from mice fed an HFHS diet or HFD, but did not worsen insulin resistance. A set of short- and long-chain acyl CoAs were elevated by diet alone in muscle, liver, and white adipose tissue (WAT), but not increased further by BCAA supplementation. HFD feeding reduced valine and leucine oxidation in WAT but not in muscle. BCAA supplementation markedly increased valine oxidation in muscle from HFD-fed mice, while leucine oxidation was unaffected by diet or BCAA treatment. Here we establish an extensive metabolome database showing tissue-specific changes in mice on 2 different HFDs, with or without BCAA supplementation. We conclude that mildly elevating circulating BCAAs and a subset of ACs by BCAA supplementation does not worsen insulin resistance or glucose tolerance in mice. This work highlights major differences in the effects of BCAAs on glucose homeostasis in diet-induced obese mice versus data reported in obese rats and in humans.


Subject(s)
Amino Acids, Branched-Chain/administration & dosage , Blood Glucose/metabolism , Diet/adverse effects , Insulin Resistance/physiology , Metabolomics , Obesity/metabolism , Amino Acids, Branched-Chain/blood , Amino Acids, Branched-Chain/metabolism , Animals , Diet, High-Fat , Dietary Sucrose/administration & dosage , Dietary Supplements , Female , Glucose Intolerance/blood , Homeostasis/drug effects , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Obesity/etiology , Oxidation-Reduction
2.
Cell Rep ; 33(6): 108375, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33176135

ABSTRACT

Glycine levels are inversely associated with branched-chain amino acids (BCAAs) and cardiometabolic disease phenotypes, but biochemical mechanisms that explain these relationships remain uncharted. Metabolites and genes related to BCAA metabolism and nitrogen handling were strongly associated with glycine in correlation analyses. Stable isotope labeling in Zucker fatty rats (ZFRs) shows that glycine acts as a carbon donor for the pyruvate-alanine cycle in a BCAA-regulated manner. Inhibition of the BCAA transaminase (BCAT) enzymes depletes plasma pools of alanine and raises glycine levels. In high-fat-fed ZFRs, dietary glycine supplementation raises urinary acyl-glycine content and lowers circulating triglycerides but also results in accumulation of long-chain acyl-coenzyme As (acyl-CoAs), lower 5' adenosine monophosphate-activated protein kinase (AMPK) phosphorylation in muscle, and no improvement in glucose tolerance. Collectively, these studies frame a mechanism for explaining obesity-related glycine depletion and also provide insight into the impact of glycine supplementation on systemic glucose, lipid, and amino acid metabolism.


Subject(s)
Glycine/metabolism , Liver/physiopathology , Muscle, Skeletal/physiopathology , Nitrogen/metabolism , Obesity/physiopathology , Amino Acids, Branched-Chain/metabolism , Animals , Male , Rats , Rats, Zucker
3.
Cell Metab ; 30(4): 735-753.e4, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31577934

ABSTRACT

Dietary sugars, fructose and glucose, promote hepatic de novo lipogenesis and modify the effects of a high-fat diet (HFD) on the development of insulin resistance. Here, we show that fructose and glucose supplementation of an HFD exert divergent effects on hepatic mitochondrial function and fatty acid oxidation. This is mediated via three different nodes of regulation, including differential effects on malonyl-CoA levels, effects on mitochondrial size/protein abundance, and acetylation of mitochondrial proteins. HFD- and HFD plus fructose-fed mice have decreased CTP1a activity, the rate-limiting enzyme of fatty acid oxidation, whereas knockdown of fructose metabolism increases CPT1a and its acylcarnitine products. Furthermore, fructose-supplemented HFD leads to increased acetylation of ACADL and CPT1a, which is associated with decreased fat metabolism. In summary, dietary fructose, but not glucose, supplementation of HFD impairs mitochondrial size, function, and protein acetylation, resulting in decreased fatty acid oxidation and development of metabolic dysregulation.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Sugars/adverse effects , Fatty Acids/metabolism , Fructose/adverse effects , Liver/metabolism , Mitochondrial Proteins , Obesity/metabolism , Animals , Cell Line , Glucose/adverse effects , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Processing, Post-Translational , Transcription, Genetic
4.
Br J Pharmacol ; 174(24): 4797-4811, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28977680

ABSTRACT

BACKGROUND AND PURPOSE: The human kinome consists of roughly 500 kinases, including 150 that have been proposed as therapeutic targets. Protein kinases regulate an array of signalling pathways that control metabolism, cell cycle progression, cell death, differentiation and survival. It is not surprising, then, that new kinase inhibitors developed to treat cancer, including sorafenib, also exhibit cardiotoxicity. We hypothesized that sorafenib cardiotoxicity is related to its deleterious effects on specific cardiac metabolic pathways given the critical roles of protein kinases in cardiac metabolism. EXPERIMENTAL APPROACH: FVB/N mice (10 per group) were challenged with sorafenib or vehicle control daily for 2 weeks. Echocardiographic assessment of the heart identified systolic dysfunction consistent with cardiotoxicity in sorafenib-treated mice compared to vehicle-treated controls. Heart, skeletal muscle, liver and plasma were flash frozen and prepped for non-targeted GC-MS metabolomics analysis. KEY RESULTS: Compared to vehicle-treated controls, sorafenib-treated hearts exhibited significant alterations in 11 metabolites, including markedly altered taurine/hypotaurine metabolism (25-fold enrichment), identified by pathway enrichment analysis. CONCLUSIONS AND IMPLICATIONS: These studies identified alterations in taurine/hypotaurine metabolism in the hearts and skeletal muscles of mice treated with sorafenib. Interventions that rescue or prevent these sorafenib-induced changes, such as taurine supplementation, may be helpful in attenuating sorafenib-induced cardiac injury.


Subject(s)
Heart/drug effects , Liver/drug effects , Metabolomics , Muscle, Skeletal/drug effects , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Plasma/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Liver/metabolism , Mice , Mice, Inbred Strains , Muscle, Skeletal/metabolism , Niacinamide/chemistry , Niacinamide/pharmacology , Phenylurea Compounds/chemistry , Plasma/metabolism , Protein Kinase Inhibitors/chemistry , Sorafenib , Tissue Distribution
5.
J Clin Invest ; 127(11): 4059-4074, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28972537

ABSTRACT

Overconsumption of high-fat diet (HFD) and sugar-sweetened beverages are risk factors for developing obesity, insulin resistance, and fatty liver disease. Here we have dissected mechanisms underlying this association using mice fed either chow or HFD with or without fructose- or glucose-supplemented water. In chow-fed mice, there was no major physiological difference between fructose and glucose supplementation. On the other hand, mice on HFD supplemented with fructose developed more pronounced obesity, glucose intolerance, and hepatomegaly as compared to glucose-supplemented HFD mice, despite similar caloric intake. Fructose and glucose supplementation also had distinct effects on expression of the lipogenic transcription factors ChREBP and SREBP1c. While both sugars increased ChREBP-ß, fructose supplementation uniquely increased SREBP1c and downstream fatty acid synthesis genes, resulting in reduced liver insulin signaling. In contrast, glucose enhanced total ChREBP expression and triglyceride synthesis but was associated with improved hepatic insulin signaling. Metabolomic and RNA sequence analysis confirmed dichotomous effects of fructose and glucose supplementation on liver metabolism in spite of inducing similar hepatic lipid accumulation. Ketohexokinase, the first enzyme of fructose metabolism, was increased in fructose-fed mice and in obese humans with steatohepatitis. Knockdown of ketohexokinase in liver improved hepatic steatosis and glucose tolerance in fructose-supplemented mice. Thus, fructose is a component of dietary sugar that is distinctively associated with poor metabolic outcomes, whereas increased glucose intake may be protective.


Subject(s)
Fructose/pharmacology , Glucose/pharmacology , Insulin/physiology , Lipogenesis/drug effects , Liver/drug effects , Adolescent , Animals , Diet, High-Fat/adverse effects , Enzyme Induction , Fatty Acids/biosynthesis , Fructokinases/genetics , Fructokinases/metabolism , Glucose Intolerance , Humans , Insulin Resistance , Liver/enzymology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/etiology , Obesity/enzymology , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Transcriptome , Up-Regulation
6.
J Lipid Res ; 57(9): 1696-711, 2016 09.
Article in English | MEDLINE | ID: mdl-27474222

ABSTRACT

We aimed to characterize the lipidomic, metabolomic, and transcriptomic profiles in preterm piglets administered enteral (ENT) formula or three parenteral lipid emulsions [parenteral nutrition (PN)], Intralipid (IL), Omegaven (OV), or SMOFlipid (SL), for 14 days. Piglets in all parenteral lipid groups showed differential organ growth versus ENT piglets; whole body growth rate was lowest in IL piglets, yet there were no differences in either energy expenditure or (13)C-palmitate oxidation. Plasma homeostatic model assessment of insulin resistance demonstrated insulin resistance in IL, but not OV or SL, compared with ENT. The fatty acid and acyl-CoA content of the liver, muscle, brain, and plasma fatty acids reflected the composition of the dietary lipids administered. Free carnitine and acylcarnitine (ACT) levels were markedly reduced in the PN groups compared with ENT piglets. Genes associated with oxidative stress and inflammation were increased, whereas those associated with alternative pathways of fatty acid oxidation were decreased in all PN groups. Our results show that new generation lipid emulsions directly enrich tissue fatty acids, especially in the brain, and lead to improved growth and insulin sensitivity compared with a soybean lipid emulsion. In all total PN groups, carnitine levels are limiting to the formation of ACTs and gene expression reflects the stress of excess lipid on liver function.


Subject(s)
Fish Oils/administration & dosage , Lipid Metabolism/genetics , Phospholipids/administration & dosage , Soybean Oil/administration & dosage , Triglycerides/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Dietary Fats, Unsaturated/administration & dosage , Emulsions/administration & dosage , Fatty Acids/metabolism , Liver/drug effects , Liver/metabolism , Male , Palmitic Acid/administration & dosage , Palmitic Acid/metabolism , Parenteral Nutrition , Swine/growth & development , Swine/metabolism
7.
J Clin Endocrinol Metab ; 99(6): 2128-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24606092

ABSTRACT

OBJECTIVE: Malnutrition is a major cause of childhood morbidity and mortality. To identify and target those at highest risk, there is a critical need to characterize biomarkers that predict complications prior to and during treatment. METHODS: We used targeted and nontargeted metabolomic analysis to characterize changes in a broad array of hormones, cytokines, growth factors, and metabolites during treatment of severe childhood malnutrition. Children aged 6 months to 5 years were studied at presentation to Mulago Hospital and during inpatient therapy with milk-based formulas and outpatient supplementation with ready-to-use food. We assessed the relationship between baseline hormone and metabolite levels and subsequent mortality. RESULTS: Seventy-seven patients were enrolled in the study; a subset was followed up from inpatient treatment to the outpatient clinic. Inpatient and outpatient therapies increased weight/height z scores and induced striking changes in the levels of fatty acids, amino acids, acylcarnitines, inflammatory cytokines, and various hormones including leptin, insulin, GH, ghrelin, cortisol, IGF-I, glucagon-like peptide-1, and peptide YY. A total of 12.2% of the patients died during hospitalization; the major biochemical factor predicting mortality was a low level of leptin (P = .0002), a marker of adipose tissue reserve and a critical modulator of immune function. CONCLUSIONS: We have used metabolomic analysis to provide a comprehensive hormonal and metabolic profile of severely malnourished children at presentation and during nutritional rehabilitation. Our findings suggest that fatty acid metabolism plays a central role in the adaptation to acute malnutrition and that low levels of the adipose tissue hormone leptin associate with, and may predict, mortality prior to and during treatment.


Subject(s)
Child Mortality , Child Nutrition Disorders , Hormones/blood , Malnutrition , Nutrition Therapy , Acute Disease , Child Nutrition Disorders/diagnosis , Child Nutrition Disorders/metabolism , Child Nutrition Disorders/mortality , Child Nutrition Disorders/therapy , Child, Preschool , Cohort Studies , Health Status , Humans , Infant , Malnutrition/diagnosis , Malnutrition/metabolism , Malnutrition/mortality , Malnutrition/therapy , Prognosis , Severity of Illness Index , Treatment Outcome , Uganda/epidemiology
8.
Am J Physiol Endocrinol Metab ; 304(4): E405-13, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23249694

ABSTRACT

Recently, we have described a strong association of branched-chain amino acids (BCAA) and aromatic amino acids (AAA) with obesity and insulin resistance. In the current study, we have investigated the potential impact of BCAA on behavioral functions. We demonstrate that supplementation of either a high-sucrose or a high-fat diet with BCAA induces anxiety-like behavior in rats compared with control groups fed on unsupplemented diets. These behavioral changes are associated with a significant decrease in the concentration of tryptophan (Trp) in brain tissues and a consequent decrease in serotonin but no difference in indices of serotonin synaptic function. The anxiety-like behaviors and decreased levels of Trp in the brain of BCAA-fed rats were reversed by supplementation of Trp in the drinking water but not by administration of fluoxetine, a selective serotonin reuptake inhibitor, suggesting that the behavioral changes are independent of the serotonergic pathway of Trp metabolism. Instead, BCAA supplementation lowers the brain levels of another Trp-derived metabolite, kynurenic acid, and these levels are normalized by Trp supplementation. We conclude that supplementation of high-energy diets with BCAA causes neurobehavioral impairment. Since BCAA are elevated spontaneously in human obesity, our studies suggest a potential mechanism for explaining the strong association of obesity and mood disorders.


Subject(s)
Amino Acids, Branched-Chain/adverse effects , Anxiety/etiology , Brain/metabolism , Diet/adverse effects , Neurons/metabolism , Amino Acids, Branched-Chain/blood , Animals , Anxiety/metabolism , Anxiety/physiopathology , Anxiety/prevention & control , Behavior, Animal , Brain/physiopathology , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Exploratory Behavior , Kynurenic Acid/metabolism , Male , Mood Disorders/etiology , Obesity/etiology , Obesity/psychology , Rats , Rats, Wistar , Serotonin/metabolism , Tryptophan/metabolism , Tryptophan/therapeutic use , Weight Gain
9.
Cell Metab ; 15(5): 606-14, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22560213

ABSTRACT

Fatty acids (FA) and FA-derived metabolites have long been implicated in the development of insulin resistance and type 2 diabetes. Surprisingly, application of metabolomics technologies has revealed that branched-chain amino acids (BCAA) and related metabolites are more strongly associated with insulin resistance than many common lipid species. Moreover, the BCAA-related signature is predictive of incident diabetes and intervention outcomes and uniquely responsive to therapeutic interventions. Nevertheless, in animal feeding studies, BCAA supplementation requires the background of a high-fat diet to promote insulin resistance. This Perspective develops a model to explain how lipids and BCAA may synergize to promote metabolic diseases.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Insulin Resistance/physiology , Lipid Metabolism/physiology , Animals , Humans , Metabolic Diseases/metabolism
10.
Cell Metab ; 9(4): 311-26, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19356713

ABSTRACT

Metabolomic profiling of obese versus lean humans reveals a branched-chain amino acid (BCAA)-related metabolite signature that is suggestive of increased catabolism of BCAA and correlated with insulin resistance. To test its impact on metabolic homeostasis, we fed rats on high-fat (HF), HF with supplemented BCAA (HF/BCAA), or standard chow (SC) diets. Despite having reduced food intake and a low rate of weight gain equivalent to the SC group, HF/BCAA rats were as insulin resistant as HF rats. Pair-feeding of HF diet to match the HF/BCAA animals or BCAA addition to SC diet did not cause insulin resistance. Insulin resistance induced by HF/BCAA feeding was accompanied by chronic phosphorylation of mTOR, JNK, and IRS1Ser307 and by accumulation of multiple acylcarnitines in muscle, and it was reversed by the mTOR inhibitor, rapamycin. Our findings show that in the context of a dietary pattern that includes high fat consumption, BCAA contributes to development of obesity-associated insulin resistance.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Insulin Resistance/physiology , Metabolomics , Obesity/metabolism , Thinness/metabolism , Animals , Cytokines/metabolism , Demography , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Dietary Supplements , Feeding Behavior/drug effects , Female , Hormones/metabolism , Humans , Insulin/metabolism , Male , Mass Spectrometry , Metabolome , Middle Aged , Rats , Rats, Wistar , Signal Transduction/drug effects
11.
J Biol Chem ; 278(29): 26597-603, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12734177

ABSTRACT

Peroxisome proliferator-activated receptor gamma coactivator (PGC)-1alpha is a coactivator of nuclear receptors and other transcription factors that regulates several components of energy metabolism, particularly certain aspects of adaptive thermogenesis in brown fat and skeletal muscle, hepatic gluconeogenesis, and fiber type switching in skeletal muscle. PGC-1alpha has been shown to induce mitochondrial biogenesis when expressed in muscle cells, and preliminary analysis has suggested that this molecule may specifically increase the fraction of uncoupled versus coupled respiration. In this paper, we have performed detailed bioenergetic analyses of the function of PGC-1alpha and its homolog PGC-1beta in muscle cells by monitoring simultaneously oxygen consumption and membrane potential. Cells expressing PGC-1alpha or PGC-1beta display higher proton leak rates at any given membrane potential than control cells. However, cells expressing PGC-1alpha have a higher proportion of their mitochondrial respiration linked to proton leak than cells expressing PGC-1beta. Although these two proteins cause a similar increase in the expression of many mitochondrial genes, PGC-1beta preferentially induces certain genes involved in the removal of reactive oxygen species, recently recognized as activators of uncoupling proteins. Together, these data indicate that PGC-1alpha and PGC-1beta profoundly alter mitochondrial metabolism and suggest that these proteins are likely to play different physiological functions.


Subject(s)
Muscle, Skeletal/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Energy Metabolism , Green Fluorescent Proteins , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potentials , Mice , Mice, Transgenic , Microscopy, Electron , Mitochondria, Muscle/metabolism , Muscle, Skeletal/cytology , Oxygen Consumption , Protons , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL