201
|
Piccolo BD, Graham JL, Stanhope KL, Fiehn O, Havel PJ, Adams SH. Plasma amino acid and metabolite signatures tracking diabetes progression in the UCD-T2DM rat model. Am J Physiol Endocrinol Metab 2016; 310:E958-69. [PMID: 27094034 PMCID: PMC4935135 DOI: 10.1152/ajpendo.00052.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/15/2016] [Indexed: 12/16/2022]
Abstract
Elevations of plasma concentrations of branched-chain amino acids (BCAAs) are observed in human insulin resistance and type 2 diabetes mellitus (T2DM); however, there has been some controversy with respect to the passive or causative nature of the BCAA phenotype. Using untargeted metabolomics, plasma BCAA and other metabolites were assessed in lean control Sprague-Dawley rats (LC) and temporally during diabetes development in the UCD-T2DM rat model, i.e., prediabetic (PD) and 2 wk (D2W), 3 mo (D3M), and 6 mo (D6M) post-onset of diabetes. Plasma leucine, isoleucine, and valine concentrations were elevated only in D6M rats compared with D2W rats (by 28, 29, and 30%, respectively). This was in contrast to decreased plasma concentrations of several other amino acids in D3M and/or D6M relative to LC rats (Ala, Arg, Glu, Gln, Met, Ser, Thr, and Trp). BCAAs were positively correlated with fasting glucose and negatively correlated with plasma insulin, total body weight, total adipose tissue weight, and gastrocnemius muscle weight in the D3M and D6M groups. Multivariate analysis revealed that D3M and D6M UCD-T2DM rats had lower concentrations of amino acids, amino acid derivatives, 1,5-anhydroglucitol, and conduritol-β-opoxide and higher concentrations of uronic acids, pantothenic acids, aconitate, benzoic acid, lactate, and monopalmitin-2-glyceride relative to PD and D2W UCD-T2DM rats. The UCD-T2DM rat does not display elevated plasma BCAA concentrations until 6 mo post-onset of diabetes. With the acknowledgement that this is a rodent model of T2DM, the results indicate that elevated plasma BCAA concentrations are not necessary or sufficient to elicit an insulin resistance or T2DM onset.
Collapse
Affiliation(s)
- Brian D Piccolo
- Arkansas Children's Nutrition Center, Little Rock, Arkansas; Department of Pediatrics, University of Arkansas for Medical Science, Little Rock, Arkansas
| | - James L Graham
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California; Department of Nutrition, University of California, Davis, California
| | - Kimber L Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California; Department of Nutrition, University of California, Davis, California
| | - Oliver Fiehn
- West Coast Metabolomics Center, Genome Center, University of California, Davis, California; and King Abdulaziz University, Biochemistry Department, Jeddah, Saudi Arabia
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California; Department of Nutrition, University of California, Davis, California
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, Arkansas; Department of Pediatrics, University of Arkansas for Medical Science, Little Rock, Arkansas; Department of Nutrition, University of California, Davis, California;
| |
Collapse
|
202
|
Allam-Ndoul B, Guénard F, Garneau V, Cormier H, Barbier O, Pérusse L, Vohl MC. Association between Metabolite Profiles, Metabolic Syndrome and Obesity Status. Nutrients 2016; 8:nu8060324. [PMID: 27240400 PMCID: PMC4924165 DOI: 10.3390/nu8060324] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/11/2016] [Accepted: 05/20/2016] [Indexed: 12/28/2022] Open
Abstract
Underlying mechanisms associated with the development of abnormal metabolic phenotypes among obese individuals are not yet clear. Our aim is to investigate differences in plasma metabolomics profiles between normal weight (NW) and overweight/obese (Ov/Ob) individuals, with or without metabolic syndrome (MetS). Mass spectrometry-based metabolite profiling was used to compare metabolite levels between each group. Three main principal components factors explaining a maximum of variance were retained. Factor 1's (long chain glycerophospholipids) metabolite profile score was higher among Ov/Ob with MetS than among Ov/Ob and NW participants without MetS. This factor was positively correlated to plasma total cholesterol (total-C) and triglyceride levels in the three groups, to high density lipoprotein -cholesterol (HDL-C) among participants without MetS. Factor 2 (amino acids and short to long chain acylcarnitine) was positively correlated to HDL-C and negatively correlated with insulin levels among NW participants. Factor 3's (medium chain acylcarnitines) metabolite profile scores were higher among NW participants than among Ov/Ob with or without MetS. Factor 3 was negatively associated with glucose levels among the Ov/Ob with MetS. Factor 1 seems to be associated with a deteriorated metabolic profile that corresponds to obesity, whereas Factors 2 and 3 seem to be rather associated with a healthy metabolic profile.
Collapse
Affiliation(s)
- Bénédicte Allam-Ndoul
- Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec City, QC G1V0A6, Canada.
- School of Nutrition, Laval University, Quebec City, QC G1V0A6, Canada.
| | - Frédéric Guénard
- Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec City, QC G1V0A6, Canada.
- School of Nutrition, Laval University, Quebec City, QC G1V0A6, Canada.
| | - Véronique Garneau
- Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec City, QC G1V0A6, Canada.
- School of Nutrition, Laval University, Quebec City, QC G1V0A6, Canada.
| | - Hubert Cormier
- Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec City, QC G1V0A6, Canada.
- School of Nutrition, Laval University, Quebec City, QC G1V0A6, Canada.
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, CHU-Quebec Research Center, and Faculty of Pharmacy, Laval University, Quebec City, QC G1V4G2, Canada.
| | - Louis Pérusse
- Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec City, QC G1V0A6, Canada.
- Department of Kinesiology, Laval University, Quebec City, QC G1V0A6, Canada.
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec City, QC G1V0A6, Canada.
- School of Nutrition, Laval University, Quebec City, QC G1V0A6, Canada.
| |
Collapse
|
203
|
Gao X, Zhang W, Wang Y, Pedram P, Cahill F, Zhai G, Randell E, Gulliver W, Sun G. Serum metabolic biomarkers distinguish metabolically healthy peripherally obese from unhealthy centrally obese individuals. Nutr Metab (Lond) 2016; 13:33. [PMID: 27175209 PMCID: PMC4865032 DOI: 10.1186/s12986-016-0095-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/03/2016] [Indexed: 12/11/2022] Open
Abstract
Background Metabolic abnormalities are more associated with central obesity than peripheral obesity, but the underlying mechanisms are largely unknown. The present study was to identify serum metabolic biomarkers which distinguish metabolically unhealthy centrally obese (MUCO) from metabolically healthy peripherally obese (MHPO) individuals. Methods A two-stage case–control study design was employed. In the discovery stage, 20 individuals (10 MHPO and 10 MUCO) were included and in the following validation stage, 79 individuals (20 normal weight (NW), 30 MHPO, 29 MUCO) were utilized. Study groups were matched for age, sex, physical activity and total dietary calorie intake with MHPO and MUCO additionally matched for BMI. Metabolic abnormality was defined as: 1) HOMA-IR > 4.27 (90th percentile), 2) high-density lipoprotein cholesterol < 1.03 mmol/L in men and < 1.30 mmol/L in women, 3) fasting blood glucose ≥ 5.6 mmol/L, and 4) waist circumference > 102 cm in men and > 88 cm in women. MUCO individuals had all of these abnormalities whereas MHPO and NW individuals had none of them. A targeted metabolomics approach was performed on fasting serum samples, which can simultaneously identify and quantify 186 metabolites. Results In the discovery stage, serum leucine, isoleucine, tyrosine, valine, phenylalanine, alpha-aminoadipic acid, methioninesulfoxide and propionylcarnitine were found to be significantly higher in MUCO, compared with MHPO group after multiple testing adjustment. Significant changes of five metabolites (leucine, isoleucine, valine, alpha-aminoadipic acid, propionylcarnitine) were confirmed in the validation stage. Conclusions Significantly higher levels of serum leucine, isoleucine, valine, alpha-aminoadipic acid, propionylcarnitine are characteristic of metabolically unhealthy centrally obese patients. The finding provides novel insights into the pathogenesis of metabolic abnormalities in obesity. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0095-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiang Gao
- College of Food Science and Engineering, Ocean University of China, No.5, Yu Shan Road, Qingdao, Shandong Province China ; Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada
| | - Weidong Zhang
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada
| | - Yongbo Wang
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada ; Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000 Liaoning China
| | - Pardis Pedram
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada
| | - Farrell Cahill
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada
| | - Guangju Zhai
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada
| | - Edward Randell
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada
| | - Wayne Gulliver
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada
| | - Guang Sun
- Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL Canada
| |
Collapse
|
204
|
Mastrangelo A, Martos-Moreno GÁ, García A, Barrios V, Rupérez FJ, Chowen JA, Barbas C, Argente J. Insulin resistance in prepubertal obese children correlates with sex-dependent early onset metabolomic alterations. Int J Obes (Lond) 2016; 40:1494-1502. [PMID: 27163744 PMCID: PMC5056960 DOI: 10.1038/ijo.2016.92] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/21/2016] [Accepted: 05/02/2016] [Indexed: 12/16/2022]
Abstract
Background: Insulin resistance (IR) is usually the first metabolic alteration diagnosed in obese children and the key risk factor for development of comorbidities. The factors determining whether or not IR develops as a result of excess body mass index (BMI) are still not completely understood. Objectives: This study aimed to elucidate the mechanisms underpinning the predisposition toward hyperinsulinemia-related complications in obese children by using a metabolomic strategy that allows a profound interpretation of metabolic profiles potentially affected by IR. Methods: Serum from 60 prepubertal obese children (30 girls/30 boys, 50% IR and 50% non-IR in each group, but with similar BMIs) were analyzed by using liquid chromatography–mass spectrometry, gas chromatography–mass spectrometry and capillary electrophoresis–mass spectrometry following an untargeted metabolomics approach. Validation was then performed on a group of 100 additional children with the same characteristics. Results: When obese children with and without IR were compared, 47 metabolites out of 818 compounds (P<0.05) obtained after data pre-processing were found to be significantly different. Bile acids exhibit the greatest changes (that is, approximately a 90% increase in IR). The majority of metabolites differing between groups were lysophospholipids (15) and amino acids (17), indicating inflammation and central carbon metabolism as the most altered processes in impaired insulin signaling. Multivariate analysis (OPLS-DA models) showed subtle differences between groups that were magnified when females were analyzed alone. Conclusions: Inflammation and central carbon metabolism, together with the contribution of the gut microbiota, are the most altered processes in obese children with impaired insulin signaling in a sex-specific fashion despite their prepubertal status.
Collapse
Affiliation(s)
- A Mastrangelo
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, San Pablo CEU University, Madrid, Spain
| | - G Á Martos-Moreno
- Department of Pediatrics & Pediatric Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain.,CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - A García
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, San Pablo CEU University, Madrid, Spain
| | - V Barrios
- Department of Pediatrics & Pediatric Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain.,CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - F J Rupérez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, San Pablo CEU University, Madrid, Spain
| | - J A Chowen
- Department of Pediatrics & Pediatric Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain.,CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - C Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, San Pablo CEU University, Madrid, Spain
| | - J Argente
- Department of Pediatrics & Pediatric Endocrinology, Instituto de Investigación La Princesa, Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain.,CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
205
|
Guasch-Ferré M, Hruby A, Toledo E, Clish CB, Martínez-González MA, Salas-Salvadó J, Hu FB. Metabolomics in Prediabetes and Diabetes: A Systematic Review and Meta-analysis. Diabetes Care 2016; 39:833-846. [PMID: 27208380 PMCID: PMC4839172 DOI: 10.2337/dc15-2251] [Citation(s) in RCA: 664] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/06/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To conduct a systematic review of cross-sectional and prospective human studies evaluating metabolite markers identified using high-throughput metabolomics techniques on prediabetes and type 2 diabetes. RESEARCH DESIGN AND METHODS We searched MEDLINE and EMBASE databases through August 2015. We conducted a qualitative review of cross-sectional and prospective studies. Additionally, meta-analyses of metabolite markers, with data estimates from at least three prospective studies, and type 2 diabetes risk were conducted, and multivariable-adjusted relative risks of type 2 diabetes were calculated per study-specific SD difference in a given metabolite. RESULTS We identified 27 cross-sectional and 19 prospective publications reporting associations of metabolites and prediabetes and/or type 2 diabetes. Carbohydrate (glucose and fructose), lipid (phospholipids, sphingomyelins, and triglycerides), and amino acid (branched-chain amino acids, aromatic amino acids, glycine, and glutamine) metabolites were higher in individuals with type 2 diabetes compared with control subjects. Prospective studies provided evidence that blood concentrations of several metabolites, including hexoses, branched-chain amino acids, aromatic amino acids, phospholipids, and triglycerides, were associated with the incidence of prediabetes and type 2 diabetes. We meta-analyzed results from eight prospective studies that reported risk estimates for metabolites and type 2 diabetes, including 8,000 individuals of whom 1,940 had type 2 diabetes. We found 36% higher risk of type 2 diabetes per study-specific SD difference for isoleucine (pooled relative risk 1.36 [1.24-1.48]; I(2) = 9.5%), 36% for leucine (1.36 [1.17-1.58]; I(2) = 37.4%), 35% for valine (1.35 [1.19-1.53]; I(2) = 45.8%), 36% for tyrosine (1.36 [1.19-1.55]; I(2) = 51.6%), and 26% for phenylalanine (1.26 [1.10-1.44]; I(2) = 56%). Glycine and glutamine were inversely associated with type 2 diabetes risk (0.89 [0.81-0.96] and 0.85 [0.82-0.89], respectively; both I(2) = 0.0%). CONCLUSIONS In studies using high-throughput metabolomics, several blood amino acids appear to be consistently associated with the risk of developing type 2 diabetes.
Collapse
Affiliation(s)
- Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Institute for Health Research, Rovira i Virgili University, Reus, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Adela Hruby
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Estefanía Toledo
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain Department of Preventive Medicine and Public Health, University of Navarra, Health Research Institute of Navarra, Pamplona, Spain
| | | | - Miguel A Martínez-González
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain Department of Preventive Medicine and Public Health, University of Navarra, Health Research Institute of Navarra, Pamplona, Spain
| | - Jordi Salas-Salvadó
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Institute for Health Research, Rovira i Virgili University, Reus, Spain CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
206
|
White PJ, Lapworth AL, An J, Wang L, McGarrah RW, Stevens RD, Ilkayeva O, George T, Muehlbauer MJ, Bain JR, Trimmer JK, Brosnan MJ, Rolph TP, Newgard CB. Branched-chain amino acid restriction in Zucker-fatty rats improves muscle insulin sensitivity by enhancing efficiency of fatty acid oxidation and acyl-glycine export. Mol Metab 2016; 5:538-551. [PMID: 27408778 PMCID: PMC4921791 DOI: 10.1016/j.molmet.2016.04.006] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 04/15/2016] [Accepted: 04/19/2016] [Indexed: 12/19/2022] Open
Abstract
Objective A branched-chain amino acid (BCAA)-related metabolic signature is strongly associated with insulin resistance and predictive of incident diabetes and intervention outcomes. To better understand the role that this metabolite cluster plays in obesity-related metabolic dysfunction, we studied the impact of BCAA restriction in a rodent model of obesity in which BCAA metabolism is perturbed in ways that mirror the human condition. Methods Zucker-lean rats (ZLR) and Zucker-fatty rats (ZFR) were fed either a custom control, low fat (LF) diet, or an isonitrogenous, isocaloric LF diet in which all three BCAA (Leu, Ile, Val) were reduced by 45% (LF-RES). We performed comprehensive metabolic and physiologic profiling to characterize the effects of BCAA restriction on energy balance, insulin sensitivity, and glucose, lipid and amino acid metabolism. Results LF-fed ZFR had higher levels of circulating BCAA and lower levels of glycine compared to LF-fed ZLR. Feeding ZFR with the LF-RES diet lowered circulating BCAA to levels found in LF-fed ZLR. Activity of the rate limiting enzyme in the BCAA catabolic pathway, branched chain keto acid dehydrogenase (BCKDH), was lower in liver but higher in skeletal muscle of ZFR compared to ZLR and was not responsive to diet in either tissue. BCAA restriction had very little impact on metabolites studied in liver of ZFR where BCAA content was low, and BCKDH activity was suppressed. However, in skeletal muscle of LF-fed ZFR compared to LF-fed ZLR, where BCAA content and BCKDH activity were increased, accumulation of fatty acyl CoAs was completely normalized by dietary BCAA restriction. BCAA restriction also normalized skeletal muscle glycine content and increased urinary acetyl glycine excretion in ZFR. These effects were accompanied by lower RER and improved skeletal muscle insulin sensitivity in LF-RES fed ZFR as measured by hyperinsulinemic-isoglycemic clamp. Conclusions Our data are consistent with a model wherein elevated circulating BCAA contribute to development of obesity-related insulin resistance by interfering with lipid oxidation in skeletal muscle. BCAA-dependent lowering of the skeletal muscle glycine pool appears to contribute to this effect by slowing acyl-glycine export to the urine. Feeding a BCAA restricted diet improves skeletal muscle insulin sensitivity in Zucker fatty rats. BCKDH activity is decreased in liver and increased in skeletal muscle in Zucker fatty versus lean rats. High BCAA levels drive the obesity-associated decline in circulating and muscle glycine levels. BCAA-driven glycine depletion restricts formation of acyl-glycine adducts for excretion in urine. High BCAA/low glycine reduces efficiency of fat oxidation in muscle leading to acyl CoA buildup.
Collapse
Affiliation(s)
- Phillip J White
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | | | - Jie An
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - Liping Wang
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - Robert W McGarrah
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - Robert D Stevens
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - Tabitha George
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA
| | - Jeff K Trimmer
- CV and Metabolic Diseases Research Unit, Pfizer, Cambridge, MA, USA
| | - M Julia Brosnan
- CV and Metabolic Diseases Research Unit, Pfizer, Cambridge, MA, USA
| | - Timothy P Rolph
- CV and Metabolic Diseases Research Unit, Pfizer, Cambridge, MA, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC, 27701, USA.
| |
Collapse
|
207
|
Mangge H, Zelzer S, Prüller F, Schnedl WJ, Weghuber D, Enko D, Bergsten P, Haybaeck J, Meinitzer A. Branched-chain amino acids are associated with cardiometabolic risk profiles found already in lean, overweight and obese young. J Nutr Biochem 2016; 32:123-7. [PMID: 27142745 DOI: 10.1016/j.jnutbio.2016.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/09/2016] [Accepted: 02/23/2016] [Indexed: 01/01/2023]
Abstract
Cardiovascular risk is increased in obese subjects. Nevertheless, some overweight and obese remain cardiometabolically healthy (CMH), and normal-weight persons develop cardiovascular disease (CVD). Herein, we investigate the potential of branched-chain amino acids (BCAAs) to identify an increased CVD risk in a cross-sectional study of 666 adults and juveniles (age 25.3±12.8years), classified as lean, overweight or obese. Cardiometabolic groups were defined by cutoffs of systolic blood pressure<130mmHg, diastolic blood pressure<85mmHg, glucose<125mg/dl, triglycerides<150mg/dl, HDL-cholesterol>40mg/dl (males), HDL-cholesterol>50mg/dl (females) and HOMA-IR<5. CMH had ≤1 cutoff, and cardiometabolically abnormal (CMA) had ≥2 cutoffs. Amino acids were measured by high-pressure lipid chromatography after precipitation of serum with perchloric acid and derivatization with o-phthalaldehyde. Valine correlated with 5, leucine correlated with 3 and isoleucine correlated with 5 of the cardiac risk classification factors. Valine and leucine were significantly higher in the obese (P<.001, P=.015, respectively), overweight (P<.001, P=.015, respectively) and lean (P=.024, P=.012, respectively) CMA compared to CMH subjects. Isoleucine showed except of the lean group the same results. Taken together, BCAAs, especially valine and leucine, are proposed as a cardiometabolic risk marker independent of body mass index (BMI) category.
Collapse
Affiliation(s)
- Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Florian Prüller
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | | | - Daniel Weghuber
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria; Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
| | - Dietmar Enko
- Institute of Laboratory Medicine, General Hospital Steyr, Steyr, Austria
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
208
|
Geidenstam N, Danielsson APH, Spégel P, Ridderstråle M. Changes in glucose-elicited blood metabolite responses following weight loss and long term weight maintenance in obese individuals with impaired glucose tolerance. Diabetes Res Clin Pract 2016; 113:187-97. [PMID: 26809903 DOI: 10.1016/j.diabres.2015.12.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/18/2015] [Accepted: 12/24/2015] [Indexed: 01/05/2023]
Abstract
AIMS Weight loss improves insulin sensitivity and glucose tolerance in obese subjects with impaired glucose tolerance (IGT), but the long term dynamic effects on blood metabolites other than glucose during an oral glucose tolerance test (OGTT), are largely unknown. Here, we studied changes in OGTT-elicited metabolite patterns in obese subjects during a diet-induced weight loss study. METHODS Blood samples from 14 obese individuals with IGT were collected at 0, 30 and 120 min during a standard 75 g OGTT at baseline (BMI 44 ± 2 kg/m(2)), after weight loss (BMI 36 ± 2 kg/m(2)) and after weight maintenance (BMI 35 ± 2 kg/m(2)). Serum metabolite levels were analyzed by gas chromatography/mass spectrometry and compared to a lean glucose tolerant group. RESULTS Changes in the OGTT-elicited metabolite patterns occurred differentially during weight loss and weight maintenance. Enhanced suppression of aromatic amino acids were associated with decreased insulinogenic index observed after weight loss (tyrosine: r=0.72, p=0.013; phenylalanine: r=0.63, p=0.039). The OGTT-elicited suppression and/or lack of increase in levels of glutamate, glutamine, isoleucine, leucine, and the fatty acids laurate, oleate and palmitate, improved towards the lean profile after weight maintenance, paralleling an improvement in glucose tolerance. The greater heterogeneity in the response before and after weight loss in the obese, compared to lean subjects, was markedly reduced after weight maintenance. CONCLUSIONS Diet-induced weight loss followed by weight maintenance results in changes in metabolite profiles associated with either hepatic insulin sensitivity or peripheral glucose tolerance. Our results highlight the importance of evaluating the effects of weight loss and weight maintenance separately.
Collapse
Affiliation(s)
- Nina Geidenstam
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Sweden.
| | - Anders P H Danielsson
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Sweden
| | - Peter Spégel
- Department of Clinical Sciences Malmö, Unit of Molecular Metabolism, Lund University Diabetes Center, Lund University, Sweden
| | - Martin Ridderstråle
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Sweden; Steno Diabetes Center A/S, Gentofte, Denmark
| |
Collapse
|
209
|
Associations between plasma branched-chain amino acids, β-aminoisobutyric acid and body composition. J Nutr Sci 2016; 5:e6. [PMID: 27313851 PMCID: PMC4791517 DOI: 10.1017/jns.2015.37] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 11/04/2015] [Accepted: 11/18/2015] [Indexed: 01/01/2023] Open
Abstract
Plasma branched-chain amino acids (BCAA) are elevated in obesity and associated with
increased cardiometabolic risk. β-Aminoisobutyric acid (B-AIBA), a recently identified
small molecule metabolite, is associated with decreased cardiometabolic risk. Therefore,
we investigated the association of BCAA and B-AIBA with each other and with detailed body
composition parameters, including abdominal visceral adipose tissue (VAT) and subcutaneous
adipose tissue (SAT). A cross-sectional study was carried out with lean
(n 15) and obese (n 33) men and women. Detailed
metabolic evaluations, including measures of body composition, insulin sensitivity and
plasma metabolomics were completed. Plasma BCAA were higher (1·6 (se 0·08)
(×107) v. 1·3 (se 0·06) (×107) arbitrary
units; P = 0·005) in obese v. lean subjects. BCAA were
positively associated with VAT (R 0·49; P = 0·0006) and
trended to an association with SAT (R 0·29; P = 0·052).
The association between BCAA and VAT, but not SAT, remained significant after controlling
for age, sex and race on multivariate modelling (P < 0·05). BCAA
were also associated with parameters of insulin sensitivity (Matsuda index:
R −0·50, P = 0·0004; glucose AUC: R
0·53, P < 0·001). BCAA were not associated with B-AIBA
(R −0·04; P = 0·79). B-AIBA was negatively associated
with SAT (R −0·37; P = 0·01) but only trended to an
association with VAT (R 0·27; P = 0·07). However,
neither relationship remained significant after multivariate modelling
(P > 0·05). Plasma B-AIBA was associated with parameters of
insulin sensitivity (Matsuda index R 0·36, P = 0·01;
glucose AUC: R −0·30, P = 0·04). Plasma BCAA levels were
positively correlated with VAT and markers of insulin resistance. The results suggest a
possible complex role of adipose tissue in BCAA homeostasis and insulin resistance.
Collapse
Key Words
- AU, arbitrary units
- B-AIBA, β-aminoisobutyric acid
- BCAA, branched-chain amino acid
- BCAT, branched-chain amino acid aminotransferase
- BCKD, branched-chain α-ketoacid dehydrogenase
- Branched-chain amino acids
- DXA, dual-energy X-ray absorptiometry
- HOMA-IR, homeostasis model assessment for insulin resistance
- Lean body mass
- Metabolomics
- OGTT, oral glucose tolerance test
- SAT, subcutaneous adipose tissue
- Subcutaneous adipose tissue
- VAT, visceral adipose tissue
- Visceral adiposity
- β-Aminoisobutyric acid
Collapse
|
210
|
Shah SH, Newgard CB. Integrated metabolomics and genomics: systems approaches to biomarkers and mechanisms of cardiovascular disease. ACTA ACUST UNITED AC 2016; 8:410-9. [PMID: 25901039 DOI: 10.1161/circgenetics.114.000223] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The genetic architecture underlying the heritability of cardiovascular disease is incompletely understood. Metabolomics is an emerging technology platform that has shown early success in identifying biomarkers and mechanisms of common chronic diseases. Integration of metabolomics, genetics, and other omics platforms in a systems biology approach holds potential for elucidating novel genetic markers and mechanisms for cardiovascular disease. We review important studies that have used metabolomic profiling in cardiometabolic diseases, approaches for integrating metabolomics with genetics and other molecular profiling platforms, and key studies showing the potential for such studies in deciphering cardiovascular disease genetics, biomarkers, and mechanisms.
Collapse
Affiliation(s)
- Svati H Shah
- From the Duke Molecular Physiology Institute (S.H.S., C.B.N.), Division of Cardiology, Department of Medicine (S.H.S., C.B.N.), Department of Pharmacology and Cancer Biology and Division of Endocrinology, Department of Medicine, and the Sarah W. Stedman Nutrition and Metabolism Center (C.B.N.), Duke University, Durham, NC.
| | - Christopher B Newgard
- From the Duke Molecular Physiology Institute (S.H.S., C.B.N.), Division of Cardiology, Department of Medicine (S.H.S., C.B.N.), Department of Pharmacology and Cancer Biology and Division of Endocrinology, Department of Medicine, and the Sarah W. Stedman Nutrition and Metabolism Center (C.B.N.), Duke University, Durham, NC
| |
Collapse
|
211
|
Zhao X, Han Q, Liu Y, Sun C, Gang X, Wang G. The Relationship between Branched-Chain Amino Acid Related Metabolomic Signature and Insulin Resistance: A Systematic Review. J Diabetes Res 2016; 2016:2794591. [PMID: 27642608 PMCID: PMC5014958 DOI: 10.1155/2016/2794591] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 07/16/2016] [Accepted: 08/01/2016] [Indexed: 01/22/2023] Open
Abstract
Recent studies have shown the positive association between increased circulating BCAAs (valine, leucine, and isoleucine) and insulin resistance (IR) in obese or diabetic patients. However, results seem to be controversial in different races, diets, and distinct tissues. Our aims were to evaluate the relationship between BCAA and IR as well as later diabetes risk and explore the phenotypic and genetic factors influencing BCAA level based on available studies. We performed systematic review, searching MEDLINE, EMASE, ClinicalTrials.gov, the Cochrane Library, and Web of Science from inception to March 2016. After selection, 23 studies including 20,091 participants were included. Based on current evidence, we found that BCAA is a useful biomarker for early detection of IR and later diabetic risk. Factors influencing BCAA level can be divided into four parts: race, gender, dietary patterns, and gene variants. These factors might not only contribute to the elevated BCAA level but also show obvious associations with insulin resistance. Genes related to BCAA catabolism might serve as potential targets for the treatment of IR associated metabolic disorders. Moreover, these factors should be controlled properly during study design and data analysis. In the future, more large-scale studies with elaborate design addressing BCAA and IR are required.
Collapse
Affiliation(s)
- Xue Zhao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Qing Han
- Hospital of Orthopedics, The Second Hospital of Jilin University, Changchun 130021, China
| | - Yujia Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Chenglin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
- *Xiaokun Gang: and
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
- *Guixia Wang:
| |
Collapse
|
212
|
Hellmuth C, Kirchberg FF, Lass N, Harder U, Peissner W, Koletzko B, Reinehr T. Tyrosine Is Associated with Insulin Resistance in Longitudinal Metabolomic Profiling of Obese Children. J Diabetes Res 2016; 2016:2108909. [PMID: 26881241 PMCID: PMC4736430 DOI: 10.1155/2016/2108909] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/28/2015] [Accepted: 09/06/2015] [Indexed: 12/18/2022] Open
Abstract
In obese children, hyperinsulinaemia induces adverse metabolic consequences related to the risk of cardiovascular and other disorders. Branched-chain amino acids (BCAA) and acylcarnitines (Carn), involved in amino acid (AA) degradation, were linked to obesity-associated insulin resistance, but these associations yet have not been studied longitudinally in obese children. We studied 80 obese children before and after a one-year lifestyle intervention programme inducing substantial weight loss >0.5 BMI standard deviation scores in 40 children and no weight loss in another 40 children. At baseline and after the 1-year intervention, we assessed insulin resistance (HOMA index), fasting glucose, HbA1c, 2 h glucose in an oral glucose tolerance test, AA, and Carn. BMI adjusted metabolite levels were associated with clinical markers at baseline and after intervention, and changes with the intervention period were evaluated. Only tyrosine was significantly associated with HOMA (p < 0.05) at baseline and end and with change during the intervention (p < 0.05). In contrast, ratios depicting BCAA metabolism were negatively associated with HOMA at baseline (p < 0.05), but not in the longitudinal profiling. Stratified analysis revealed that the children with substantial weight loss drove this association. We conclude that tyrosine alterations in association with insulin resistance precede alteration in BCAA metabolism. This trial is registered with ClinicalTrials.gov Identifier NCT00435734.
Collapse
Affiliation(s)
- Christian Hellmuth
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich, Lindwurmstraße 4, 80337 Munich, Germany
| | - Franca Fabiana Kirchberg
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich, Lindwurmstraße 4, 80337 Munich, Germany
| | - Nina Lass
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents, University of Witten-Herdecke, Dr. Friedrich Steiner Strasse 5, 45711 Datteln, Germany
| | - Ulrike Harder
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich, Lindwurmstraße 4, 80337 Munich, Germany
| | - Wolfgang Peissner
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich, Lindwurmstraße 4, 80337 Munich, Germany
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich, Lindwurmstraße 4, 80337 Munich, Germany
- *Berthold Koletzko:
| | - Thomas Reinehr
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents, University of Witten-Herdecke, Dr. Friedrich Steiner Strasse 5, 45711 Datteln, Germany
| |
Collapse
|
213
|
Hu W, Sun L, Gong Y, Zhou Y, Yang P, Ye Z, Fu J, Huang A, Fu Z, Yu W, Zhao Y, Yang T, Zhou H. Relationship between Branched-Chain Amino Acids, Metabolic Syndrome, and Cardiovascular Risk Profile in a Chinese Population: A Cross-Sectional Study. Int J Endocrinol 2016; 2016:8173905. [PMID: 27528871 PMCID: PMC4977397 DOI: 10.1155/2016/8173905] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/28/2016] [Indexed: 02/07/2023] Open
Abstract
Objective. This study aimed to evaluate the relationship between branched-chain amino acids (BCAAs), metabolic syndrome (MS), and other cardiovascular (CV) risk factors in middle-aged and elderly Chinese population at high risk for the development of cardiovascular disease (CVD). Methods. 1302 subjects were enrolled from the Huai'an Diabetes Prevention Program. Results. BCAAs levels were positively correlated with MS, its components, and CV risk profile. The odds ratio (OR) for MS among subjects in the fourth quartile of BCAAs levels showed a 2.17-fold increase compared with those in the first quartile. BCAAs were independently associated with high Framingham risk score even after adjusting for MS and its components (P < 0.0001). Additionally, the OR for high CV risk was 3.20-fold (P < 0.0001) in participants in the fourth BCAAs quartile with MS compared with participants in the first BCAAs quartile without MS. Conclusions. Increased BCAAs levels are independent risk factors of MS and CVD in addition to the traditional factors in middle-aged and elderly Chinese population. The development of CVD in MS patients with high level BCAAs is accelerated. Intervention studies are needed to investigate whether the strategy of BCAAs reduction has impacts on endpoints in patients with higher CV risk. This study is registered with ChiCTR-TRC-14005029.
Collapse
Affiliation(s)
- Wen Hu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an 223001, China
| | - Luning Sun
- Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yingyun Gong
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ying Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Panpan Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhengqin Ye
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jinxiang Fu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Aijie Huang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhenzhen Fu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Weinan Yu
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an 223001, China
| | - Yang Zhao
- School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- *Hongwen Zhou:
| |
Collapse
|
214
|
Abstract
Metabolomics is a promising approach for the identification of chemical compounds that serve for early detection, diagnosis, prediction of therapeutic response and prognosis of disease. Moreover, metabolomics has shown to increase the diagnostic threshold and prediction of type 2 diabetes. Evidence suggests that branched-chain amino acids, acylcarnitines and aromatic amino acids may play an early role on insulin resistance, exposing defects on amino acid metabolism, β-oxidation, and tricarboxylic acid cycle. This review aims to provide a panoramic view of the metabolic shifts that antecede or follow type 2 diabetes. Key messages BCAAs, AAAs and acylcarnitines are strongly associated with early insulin resistance. Diabetes risk prediction has been improved when adding metabolomic markers of dysglycemia to standard clinical and biochemical factors.
Collapse
Affiliation(s)
| | - Carlos A Aguilar-Salinas
- a Instituto Nacional De Ciencias Médicas Y Nutrición "Salvador Zubirán" , Ciudad De México , D.F
| | - Ivette Cruz-Bautista
- a Instituto Nacional De Ciencias Médicas Y Nutrición "Salvador Zubirán" , Ciudad De México , D.F
| | | |
Collapse
|
215
|
Catabolism of Branched Chain Amino Acids Contributes Significantly to Synthesis of Odd-Chain and Even-Chain Fatty Acids in 3T3-L1 Adipocytes. PLoS One 2015; 10:e0145850. [PMID: 26710334 PMCID: PMC4692509 DOI: 10.1371/journal.pone.0145850] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/09/2015] [Indexed: 12/21/2022] Open
Abstract
The branched chain amino acids (BCAA) valine, leucine and isoleucine have been implicated in a number of diseases including obesity, insulin resistance, and type 2 diabetes mellitus, although the mechanisms are still poorly understood. Adipose tissue plays an important role in BCAA homeostasis by actively metabolizing circulating BCAA. In this work, we have investigated the link between BCAA catabolism and fatty acid synthesis in 3T3-L1 adipocytes using parallel 13C-labeling experiments, mass spectrometry and model-based isotopomer data analysis. Specifically, we performed parallel labeling experiments with four fully 13C-labeled tracers, [U-13C]valine, [U-13C]leucine, [U-13C]isoleucine and [U-13C]glutamine. We measured mass isotopomer distributions of fatty acids and intracellular metabolites by GC-MS and analyzed the data using the isotopomer spectral analysis (ISA) framework. We demonstrate that 3T3-L1 adipocytes accumulate significant amounts of even chain length (C14:0, C16:0 and C18:0) and odd chain length (C15:0 and C17:0) fatty acids under standard cell culture conditions. Using a novel GC-MS method, we demonstrate that propionyl-CoA acts as the primer on fatty acid synthase for the production of odd chain fatty acids. BCAA contributed significantly to the production of all fatty acids. Leucine and isoleucine contributed at least 25% to lipogenic acetyl-CoA pool, and valine and isoleucine contributed 100% to lipogenic propionyl-CoA pool. Our results further suggest that low activity of methylmalonyl-CoA mutase and mass action kinetics of propionyl-CoA on fatty acid synthase result in high rates of odd chain fatty acid synthesis in 3T3-L1 cells. Overall, this work provides important new insights into the connection between BCAA catabolism and fatty acid synthesis in adipocytes and underscores the high capacity of adipocytes for metabolizing BCAA.
Collapse
|
216
|
Gralka E, Luchinat C, Tenori L, Ernst B, Thurnheer M, Schultes B. Metabolomic fingerprint of severe obesity is dynamically affected by bariatric surgery in a procedure-dependent manner. Am J Clin Nutr 2015; 102:1313-22. [PMID: 26581381 DOI: 10.3945/ajcn.115.110536] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity is associated with multiple diseases. Bariatric surgery is the most effective therapy for severe obesity that can reduce body weight and obesity-associated morbidity. The metabolic alterations associated with obesity and respective changes after bariatric surgery are incompletely understood. OBJECTIVE We comprehensively assessed metabolic alterations associated with severe obesity and distinct bariatric procedures. DESIGN In our longitudinal observational study, we applied a (1)H-nuclear magnetic resonance-based global, untargeted metabolomics strategy on human serum samples that were collected before and repeatedly ≤1 y after distinct bariatric procedures [i.e., a sleeve gastrectomy, proximal Roux-en Y gastric bypass (RYGB), and distal RYGB]. For comparison, we also analyzed serum samples from normal-weight and less-obese subjects who were matched for 1-y postoperative body mass index (BMI) values of the surgical groups. RESULTS We identified a metabolomic fingerprint in obese subjects that was clearly discriminated from that of normal-weight subjects. Furthermore, we showed that bariatric surgery (sleeve gastrectomy and proximal and distal RYGB) dynamically affected this fingerprint in a procedure-dependent manner, thereby establishing new fingerprints that could be discriminated from those of BMI-matched and normal-weight control subjects. Metabolites that largely contributed to the metabolomic fingerprints of severe obesity were aromatic and branched-chain amino acids (elevated), metabolites related to energy metabolism (pyruvate and citrate; elevated), and metabolites suggested to be derived from gut microbiota (formate, methanol, and isopropanol; all elevated). CONCLUSION Our data indicate that bariatric surgery, irrespective of the specific kind of procedure used, reverses most of the metabolic alterations associated with obesity and suggest profound changes in gut microbiome-host interactions after the surgery. This trial was registered at clinicaltrials.gov as NCT02480322.
Collapse
Affiliation(s)
- Ewa Gralka
- FiorGen Foundation, Sesto Fiorentino, Italy; Magnetic Resonance Center and
| | - Claudio Luchinat
- Magnetic Resonance Center and Chemistry Department, University of Florence, Sesto Fiorentino, Italy; and
| | | | - Barbara Ernst
- eSwiss Medical & Surgical Center, Interdisciplinary Obesity Center, St. Gallen, Switzerland
| | - Martin Thurnheer
- eSwiss Medical & Surgical Center, Interdisciplinary Obesity Center, St. Gallen, Switzerland
| | - Bernd Schultes
- eSwiss Medical & Surgical Center, Interdisciplinary Obesity Center, St. Gallen, Switzerland
| |
Collapse
|
217
|
Anhê FF, Varin TV, Le Barz M, Desjardins Y, Levy E, Roy D, Marette A. Gut Microbiota Dysbiosis in Obesity-Linked Metabolic Diseases and Prebiotic Potential of Polyphenol-Rich Extracts. Curr Obes Rep 2015; 4:389-400. [PMID: 26343880 DOI: 10.1007/s13679-015-0172-9] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Trillions of microorganisms inhabit the human body, strongly colonizing the gastro-intestinal tract and outnumbering our own cells. High-throughput sequencing techniques and new bioinformatic tools have enabled scientists to extend our knowledge on the relationship between the gut microbiota and host's physiology. Disruption of the ecological equilibrium in the gut (i.e., dysbiosis) has been associated with several pathological processes, including obesity and its related comorbidities, with diet being a strong determinant of gut microbial balance. In this review, we discuss the potential prebiotic effect of polyphenol-rich foods and extracts and how they can reshape the gut microbiota, emphasizing the novel role of the mucin-degrading bacterium Akkermansia muciniphila in their metabolic benefits.
Collapse
Affiliation(s)
- Fernando F Anhê
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Quebec, Canada.
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 boulevard Hochelaga, Québec, QC, G1V 0A6, Canada.
- Hôpital Laval, Pavillon Marguerite d'Youville, Quebec, QC, G1V 4G5, Canada.
| | - Thibault V Varin
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 boulevard Hochelaga, Québec, QC, G1V 0A6, Canada.
| | - Mélanie Le Barz
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Quebec, Canada.
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 boulevard Hochelaga, Québec, QC, G1V 0A6, Canada.
- Hôpital Laval, Pavillon Marguerite d'Youville, Quebec, QC, G1V 4G5, Canada.
| | - Yves Desjardins
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 boulevard Hochelaga, Québec, QC, G1V 0A6, Canada.
| | - Emile Levy
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 boulevard Hochelaga, Québec, QC, G1V 0A6, Canada.
- Research Centre, CHU-Sainte-Justine and Department of Nutrition, Université de Montréal, 3175, Chemin de la Côte Ste Catherine, bureau 5731A, Montreal, QC, H3T 1C5, Canada.
- Department of Nutrition, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.
| | - Denis Roy
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 boulevard Hochelaga, Québec, QC, G1V 0A6, Canada.
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Quebec, Canada.
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 boulevard Hochelaga, Québec, QC, G1V 0A6, Canada.
- Hôpital Laval, Pavillon Marguerite d'Youville, Quebec, QC, G1V 4G5, Canada.
| |
Collapse
|
218
|
Boulet MM, Chevrier G, Grenier-Larouche T, Pelletier M, Nadeau M, Scarpa J, Prehn C, Marette A, Adamski J, Tchernof A. Alterations of plasma metabolite profiles related to adipose tissue distribution and cardiometabolic risk. Am J Physiol Endocrinol Metab 2015; 309:E736-46. [PMID: 26306599 DOI: 10.1152/ajpendo.00231.2015] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/11/2015] [Indexed: 12/17/2022]
Abstract
Metabolomic profiling of obese individuals revealed altered concentrations of many metabolites, especially branched-chain amino acids (BCAA), possibly linked to altered adipose tissue BCAA catabolism. We tested the hypothesis that some features of this metabolite signature relate closely to visceral obesity and concomitant alterations in cardiometabolic risk factors. We also postulated that alterations in BCAA-catabolizing enzymes are predominant in visceral adipose tissue. Fifty-nine women (BMI 20-41 kg/m(2)) undergoing gynecologic surgery were recruited and characterized for overall and regional adiposity, blood metabolite levels using targeted metabolomics, and cardiometabolic risk factors. Adipose samples (visceral and subcutaneous) were obtained and used for gene expression and Western blot analyses. Obese women had significantly higher circulating BCAA and kynurenine/tryptophan (Kyn/Trp) ratio than lean or overweight women (P < 0.01). Principal component analysis confirmed that factors related to AA and the Kyn/Trp ratio were positively associated with BMI, fat mass, visceral or subcutaneous adipose tissue area, and subcutaneous adipocyte size (P ≤ 0.05). AA-related factor was positively associated with HOMA-IR (P ≤ 0.01). Factors reflecting glycerophospholipids and sphingolipids levels were mostly associated with altered blood lipid concentrations (P ≤ 0.05). Glutamate level was the strongest independent predictor of visceral adipose tissue area (r = 0.46, P < 0.001). Obese women had lower expression and protein levels of BCAA-catabolizing enzymes in visceral adipose tissue than overweight or lean women (P ≤ 0.05). We conclude that among metabolites altered in obesity plasma concentrations of BCAA and the Kyn/Trp ratio are closely related to increased adiposity. Alterations in expression and protein levels of BCAA-catabolizing enzymes are predominant in visceral adipose tissue.
Collapse
Affiliation(s)
- Marie Michèle Boulet
- Endocrinology and Nephrology, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada; School of Nutrition, Laval University, Quebec City, Canada; Quebec Heart and Lung Institute, Quebec City, Canada
| | | | | | - Mélissa Pelletier
- Endocrinology and Nephrology, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada
| | | | - Julia Scarpa
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
| | - Cornelia Prehn
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
| | - André Marette
- Quebec Heart and Lung Institute, Quebec City, Canada
| | - Jerzy Adamski
- Helmholtz Zentrum München, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany; Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan, Germany; and German Center for Diabetes Research, Neuherberg, Germany
| | - André Tchernof
- Endocrinology and Nephrology, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada; School of Nutrition, Laval University, Quebec City, Canada; Quebec Heart and Lung Institute, Quebec City, Canada;
| |
Collapse
|
219
|
Glynn EL, Piner LW, Huffman KM, Slentz CA, Elliot-Penry L, AbouAssi H, White PJ, Bain JR, Muehlbauer MJ, Ilkayeva OR, Stevens RD, Porter Starr KN, Bales CW, Volpi E, Brosnan MJ, Trimmer JK, Rolph TP, Newgard CB, Kraus WE. Impact of combined resistance and aerobic exercise training on branched-chain amino acid turnover, glycine metabolism and insulin sensitivity in overweight humans. Diabetologia 2015; 58:2324-35. [PMID: 26254576 PMCID: PMC4793723 DOI: 10.1007/s00125-015-3705-6] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 06/24/2015] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESES Obesity is associated with decreased insulin sensitivity (IS) and elevated plasma branched-chain amino acids (BCAAs). The purpose of this study was to investigate the relationship between BCAA metabolism and IS in overweight (OW) individuals during exercise intervention. METHODS Whole-body leucine turnover, IS by hyperinsulinaemic-euglycaemic clamp, and circulating and skeletal muscle amino acids, branched-chain α-keto acids and acylcarnitines were measured in ten healthy controls (Control) and nine OW, untrained, insulin-resistant individuals (OW-Untrained). OW-Untrained then underwent a 6 month aerobic and resistance exercise programme and repeated testing (OW-Trained). RESULTS IS was higher in Control vs OW-Untrained and increased significantly following exercise. IS was lower in OW-Trained vs Control expressed relative to body mass, but was not different from Control when normalised to fat-free mass (FFM). Plasma BCAAs and leucine turnover (relative to FFM) were higher in OW-Untrained vs Control, but did not change on average with exercise. Despite this, within individuals, the decrease in molar sum of circulating BCAAs was the best metabolic predictor of improvement in IS. Circulating glycine levels were higher in Control and OW-Trained vs OW-Untrained, and urinary metabolic profiling suggests that exercise induces more efficient elimination of excess acyl groups derived from BCAA and aromatic amino acid (AA) metabolism via formation of urinary glycine adducts. CONCLUSIONS/INTERPRETATION A mechanism involving more efficient elimination of excess acyl groups derived from BCAA and aromatic AA metabolism via glycine conjugation in the liver, rather than increased BCAA disposal through oxidation and turnover, may mediate interactions between exercise, BCAA metabolism and IS. TRIAL REGISTRATION Clinicaltrials.gov NCT01786941.
Collapse
Affiliation(s)
- Erin L Glynn
- Sarah W. Stedman Nutrition & Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
| | - Lucy W Piner
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
| | - Kim M Huffman
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
| | - Cris A Slentz
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Lorraine Elliot-Penry
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
| | - Hiba AbouAssi
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Division of Endocrinology, Duke University Medical Center, Durham, NC, USA
| | - Phillip J White
- Sarah W. Stedman Nutrition & Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
| | - James R Bain
- Sarah W. Stedman Nutrition & Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Division of Endocrinology, Duke University Medical Center, Durham, NC, USA
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition & Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
| | - Olga R Ilkayeva
- Sarah W. Stedman Nutrition & Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
| | - Robert D Stevens
- Sarah W. Stedman Nutrition & Metabolism Center, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Division of Endocrinology, Duke University Medical Center, Durham, NC, USA
| | | | - Connie W Bales
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Division of Geriatrics, Duke University Medical Center, Durham, NC, USA
- GRECC, Durham VA Medical Center, Durham, NC, USA
| | - Elena Volpi
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX, USA
| | - M Julia Brosnan
- The CV and Metabolic Diseases Research Unit, Pfizer, Cambridge, MA, USA
| | - Jeff K Trimmer
- The CV and Metabolic Diseases Research Unit, Pfizer, Cambridge, MA, USA
| | - Timothy P Rolph
- The CV and Metabolic Diseases Research Unit, Pfizer, Cambridge, MA, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition & Metabolism Center, Duke University Medical Center, Durham, NC, USA.
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA.
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.
- Division of Endocrinology, Duke University Medical Center, Durham, NC, USA.
| | - William E Kraus
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA.
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.
- Department of Cardiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
220
|
Yang RY, Wang SM, Sun L, Liu JM, Li HX, Sui XF, Wang M, Xiu HL, Wang S, He Q, Dong J, Chen WX. Association of branched-chain amino acids with coronary artery disease: A matched-pair case-control study. Nutr Metab Cardiovasc Dis 2015; 25:937-942. [PMID: 26231617 DOI: 10.1016/j.numecd.2015.06.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/25/2015] [Accepted: 06/05/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND AIM Several recent studies have found an independent relationship between levels of plasma branched-chain amino acids (BCAAs) and risk factors for coronary artery disease (CAD); however, few studies have investigated the associations of BCAAs with CAD and the risk of cardiovascular events. Therefore, the aim of this study was to investigate the relationship between BCAAs and CAD. METHODS AND RESULTS We studied 143 patients with CAD diagnosed by coronary angiography at Beijing Hospital (Beijing, China) during 2008-2011. Apparently healthy control individuals (n = 286) and the patients with CAD were matched (2:1 ratio) by age and gender. The healthy control individuals were selected at random from a set of subjects who attended an annual physical examination at the same hospital in 2011. Conditional logistic regression models were used to evaluate the associations between measured variables and CAD. After multivariate adjustment for traditional CAD risk factors, each one-standard-deviation increase in BCAA concentration was associated with an approximately twofold increase in the risk of CAD (odds ratio = 1.63, 95% confidence interval (CI): 1.21-2.20, P = 0.001). As compared with subjects in the lowest quartile of BCAA levels, the odds ratios (95% CIs) for CAD risk in subjects belonging to quartiles 2, 3, and 4 were 1.65 (0.75-3.61), 2.04 (0.92-4.53), and 3.86 (1.71-8.69), respectively (P trend = 0.01). CONCLUSION Our results demonstrate that BCAAs are significantly related to CAD development. This relationship is independent of diabetes, hypertension, dyslipidemia, and body mass index.
Collapse
Affiliation(s)
- R Y Yang
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - S M Wang
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - L Sun
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - J M Liu
- Department of Cardiology, Beijing Hospital, Ministry of Health, Beijing, China
| | - H X Li
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - X F Sui
- The First Affiliated Hospital, Jiamusi University, Heilongjiang, China
| | - M Wang
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China; Beijing Hospital, National Center for Clinical Laboratories, Ministry of Health, Beijing, China
| | - H L Xiu
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - S Wang
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Q He
- Department of Cardiology, Beijing Hospital, Ministry of Health, Beijing, China
| | - J Dong
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China.
| | - W X Chen
- The Key Laboratory of Geriatrics, Beijing Hospital, Beijing Institute of Geriatrics, Ministry of Health, Beijing, China; Beijing Hospital, National Center for Clinical Laboratories, Ministry of Health, Beijing, China
| |
Collapse
|
221
|
Connelly MA. Nuclear Magnetic Resonance Measured Serum Biomarkers and Type 2 Diabetes Risk Stratification. ACTA ACUST UNITED AC 2015. [DOI: 10.15406/jdmdc.2015.02.00050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
222
|
Kang I, Choi S, Ha TJ, Choi M, Wi HR, Lee BW, Lee M. Effects of Mung Bean (Vigna radiata L.) Ethanol Extracts Decrease Proinflammatory Cytokine-Induced Lipogenesis in the KK-Ay Diabese Mouse Model. J Med Food 2015; 18:841-9. [PMID: 25826234 PMCID: PMC4523062 DOI: 10.1089/jmf.2014.3364] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/13/2015] [Indexed: 01/27/2023] Open
Abstract
Rapid increase in the prevalence of obesity-related metabolic inflammatory diseases has led to research focused on nutraceuticals for their treatment. This study investigated the effects of the ethanol extracts of mung bean testa (MBT) on the metabolic inflammation-induced lipogenesis in gastrocnemius muscle of KK-Ay diabese mice. Ethanol extracts of MBT were orally administered to diabese KK-Ay mice for 4 weeks after diet-induced obesity model was generated by feeding a 60% high-fat diet for 3 weeks. Although there were no changes in body weight gain, MBT treatments decreased total weight of white adipose tissue. MBT also decreased triacylglycerol and total cholesterol levels in the muscle by 30%, which was correlated with suppression of lipogenic genes such as ACC, C/EBP alpha, PGC-1 alpha, and PPAR gamma. In particular, decreased levels of p-ERK1/2, PPAR gamma, and C/EBP alpha in the MBT-treated groups suggest that MBT might inhibit adipogenesis and decrease differentiation via the MEK/ERK pathway. Furthermore, significantly lower amounts of plasma interleukin (IL)-6 and intramuscular tumor necrosis factor (TNF)-alpha and monocyte chemoattractant protein-1 (MCP-1) were detected in MBT groups, confirming the anti-inflammatory effect of mung bean. In addition, our in vitro pilot study with 3T3-L1 cells showed that vitexin, the functional chemical in MBT, inhibited inflammation-induced lipogenesis with significantly lower amounts of IL-6 and MCP-1 after 14 days of vitexin treatment. Thus, the functional compounds in the mung bean ethanol extracts such as vitexin and isovitexin may regulate intracellular lipogenesis and adipogenesis via anti-inflammatory mechanisms and MEK/ERK pathway in the KK-Ay mouse model.
Collapse
Affiliation(s)
- Inhae Kang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Seojin Choi
- Department of Food and Nutrition, Research Institute of Obesity Sciences, Sungshin Women's University, Seoul, Korea
| | - Tae Joung Ha
- Department of Functional Crop, National Institute of Crop Science, RDA, Miryang, Korea
| | - Munji Choi
- Department of Food and Nutrition, Research Institute of Obesity Sciences, Sungshin Women's University, Seoul, Korea
| | - Hae-Ri Wi
- Department of Food and Nutrition, Research Institute of Obesity Sciences, Sungshin Women's University, Seoul, Korea
| | - Byong Won Lee
- Department of Functional Crop, National Institute of Crop Science, RDA, Miryang, Korea
| | - Myoungsook Lee
- Department of Food and Nutrition, Research Institute of Obesity Sciences, Sungshin Women's University, Seoul, Korea
| |
Collapse
|
223
|
Yamakado M, Nagao K, Imaizumi A, Tani M, Toda A, Tanaka T, Jinzu H, Miyano H, Yamamoto H, Daimon T, Horimoto K, Ishizaka Y. Plasma Free Amino Acid Profiles Predict Four-Year Risk of Developing Diabetes, Metabolic Syndrome, Dyslipidemia, and Hypertension in Japanese Population. Sci Rep 2015; 5:11918. [PMID: 26156880 PMCID: PMC4496670 DOI: 10.1038/srep11918] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 06/11/2015] [Indexed: 02/07/2023] Open
Abstract
Plasma free amino acid (PFAA) profile is highlighted in its association with visceral obesity and hyperinsulinemia, and future diabetes. Indeed PFAA profiling potentially can evaluate individuals' future risks of developing lifestyle-related diseases, in addition to diabetes. However, few studies have been performed especially in Asian populations, about the optimal combination of PFAAs for evaluating health risks. We quantified PFAA levels in 3,701 Japanese subjects, and determined visceral fat area (VFA) and two-hour post-challenge insulin (Ins120 min) values in 865 and 1,160 subjects, respectively. Then, models between PFAA levels and the VFA or Ins120 min values were constructed by multiple linear regression analysis with variable selection. Finally, a cohort study of 2,984 subjects to examine capabilities of the obtained models for predicting four-year risk of developing new-onset lifestyle-related diseases was conducted. The correlation coefficients of the obtained PFAA models against VFA or Ins120 min were higher than single PFAA level. Our models work well for future risk prediction. Even after adjusting for commonly accepted multiple risk factors, these models can predict future development of diabetes, metabolic syndrome, and dyslipidemia. PFAA profiles confer independent and differing contributions to increasing the lifestyle-related disease risks in addition to the currently known factors in a general Japanese population.
Collapse
Affiliation(s)
- Minoru Yamakado
- Center for Multiphasic Health Testing and Services, Mitsui Memorial Hospital, 1 Kanda, Izumicho, Chiyoda-ku, Tokyo 101-8643, Japan
| | - Kenji Nagao
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Akira Imaizumi
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Mizuki Tani
- Center for Multiphasic Health Testing and Services, Mitsui Memorial Hospital, 1 Kanda, Izumicho, Chiyoda-ku, Tokyo 101-8643, Japan
| | - Akiko Toda
- Center for Multiphasic Health Testing and Services, Mitsui Memorial Hospital, 1 Kanda, Izumicho, Chiyoda-ku, Tokyo 101-8643, Japan
| | - Takayuki Tanaka
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Hiroko Jinzu
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Hiroshi Miyano
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Hiroshi Yamamoto
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Takashi Daimon
- Department of Biostatistics, Hyogo College of Medicine, 1-1, Mukogawa-cho, Nishinomiya, Japan
| | - Katsuhisa Horimoto
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Yuko Ishizaka
- Center for Multiphasic Health Testing and Services, Mitsui Memorial Hospital, 1 Kanda, Izumicho, Chiyoda-ku, Tokyo 101-8643, Japan
| |
Collapse
|
224
|
Liu L, Wang X, Li Y, Sun C. Postprandial Differences in the Amino Acid and Biogenic Amines Profiles of Impaired Fasting Glucose Individuals after Intake of Highland Barley. Nutrients 2015; 7:5556-5571. [PMID: 26184292 PMCID: PMC4517015 DOI: 10.3390/nu7075238] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/15/2015] [Accepted: 07/01/2015] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was to measure the postprandial changes in amino acid and biogenic amine profiles in individuals with impaired fasting glucose (IFG) and to investigate the changes of postprandial amino acid and biogenic amine profiles after a meal of highland barley (HB). Firstly, 50 IFG and 50 healthy individuals were recruited for the measurement of 2 h postprandial changes of amino acid and biogenic amine profiles after a glucose load. Secondly, IFG individuals received three different loads: Glucose (GL), white rice (WR) and HB. Amino acid and biogenic amine profiles, glucose and insulin were assayed at time zero and 30, 60, 90 and 120 min after the test load. The results showed fasting and postprandial amino acid and biogenic amine profiles were different between the IFG group and the controls. The level of most amino acids and their metabolites decreased after an oral glucose tolerance test, while the postprandial level of γ-aminobutyric acid (GABA) increased significantly in IFG individuals. After three different test loads, the area under the curve for glucose, insulin, lysine and GABA after a HB load decreased significantly compared to GL and WR loads. Furthermore, the postprandial changes in the level of GABA between time zero and 120 min during a HB load were associated positively with 2 h glucose and fasting insulin secretion in the IFG individuals. Thus, the HB load produced low postprandial glucose and insulin responses, which induced changes in amino acid and biogenic amine profiles and improved insulin sensitivity.
Collapse
Affiliation(s)
- Liyan Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin 150086, China.
| | - Xinyang Wang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin 150086, China.
| | - Ying Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin 150086, China.
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin 150086, China.
| |
Collapse
|
225
|
Tripathy D, Cobb JE, Gall W, Adam KP, George T, Schwenke DC, Banerji M, Bray GA, Buchanan TA, Clement SC, Henry RR, Kitabchi AE, Mudaliar S, Ratner RE, Stentz FB, Reaven PD, Musi N, Ferrannini E, DeFronzo RA. A novel insulin resistance index to monitor changes in insulin sensitivity and glucose tolerance: the ACT NOW study. J Clin Endocrinol Metab 2015; 100:1855-62. [PMID: 25603459 PMCID: PMC4422894 DOI: 10.1210/jc.2014-3824] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/15/2015] [Indexed: 01/22/2023]
Abstract
OBJECTIVE The objective was to test the clinical utility of Quantose M(Q) to monitor changes in insulin sensitivity after pioglitazone therapy in prediabetic subjects. Quantose M(Q) is derived from fasting measurements of insulin, α-hydroxybutyrate, linoleoyl-glycerophosphocholine, and oleate, three nonglucose metabolites shown to correlate with insulin-stimulated glucose disposal. RESEARCH DESIGN AND METHODS Participants were 428 of the total of 602 ACT NOW impaired glucose tolerance (IGT) subjects randomized to pioglitazone (45 mg/d) or placebo and followed for 2.4 years. At baseline and study end, fasting plasma metabolites required for determination of Quantose, glycated hemoglobin, and oral glucose tolerance test with frequent plasma insulin and glucose measurements to calculate the Matsuda index of insulin sensitivity were obtained. RESULTS Pioglitazone treatment lowered IGT conversion to diabetes (hazard ratio = 0.25; 95% confidence interval = 0.13-0.50; P < .0001). Although glycated hemoglobin did not track with insulin sensitivity, Quantose M(Q) increased in pioglitazone-treated subjects (by 1.45 [3.45] mg·min(-1)·kgwbm(-1)) (median [interquartile range]) (P < .001 vs placebo), as did the Matsuda index (by 3.05 [4.77] units; P < .0001). Quantose M(Q) correlated with the Matsuda index at baseline and change in the Matsuda index from baseline (rho, 0.85 and 0.79, respectively; P < .0001) and was progressively higher across closeout glucose tolerance status (diabetes, IGT, normal glucose tolerance). In logistic models including only anthropometric and fasting measurements, Quantose M(Q) outperformed both Matsuda and fasting insulin in predicting incident diabetes. CONCLUSIONS In IGT subjects, Quantose M(Q) parallels changes in insulin sensitivity and glucose tolerance with pioglitazone therapy. Due to its strong correlation with improved insulin sensitivity and its ease of use, Quantose M(Q) may serve as a useful clinical test to identify and monitor therapy in insulin-resistant patients.
Collapse
Affiliation(s)
- Devjit Tripathy
- Texas Diabetes Institute (D.T., N.M., R.A.D.), University of Texas Health Science Center, San Antonio, Texas 78207; South Texas Veterans Health Care System (D.T., N.M., R.A.D.), Audie L. Murphy Division, San Antonio, Texas 78228; Metabolon, Inc (J.E.C., W.G., K.-P.A., T.G.), Durham, North Carolina 27713; Phoenix VA Health Care System (D.C.S., P.D.R.), Phoenix, Arizona 85012; College of Nursing and Health Care Innovation (D.C.S.), Arizona State University, Phoenix, Arizona 85004; SUNY Health Science Center at Brooklyn (M.A.B.), Brooklyn, New York 11203; Pennington Biomedical Research Center/Louisiana State University (G.A.B.), Baton Rouge, Louisiana 70808; University of Southern California Keck School of Medicine (T.A.B.), Los Angeles, California 90033; VA San Diego Healthcare System and University of California at San Diego (R.R.H., S.M.), San Diego, California 92161; Division of Endocrinology, Diabetes and Metabolism (A.E.K., F.B.S.), University of Tennessee, Memphis, Tennessee 38163; Inova Fairfax Hospital (S.C.C.), Falls Church, Virginia 22042; Medstar Research Institute (R.E.R.), Hyattsville, Maryland 20782; and Department of Clinical and Experimental Medicine (E.F.), CNR Institute of Clinical Physiology, 56126 Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Yamada C, Kondo M, Kishimoto N, Shibata T, Nagai Y, Imanishi T, Oroguchi T, Ishii N, Nishizaki Y. Association between insulin resistance and plasma amino acid profile in non-diabetic Japanese subjects. J Diabetes Investig 2015. [PMID: 26221519 PMCID: PMC4511300 DOI: 10.1111/jdi.12323] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aims/Introduction Elevation of the branched-chain amino acids (BCAAs), valine, leucine and isoleucine; and the aromatic amino acids, tyrosine and phenylalanine, has been observed in obesity-related insulin resistance. However, there have been few studies on Asians, who are generally less obese and less insulin-resistant than Caucasian or African-Americans. In the present study, we investigated the relationship between homeostasis model assessment of insulin resistance (HOMA-IR) and plasma amino acid concentration in non-diabetic Japanese participants. Materials and Methods A total of 94 healthy men and women were enrolled, and plasma amino acid concentration was measured by liquid chromatography/mass spectrometry after overnight fasting. The associations between HOMA-IR and 20 amino acid concentrations, and anthropometric and clinical parameters of lifestyle-related diseases were evaluated. Results The mean age and body mass index were 40.1 ± 9.6 years and 22.7 ± 3.9, respectively. Significantly positive correlations were observed between HOMA-IR and valine, isoleucine, leucine, tyrosine, phenylalanine and total BCAA concentration. Compared with the HOMA-IR ≤ 1.6 group, the HOMA-IR > 1.6 group showed significantly exacerbated anthropometric and clinical parameters, and significantly elevated levels of valine, isoleucine, leucine, tyrosine, phenylalanine and BCAA. Conclusions The present study shows that the insulin resistance-related change in amino acid profile is also observed in non-diabetic Japanese subjects. These amino acids include BCAAs (valine, isoleucine and leucine) and aromatic amino acids (tyrosine and phenylalanine), in agreement with previous studies carried out using different ethnic groups with different degrees of obesity and insulin resistance.
Collapse
Affiliation(s)
| | | | | | - Takeo Shibata
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine Tokyo, Japan
| | - Yoko Nagai
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine Tokyo, Japan
| | - Tadashi Imanishi
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine Tokyo, Japan
| | - Takashige Oroguchi
- Life Care Center, Graduate School of Medicine, Tokai University Tokyo, Japan
| | - Naoaki Ishii
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine Tokyo, Japan ; Life Care Center, Graduate School of Medicine, Tokai University Tokyo, Japan
| | - Yasuhiro Nishizaki
- Tokai University Tokyo Hospital Tokyo, Japan ; Life Care Center, Graduate School of Medicine, Tokai University Tokyo, Japan
| |
Collapse
|
227
|
Piccolo BD, Comerford KB, Karakas SE, Knotts TA, Fiehn O, Adams SH. Whey protein supplementation does not alter plasma branched-chained amino acid profiles but results in unique metabolomics patterns in obese women enrolled in an 8-week weight loss trial. J Nutr 2015; 145:691-700. [PMID: 25833773 DOI: 10.3945/jn.114.203943] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/29/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND It has been suggested that perturbations in branched-chain amino acid (BCAA) catabolism are associated with insulin resistance and contribute to elevated systemic BCAAs. Evidence in rodents suggests dietary protein rich in BCAAs can increase BCAA catabolism, but there is limited evidence in humans. OBJECTIVE We hypothesize that a diet rich in BCAAs will increase BCAA catabolism, which will manifest in a reduction of fasting plasma BCAA concentrations. METHODS The metabolome of 27 obese women with metabolic syndrome before and after weight loss was investigated to identify changes in BCAA metabolism using GC-time-of-flight mass spectrometry. Subjects were enrolled in an 8-wk weight-loss study including either a 20-g/d whey (whey group, n = 16) or gelatin (gelatin group, n = 11) protein supplement. When matched for total protein by weight, whey protein has 3 times the amount of BCAAs compared with gelatin protein. RESULTS Postintervention plasma abundances of Ile (gelatin group: 637 ± 18, quantifier ion peak height ÷ 100; whey group: 744 ± 65), Leu (gelatin group: 1210 ± 33; whey group: 1380 ± 79), and Val (gelatin group: 2080 ± 59; whey group: 2510 ± 230) did not differ between treatment groups. BCAAs were significantly correlated with homeostasis model assessment of insulin resistance at baseline (r = 0.52, 0.43, and 0.49 for Leu, Ile, and Val, respectively; all, P < 0.05), but correlations were no longer significant at postintervention. Pro- and Cys-related pathways were found discriminant of whey protein vs. gelatin protein supplementation in multivariate statistical analyses. CONCLUSIONS These findings suggest that BCAA metabolism is, at best, only modestly affected at a whey protein supplementation dose of 20 g/d. Furthermore, the loss of an association between postintervention BCAA and homeostasis model assessment suggests that factors associated with calorie restriction or protein intake affect how plasma BCAAs relate to insulin sensitivity. This trial was registered at clinicaltrials.gov as NCT00739479.
Collapse
Affiliation(s)
- Brian D Piccolo
- Obesity and Metabolism Research Unit, USDA, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA
| | - Kevin B Comerford
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The University of California Davis Medical Center, Sacramento, CA; and Department of Nutrition
| | - Sidika E Karakas
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The University of California Davis Medical Center, Sacramento, CA; and
| | - Trina A Knotts
- Obesity and Metabolism Research Unit, USDA, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA; Department of Nutrition
| | - Oliver Fiehn
- West Coast Metabolomics Center, and Genome Center, University of California, Davis, Davis, CA
| | - Sean H Adams
- Obesity and Metabolism Research Unit, USDA, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA; Department of Nutrition,
| |
Collapse
|
228
|
Tso SC, Gui WJ, Wu CY, Chuang JL, Qi X, Skvora KJ, Dork K, Wallace AL, Morlock LK, Lee BH, Hutson SM, Strom SC, Williams NS, Tambar UK, Wynn RM, Chuang DT. Benzothiophene carboxylate derivatives as novel allosteric inhibitors of branched-chain α-ketoacid dehydrogenase kinase. J Biol Chem 2015; 289:20583-93. [PMID: 24895126 DOI: 10.1074/jbc.m114.569251] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mitochondrial branched-chain α-ketoacid dehydrogenase complex (BCKDC) is negatively regulated by reversible phosphorylation.BCKDC kinase (BDK) inhibitors that augment BCKDC flux have been shown to reduce branched-chain amino acid (BCAA) concentrations in vivo. In the present study, we employed high-throughput screens to identify compound 3,6- dichlorobenzo[b]thiophene-2-carboxylic acid (BT2) as a novel BDK inhibitor (IC(50) = 3.19 μM). BT2 binds to the same site in BDK as other known allosteric BDK inhibitors, including (S)-α-cholorophenylproprionate ((S)-CPP). BT2 binding to BDK triggers helix movements in the N-terminal domain, resulting in the dissociation of BDK from the BCKDC accompanied by accelerated degradation of the released kinase in vivo. BT2 shows excellent pharmacokinetics (terminal T(1⁄2) = 730 min) and metabolic stability (no degradation in 240 min), which are significantly better than those of (S)-CPP. BT2, its analog 3-chloro-6-fluorobenzo[ b]thiophene-2-carboxylic acid (BT2F), and a prodrug of BT2 (i.e. N-(4-acetamido-1,2,5-oxadiazol-3-yl)-3,6-dichlorobenzo[ b]thiophene-2-carboxamide (BT3)) significantly increase residual BCKDC activity in cultured cells and primary hepatocytes from patients and a mouse model of maple syrup urine disease. Administration of BT2 at 20 mg/kg/day to wild-type mice for 1 week leads to nearly complete dephosphorylation and maximal activation of BCKDC in heart, muscle, kidneys, and liver with reduction in plasma BCAA concentrations. The availability of benzothiophene carboxylate derivatives as stable BDK inhibitors may prove useful for the treatment of metabolic disease caused by elevated BCAA concentrations.
Collapse
|
229
|
Su X, Magkos F, Zhou D, Eagon JC, Fabbrini E, Okunade AL, Klein S. Adipose tissue monomethyl branched-chain fatty acids and insulin sensitivity: Effects of obesity and weight loss. Obesity (Silver Spring) 2015; 23:329-34. [PMID: 25328153 PMCID: PMC4310778 DOI: 10.1002/oby.20923] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 01/24/2023]
Abstract
OBJECTIVES An increase in circulating branched-chain amino acids (BCAA) is associated with insulin resistance. Adipose tissue is a potentially important site for BCAA metabolism. It was evaluated whether monomethyl branched-chain fatty acids (mmBCFA) in adipose tissue, which are likely derived from BCAA catabolism, are associated with insulin sensitivity. METHODS Insulin-stimulated glucose disposal was determined by using the hyperinsulinemic-euglycemic clamp procedure with stable isotope glucose tracer infusion in nine lean and nine obese subjects, and in a separate group of nine obese subjects before and 1 year after Roux-en-Y gastric bypass (RYGB) surgery (38% weight loss). Adipose tissue mmBCFA content was measured in tissue biopsies taken in the basal state. RESULTS Total adipose tissue mmBCFA content was ∼30% lower in obese than lean subjects (P=0.02) and increased by ∼65% after weight loss in the RYGB group (P=0.01). Adipose tissue mmBCFA content correlated positively with skeletal muscle insulin sensitivity (R(2) =35%, P=0.01, n=18). CONCLUSIONS These results demonstrate a novel association between adipose tissue mmBCFA content and obesity-related insulin resistance. Additional studies are needed to determine whether the association between adipose tissue mmBCFA and muscle insulin sensitivity is causal or a simple association.
Collapse
Affiliation(s)
- Xiong Su
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, China
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Faidon Magkos
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Dequan Zhou
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - J. Christopher Eagon
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Elisa Fabbrini
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Adewole L. Okunade
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
230
|
Lian K, Du C, Liu Y, Zhu D, Yan W, Zhang H, Hong Z, Liu P, Zhang L, Pei H, Zhang J, Gao C, Xin C, Cheng H, Xiong L, Tao L. Impaired adiponectin signaling contributes to disturbed catabolism of branched-chain amino acids in diabetic mice. Diabetes 2015; 64:49-59. [PMID: 25071024 DOI: 10.2337/db14-0312] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The branched-chain amino acids (BCAA) accumulated in type 2 diabetes are independent contributors to insulin resistance. The activity of branched-chain α-keto acid dehydrogenase (BCKD) complex, rate-limiting enzyme in BCAA catabolism, is reduced in diabetic states, which contributes to elevated BCAA concentrations. However, the mechanisms underlying decreased BCKD activity remain poorly understood. Here, we demonstrate that mitochondrial phosphatase 2C (PP2Cm), a newly identified BCKD phosphatase that increases BCKD activity, was significantly downregulated in ob/ob and type 2 diabetic mice. Interestingly, in adiponectin (APN) knockout (APN(-/-)) mice fed with a high-fat diet (HD), PP2Cm expression and BCKD activity were significantly decreased, whereas BCKD kinase (BDK), which inhibits BCKD activity, was markedly increased. Concurrently, plasma BCAA and branched-chain α-keto acids (BCKA) were significantly elevated. APN treatment markedly reverted PP2Cm, BDK, BCKD activity, and BCAA and BCKA levels in HD-fed APN(-/-) and diabetic animals. Additionally, increased BCKD activity caused by APN administration was partially but significantly inhibited in PP2Cm knockout mice. Finally, APN-mediated upregulation of PP2Cm expression and BCKD activity were abolished when AMPK was inhibited. Collectively, we have provided the first direct evidence that APN is a novel regulator of PP2Cm and systematic BCAA levels, suggesting that targeting APN may be a pharmacological approach to ameliorating BCAA catabolism in the diabetic state.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/genetics
- AMP-Activated Protein Kinases/metabolism
- Adiponectin/genetics
- Adiponectin/metabolism
- Amino Acids, Branched-Chain/metabolism
- Animals
- Cells, Cultured
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diet, High-Fat
- Hepatocytes/cytology
- Hepatocytes/metabolism
- Male
- Maple Syrup Urine Disease/genetics
- Maple Syrup Urine Disease/metabolism
- Metabolism/physiology
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Obese
- Phosphoprotein Phosphatases/metabolism
- Protein Phosphatase 2C
- RNA, Small Interfering/genetics
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Kun Lian
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chaosheng Du
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yi Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Di Zhu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Haifeng Zhang
- Experiment Teaching Center, Fourth Military Medical University, Xi'an, China
| | - Zhibo Hong
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peilin Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China Department of Cardiology, 306th Hospital of PLA, Beijing, China
| | - Lijian Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Haifeng Pei
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinglong Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chao Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chao Xin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hexiang Cheng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lize Xiong
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
231
|
Yokoi N, Beppu M, Yoshida E, Hoshikawa R, Hidaka S, Matsubara T, Shinohara M, Irino Y, Hatano N, Seino S. Identification of putative biomarkers for prediabetes by metabolome analysis of rat models of type 2 diabetes. Metabolomics 2015; 11:1277-1286. [PMID: 26366137 PMCID: PMC4559098 DOI: 10.1007/s11306-015-0784-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 02/05/2015] [Indexed: 10/26/2022]
Abstract
Biomarkers for the development of type 2 diabetes (T2D) are useful for prediction and intervention of the disease at earlier stages. In this study, we performed a longitudinal study of changes in metabolites using an animal model of T2D, the spontaneously diabetic Torii (SDT) rat. Fasting plasma samples of SDT and control Sprague-Dawley (SD) rats were collected from 6 to 24 weeks of age, and subjected to gas chromatography-mass spectrometry-based metabolome analysis. Fifty-nine hydrophilic metabolites were detected in plasma samples, including amino acids, carbohydrates, sugars and organic acids. At 12 weeks of age, just before the onset of diabetes in SDT rats, the amounts of nine of these metabolites (asparagine, glutamine, glycerol, kynurenine, mannose, n-alpha-acetyllysine, taurine, threonine, and tryptophan) in SDT rats were significantly different from those in SD rats. In particular, metabolites in the tryptophan metabolism pathway (tryptophan and kynurenine) were decreased in SDT rats at 12 weeks of age and later. The lower tryptophan and kynurenine levels in the prediabetic state and later were further confirmed by a replication study on SDT rats and by a longitudinal study on another animal model of T2D, the Otsuka Long-Evans Tokushima Fatty rat. Our data indicate that tryptophan and its metabolites are potential biomarkers for prediabetes and that tryptophan metabolism may be a potential target of intervention for treatment of the disease.
Collapse
Affiliation(s)
- Norihide Yokoi
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| | - Masayuki Beppu
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, 650-0017 Japan
| | - Eri Yoshida
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, 650-0017 Japan
| | - Ritsuko Hoshikawa
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| | - Shihomi Hidaka
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| | - Toshiya Matsubara
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
- Life Science Research Center, Technology Research Laboratory, Shimadzu Corporation, Kyoto, 619-0237 Japan
| | - Masami Shinohara
- Tokyo Animal and Diet Department, CLEA Japan, Inc., Meguro-ku, Tokyo, 153-8533 Japan
| | - Yasuhiro Irino
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017 Japan
- Division of Evidenced-based Laboratory Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017 Japan
| | - Naoya Hatano
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017 Japan
| | - Susumu Seino
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, 650-0017 Japan
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017 Japan
| |
Collapse
|
232
|
Lips MA, Van Klinken JB, van Harmelen V, Dharuri HK, 't Hoen PAC, Laros JFJ, van Ommen GJ, Janssen IM, Van Ramshorst B, Van Wagensveld BA, Swank DJ, Van Dielen F, Dane A, Harms A, Vreeken R, Hankemeier T, Smit JWA, Pijl H, Willems van Dijk K. Roux-en-Y gastric bypass surgery, but not calorie restriction, reduces plasma branched-chain amino acids in obese women independent of weight loss or the presence of type 2 diabetes. Diabetes Care 2014; 37:3150-6. [PMID: 25315204 DOI: 10.2337/dc14-0195] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Obesity and type 2 diabetes mellitus (T2DM) have been associated with increased levels of circulating branched-chain amino acids (BCAAs) that may be involved in the pathogenesis of insulin resistance. However, weight loss has not been consistently associated with the reduction of BCAA levels. RESEARCH DESIGN AND METHODS We included 30 obese normal glucose-tolerant (NGT) subjects, 32 obese subjects with T2DM, and 12 lean female subjects. Obese subjects underwent either a restrictive procedure (gastric banding [GB], a very low-calorie diet [VLCD]), or a restrictive/bypass procedure (Roux-en-Y gastric bypass [RYGB] surgery). Fasting blood samples were taken for the determination of amine group containing metabolites 4 weeks before, as well as 3 weeks and 3 months after the intervention. RESULTS BCAA levels were higher in T2DM subjects, but not in NGT subjects, compared with lean subjects. Principal component (PC) analysis revealed a concise PC consisting of all BCAAs, which showed a correlation with measures of insulin sensitivity and glucose tolerance. Only after the RYGB procedure, and at both 3 weeks and 3 months, were circulating BCAA levels reduced. CONCLUSIONS Our data confirm an association between deregulation of BCAA metabolism in plasma and insulin resistance and glucose intolerance. Three weeks after undergoing RYGB surgery, a significant decrease in BCAAs in both NGT as well as T2DM subjects was observed. After 3 months, despite inducing significant weight loss, neither GB nor VLCD induced a reduction in BCAA levels. Our results indicate that the bypass procedure of RYGB surgery, independent of weight loss or the presence of T2DM, reduces BCAA levels in obese subjects.
Collapse
Affiliation(s)
- Mirjam A Lips
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan B Van Klinken
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Vanessa van Harmelen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Harish K Dharuri
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeroen F J Laros
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Gert-Jan van Ommen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Ignace M Janssen
- Department of Surgery, Rijnstate Ziekenhuis, Arnhem, the Netherlands
| | - Bert Van Ramshorst
- Department of Surgery, St. Antonius Ziekenhuis, Nieuwegein, the Netherlands
| | | | | | | | - Adrie Dane
- Leiden Amsterdam Centre for Drug Research, Netherlands Metabolomics Centre, Leiden, the Netherlands
| | - Amy Harms
- Leiden Amsterdam Centre for Drug Research, Netherlands Metabolomics Centre, Leiden, the Netherlands
| | - Rob Vreeken
- Leiden Amsterdam Centre for Drug Research, Netherlands Metabolomics Centre, Leiden, the Netherlands
| | - Thomas Hankemeier
- Leiden Amsterdam Centre for Drug Research, Netherlands Metabolomics Centre, Leiden, the Netherlands
| | - Johannes W A Smit
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - Hanno Pijl
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - Ko Willems van Dijk
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, the Netherlands Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| |
Collapse
|
233
|
Abstract
Branched-chain amino acids (BCAAs) are important nutrient signals that have direct and indirect effects. Frequently, BCAAs have been reported to mediate antiobesity effects, especially in rodent models. However, circulating levels of BCAAs tend to be increased in individuals with obesity and are associated with worse metabolic health and future insulin resistance or type 2 diabetes mellitus (T2DM). A hypothesized mechanism linking increased levels of BCAAs and T2DM involves leucine-mediated activation of the mammalian target of rapamycin complex 1 (mTORC1), which results in uncoupling of insulin signalling at an early stage. A BCAA dysmetabolism model proposes that the accumulation of mitotoxic metabolites (and not BCAAs per se) promotes β-cell mitochondrial dysfunction, stress signalling and apoptosis associated with T2DM. Alternatively, insulin resistance might promote aminoacidaemia by increasing the protein degradation that insulin normally suppresses, and/or by eliciting an impairment of efficient BCAA oxidative metabolism in some tissues. Whether and how impaired BCAA metabolism might occur in obesity is discussed in this Review. Research on the role of individual and model-dependent differences in BCAA metabolism is needed, as several genes (BCKDHA, PPM1K, IVD and KLF15) have been designated as candidate genes for obesity and/or T2DM in humans, and distinct phenotypes of tissue-specific branched chain ketoacid dehydrogenase complex activity have been detected in animal models of obesity and T2DM.
Collapse
Affiliation(s)
- Christopher J Lynch
- Cellular and Molecular Physiology Department, The Pennsylvania State University, 500 University Drive, MC-H166, Hershey, PA 17033, USA
| | - Sean H Adams
- Arkansas Children's Nutrition Center, and Department of Pediatrics, University of Arkansas for Medical Sciences, 15 Children's Way, Little Rock, AR 72202, USA
| |
Collapse
|
234
|
Muscelli E, Frascerra S, Casolaro A, Baldi S, Mari A, Gall W, Cobb J, Ferrannini E. The amino acid response to a mixed meal in patients with type 2 diabetes: effect of sitagliptin treatment. Diabetes Obes Metab 2014; 16:1140-7. [PMID: 25040945 DOI: 10.1111/dom.12350] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/05/2014] [Accepted: 06/30/2014] [Indexed: 11/30/2022]
Abstract
AIMS Amino acid (AA) metabolism is altered in type 2 diabetes (T2D), and fasting levels of α-hydroxybutyrate (α-HB), a biomarker for insulin resistance, have been suggested to track AA metabolism. We investigated the changes in AA and α-HB induced by a mixed-meal tolerance test (MTT) and the effects of sitagliptin treatment. METHODS Forty-seven T2D patients [56 ± 7 years, body mass index (BMI) 29.9 ± 4.2 kg/m(2) ] were randomized to sitagliptin (100 mg/day, 6 weeks) or placebo. Seven age- and BMI-matched non-diabetic subjects served as control (CT). RESULTS During a 5-h MTT, branched-chain AA (BCAA) peaked earlier in T2D than CT [75(25) vs. 62(3) mmol/l · h over 2 h, median(interquartile range), p = 0.05], and rose higher [5-h increment: 31(23) vs. 19(24) mmol/l · h, p = 0.05]. Fasting α-HB was higher [7.5(2.7) vs. 5.9(1.3) µg/ml, p = 0.04 T2D vs. CT], and its meal-induced increments were larger [24(99) vs. -41(86) µg/ml · h, p = 0.006]. Plasma non-esterified fatty acids (NEFA) declined during MTT, but their increments were greater in patients (53 ± 16 vs. 35 ± 10 mEq/l · h, p = 0.005). Compared to placebo, both BCAA [-6.4(21.1) vs. 0.0(48.0) mmol/l · h, p = 0.01] and α-HB increments [-114(250) vs. 114(428) µg/ml · h, p = 0.002] decreased with sitagliptin, and meal-induced NEFA suppression was improved. Changes in BCAA and α-HB were reciprocally related to changes in insulin sensitivity (ρ = -0.37 and -0.43, p ≤ 0.01). CONCLUSIONS T2D is associated with a hyperaminoacidaemic response to MTT, which circulating α-HB levels track. Sitagliptin-induced glycaemic improvement was associated with reductions in BCAA and α-HB excursions and better NEFA suppression, in parallel with improved insulin sensitivity, confirming that α-HB is a readout of metabolic overload.
Collapse
Affiliation(s)
- E Muscelli
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Chiu S, Williams PT, Dawson T, Bergman RN, Stefanovski D, Watkins SM, Krauss RM. Diets high in protein or saturated fat do not affect insulin sensitivity or plasma concentrations of lipids and lipoproteins in overweight and obese adults. J Nutr 2014; 144:1753-9. [PMID: 25332473 PMCID: PMC4195419 DOI: 10.3945/jn.114.197624] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Previous human studies reported inconsistent effects of dietary protein and branched-chain amino acids (BCAAs) on insulin action and glucose metabolism. Similarly, it is unclear whether saturated fat (SF) intake influences these metabolic variables. OBJECTIVE The objective of this study was to test the effects of high [30% of energy (%E)] vs. moderate (20%E) intakes of protein (primarily whey) on insulin action and lipid and lipoprotein concentrations in the context of both high (15%E) and low (7%E) SF diets. METHODS The study was conducted as a randomized controlled trial in 158 overweight and obese men and women. After a 4-wk baseline diet [55%E carbohydrate, 15%E protein, 30%E fat (7%E SF)], participants were randomly assigned to 4 wk of either the baseline diet or 1 of 4 test diets containing 35%E carbohydrate and either 20%E or 30%E protein and either 7%E or 15%E SF. Frequently sampled i.v. glucose tolerance tests were administered after each dietary period. RESULTS Other than significantly higher fasting glucose concentrations for high vs. moderate protein intakes with a low-fat diet (difference ± SE: 0.47 ± 0.14 mmol/L; P = 0.001), there were no significant effects of dietary protein or SF on glucose metabolism, plasma insulin, or concentrations of lipids and lipoproteins. Changes in plasma BCAAs across all diets were negatively correlated with changes in the metabolic clearance rate of insulin (ρ = -0.18, P = 0.03) and positively correlated with changes in the acute insulin response to glucose (ρ = 0.15, P = 0.05). CONCLUSIONS These findings suggest that short-term intake of BCAAs can influence insulin dynamics. However, in this group of overweight and obese individuals, neither high protein nor SF intake affected insulin sensitivity or plasma concentrations of lipids and lipoproteins. This trial was registered at clinicaltrials.gov as NCT00508937.
Collapse
Affiliation(s)
- Sally Chiu
- Children’s Hospital Oakland Research Institute, Oakland, CA
| | | | - Taylor Dawson
- Children’s Hospital Oakland Research Institute, Oakland, CA
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA; and
| | - Darko Stefanovski
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA; and
| | | | - Ronald M. Krauss
- Children’s Hospital Oakland Research Institute, Oakland, CA,To whom correspondence should be addressed. E-mail:
| |
Collapse
|
236
|
Omental adipose tissue gene expression, gene variants, branched-chain amino acids, and their relationship with metabolic syndrome and insulin resistance in humans. GENES AND NUTRITION 2014; 9:431. [PMID: 25260659 PMCID: PMC4176839 DOI: 10.1007/s12263-014-0431-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/19/2014] [Indexed: 12/31/2022]
Abstract
Obesity is a complex disorder caused by several factors. Thus, the aim of the present study was to assess whether the expression of genes in the omental white adipose tissue (AT) of subjects with insulin resistance (IR) or metabolic syndrome (MetS) is associated with an elevation in serum branched-chain amino acids (BCAAs) and whether this response depends on specific genetic variants. Serum BCAA concentration, the adipocyte area, and gene variants of PPARγ, ABCA1, FTO, TCF7L2, GFOD2,BCAT2, and BCKDH were determined in 115 Mexican subjects. The gene expression in the AT and adipocytes of BCAT, BCKDH E1α, C/EBPα, PPARγ2, SREBP-1, PPARα, UCP1, leptin receptor, leptin, adiponectin, and TNFα was measured in 51 subjects. Subjects with IR showed higher values for the BMI, HOMA-IR, and adipocyte area and higher levels of serum glucose, insulin, leptin, and C-reactive protein, as well as an elevation of the AT gene expression of SREBP-1, leptin, and TNFα and a significant reduction in the expression of adiponectin, BCAT2, and BCKDH E1α, compared with non-IR subjects. The presence of MetS was associated with higher HOMA-IR as well as higher serum BCAA concentrations. Subjects with the genetic variants for BCAT2 and BCKDH E1 α showed a lower serum BCAA concentration, and those with the ABCA1 and FTO gene variant showed higher levels of insulin and HOMA-IR than non-IR subjects. AT dysfunction is the result of a combination of the presence of some genetic variants, altered AT gene expression, the presence of MetS risk factors, IR, and serum BCAA concentrations.
Collapse
|
237
|
Demine S, Reddy N, Renard P, Raes M, Arnould T. Unraveling biochemical pathways affected by mitochondrial dysfunctions using metabolomic approaches. Metabolites 2014; 4:831-78. [PMID: 25257998 PMCID: PMC4192695 DOI: 10.3390/metabo4030831] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 09/02/2014] [Accepted: 09/18/2014] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial dysfunction(s) (MDs) can be defined as alterations in the mitochondria, including mitochondrial uncoupling, mitochondrial depolarization, inhibition of the mitochondrial respiratory chain, mitochondrial network fragmentation, mitochondrial or nuclear DNA mutations and the mitochondrial accumulation of protein aggregates. All these MDs are known to alter the capacity of ATP production and are observed in several pathological states/diseases, including cancer, obesity, muscle and neurological disorders. The induction of MDs can also alter the secretion of several metabolites, reactive oxygen species production and modify several cell-signalling pathways to resolve the mitochondrial dysfunction or ultimately trigger cell death. Many metabolites, such as fatty acids and derived compounds, could be secreted into the blood stream by cells suffering from mitochondrial alterations. In this review, we summarize how a mitochondrial uncoupling can modify metabolites, the signalling pathways and transcription factors involved in this process. We describe how to identify the causes or consequences of mitochondrial dysfunction using metabolomics (liquid and gas chromatography associated with mass spectrometry analysis, NMR spectroscopy) in the obesity and insulin resistance thematic.
Collapse
Affiliation(s)
- Stéphane Demine
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| | - Nagabushana Reddy
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| | - Patricia Renard
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| | - Martine Raes
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| | - Thierry Arnould
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| |
Collapse
|
238
|
Bergman RN, Stefanovski D, Kim SP. Systems analysis and the prediction and prevention of Type 2 diabetes mellitus. Curr Opin Biotechnol 2014; 28:165-70. [PMID: 24976265 PMCID: PMC5931209 DOI: 10.1016/j.copbio.2014.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 05/29/2014] [Accepted: 05/30/2014] [Indexed: 01/22/2023]
Abstract
Prevalence of Type 2 diabetes has increased at an alarming rate, highlighting the need to correctly predict the development of this disease in order to allow intervention and thus, slow progression of the disease and resulting metabolic derangement. There have been many recent 'advances' geared toward the detection of pre-diabetes, including genome wide association studies and metabolomics. Although these approaches generate a large amount of data with a single blood sample, studies have indicated limited success using genetic and metabolomics information alone for identification of disease risk. Clinical assessment of the disposition index (DI), based on the hyperbolic law of glucose tolerance, is a powerful predictor of Type 2 diabetes, but is not easily assessed in the clinical setting. Thus, it is evident that combining genetic or metabolomic approaches for a more simple assessment of DI may provide a useful tool to identify those at highest risk for Type 2 diabetes, allowing for intervention and prevention.
Collapse
Affiliation(s)
- Richard N Bergman
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA 90048, USA.
| | - Darko Stefanovski
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA 90048, USA
| | - Stella P Kim
- Cedars-Sinai Diabetes and Obesity Research Institute, Los Angeles, CA 90048, USA
| |
Collapse
|
239
|
Zheng Y, Qi L. Diet and lifestyle interventions on lipids: combination with genomics and metabolomics. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/clp.14.30] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
240
|
Kitsy A, Carney S, Vivar JC, Knight MS, Pointer MA, Gwathmey JK, Ghosh S. Effects of leucine supplementation and serum withdrawal on branched-chain amino acid pathway gene and protein expression in mouse adipocytes. PLoS One 2014; 9:e102615. [PMID: 25050624 PMCID: PMC4106850 DOI: 10.1371/journal.pone.0102615] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/21/2014] [Indexed: 12/25/2022] Open
Abstract
The essential branched-chain amino acids (BCAA), leucine, valine and isoleucine, are traditionally associated with skeletal muscle growth and maintenance, energy production, and generation of neurotransmitter and gluconeogenic precursors. Recent evidence from human and animal model studies has established an additional link between BCAA levels and obesity. However, details of the mechanism of regulation of BCAA metabolism during adipogenesis are largely unknown. We interrogated whether the expression of genes and proteins involved in BCAA metabolism are sensitive to the adipocyte differentiation process, and responsive to nutrient stress from starvation or BCAA excess. Murine 3T3-L1 preadipocytes were differentiated to adipocytes under control conditions and under conditions of L-leucine supplementation or serum withdrawal. RNA and proteins were isolated at days 0, 4 and 10 of differentiation to represent pre-differentiation, early differentiation and late differentiation stages. Expression of 16 BCAA metabolism genes was quantified by quantitative real-time PCR. Expression of the protein levels of branched-chain amino acid transaminase 2 (Bcat2) and branched-chain alpha keto acid dehydrogenase (Bckdha) was quantified by immunoblotting. Under control conditions, all genes displayed induction of gene expression during early adipogenesis (Day 4) compared to Day 0. Leucine supplementation resulted in an induction of Bcat2 and Bckdha genes during early and late differentiation. Western blot analysis demonstrated condition-specific concordance between gene and protein expression. Serum withdrawal resulted in undetectable Bcat2 and Bckdha protein levels at all timepoints. These results demonstrate that the expression of genes related to BCAA metabolism are regulated during adipocyte differentiation and influenced by nutrient levels. These results provide additional insights on how BCAA metabolism is associated with adipose tissue function and extends our understanding of the transcriptomic response of this pathway to variations in nutrient availability.
Collapse
Affiliation(s)
- Abderrazak Kitsy
- Division of Cardiometabolic Disorders, Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, United States of America
| | - Skyla Carney
- Division of Cardiometabolic Disorders, Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, United States of America
| | - Juan C. Vivar
- Division of Cardiometabolic Disorders, Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, United States of America
| | - Megan S. Knight
- Division of Cardiometabolic Disorders, Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, United States of America
| | - Mildred A. Pointer
- Division of Cardiometabolic Disorders, Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, United States of America
| | - Judith K. Gwathmey
- Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sujoy Ghosh
- Division of Cardiometabolic Disorders, Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, United States of America
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore
- * E-mail:
| |
Collapse
|
241
|
Rietman A, Schwarz J, Tomé D, Kok FJ, Mensink M. High dietary protein intake, reducing or eliciting insulin resistance? Eur J Clin Nutr 2014; 68:973-9. [DOI: 10.1038/ejcn.2014.123] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 05/13/2014] [Accepted: 05/21/2014] [Indexed: 02/07/2023]
|
242
|
Yang R, Dong J, Zhao H, Li H, Guo H, Wang S, Zhang C, Wang S, Wang M, Yu S, Chen W. Association of branched-chain amino acids with carotid intima-media thickness and coronary artery disease risk factors. PLoS One 2014; 9:e99598. [PMID: 24910999 PMCID: PMC4049830 DOI: 10.1371/journal.pone.0099598] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/16/2014] [Indexed: 12/26/2022] Open
Abstract
Background Recent studies have determined that branched-chain (BCAAs) and aromatic (AAAs) amino acids are strongly correlated with obesity and atherogenic dyslipidemia and are strong predictors of diabetes. However, it is not clear if these amino acids are capable of identifying subjects with coronary artery disease (CAD), particularly with subclinical atherosclerosis who are at risk of developing CAD. Methods Four hundred and seventy two Chinese subjects (272 males and 200 females, 42–97 y of age) undergoing physical exams were recruited at random for participation in the cross-sectional study. Serum BCAAs and AAAs were measured using our previously reported isotope dilution liquid chromatography tandem mass spectrometry method. Bilateral B-mode carotid artery images for carotid intima-media thickness (cIMT) were acquired at end diastole and cIMT values more than 0.9 mm were categorized as increased. Correlations of BCAAs with cIMT and other CAD risk factors were analyzed. Results BCAAs and AAAs were significantly and positively associated with risk factors of CAD, e.g., cIMT, BMI, waist circumference, blood pressure, fasting blood glucose, TG, apoB, apoB/apoAI ratio, apoCII, apoCIII and hsCRP, and were significantly and negatively associated with HDL-C and apoAI. Stepwise multiple linear regression analysis revealed that age (β = 0.175, P<0.001), log BCAA (β = 0.147, P<0.001) and systolic blood pressure (β = 0.141, P = 0.012) were positively and independently associated with cIMT. In the logistic regression model, the most and only powerful laboratory factor correlated with increased cIMT was BCAA (the odds ratio of the fourth quartile compared to the first quartile was 2.679; P = 0.009). Conclusion BCAAs are independently correlated with increased cIMT. This correlation would open a new field of research in the mechanistic understanding and risk assessment of CAD.
Collapse
Affiliation(s)
- Ruiyue Yang
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Jun Dong
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Haijian Zhao
- Beijing Hospital and National Center for Clinical Laboratories, Ministry of Health, Beijing, China
| | - Hongxia Li
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Hanbang Guo
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Shu Wang
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Chuanbao Zhang
- Beijing Hospital and National Center for Clinical Laboratories, Ministry of Health, Beijing, China
| | - Siming Wang
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
| | - Mo Wang
- Beijing Hospital and National Center for Clinical Laboratories, Ministry of Health, Beijing, China
| | - Songlin Yu
- Beijing Hospital and National Center for Clinical Laboratories, Ministry of Health, Beijing, China
| | - Wenxiang Chen
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, China
- Beijing Hospital and National Center for Clinical Laboratories, Ministry of Health, Beijing, China
- * E-mail:
| |
Collapse
|
243
|
Beasley JM, Wylie-Rosett J. The role of dietary proteins among persons with diabetes. Curr Atheroscler Rep 2014; 15:348. [PMID: 23881544 DOI: 10.1007/s11883-013-0348-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Examining the role of dietary protein and establishing intake guidelines among individuals with diabetes is complex. The 2013 American Diabetes Association (ADA) standards of care recommend an individualized approach to decision making with regard to protein intake and dietary macronutrient composition. Needs may vary based on cardiometabolic risk factors and renal function. Among individuals with impaired renal function, the ADA recommends reducing protein intake to 0.8-1.0 g/kg per day in earlier stages of chronic kidney disease (CKD), and to 0.8 g/kg per day in the later stages of CKD. Epidemiological studies suggest animal protein may increase risk of diabetes; however, few data are available to suggest how protein sources influence diabetes complications.
Collapse
Affiliation(s)
- Jeannette M Beasley
- Albert Einstein College of Medicine, Department of Epidemiology and Population Health, 1300 Morris Park Avenue Suite 1312c, Bronx, NY 10461, USA.
| | | |
Collapse
|
244
|
Hanzu FA, Vinaixa M, Papageorgiou A, Párrizas M, Correig X, Delgado S, Carmona F, Samino S, Vidal J, Gomis R. Obesity rather than regional fat depots marks the metabolomic pattern of adipose tissue: an untargeted metabolomic approach. Obesity (Silver Spring) 2014; 22:698-704. [PMID: 23804579 DOI: 10.1002/oby.20541] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 05/25/2013] [Accepted: 05/26/2013] [Indexed: 01/04/2023]
Abstract
OBJECTIVE This study compares the patterns of visceral (VIS) and subcutaneous (SC) adipose tissue (AT)-derived metabolites from non-obese (BMI 24-26 kg/m2) and obese subjects (BMI > 40 kg/m2) with no major metabolic risk factors other than BMI. METHODS SC- and VIS- AT obtained from obese (Ob) and non-obese (NOb) subjects during surgery were incubated to obtain their metabolites. Differences related to obesity or anatomical provenances of AT were assessed using an untargeted metabolomics approach based on gas chromatography-mass spectrometry. RESULTS The overall effect of obesity on the metabolite profile resulted more remarkable than the effect of regional AT. Only the depletion of 2-ketoisocaproic (2-KIC) acid reached statistical significance for the SC-AT alone, although it was observed in both depots. Obesity induced more significant changes in several amino acids levels of the VIS-AT metabolites. On the one hand, higher released levels of glutamine and alanine were detected in the VIS- obese AT, whereas on the other, the VIS- obese AT presented a diminished uptake of essential amino acids (methionine, threonine, lysine), BCAAs, leucine, and serine. CONCLUSION This study shows that obesity markedly affects the amino acid metabolic signature of the AT before the clinical onset of other significant metabolic alterations aside from BMI.
Collapse
Affiliation(s)
- F A Hanzu
- Department of Endocrinology and Nutrition, Diabetes and Obesity Research Laboratory, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Spégel P, Lindqvist A, Sandberg M, Wierup N. Glucose-dependent insulinotropic polypeptide lowers branched chain amino acids in hyperglycemic rats. ACTA ACUST UNITED AC 2014; 189:11-6. [PMID: 24412429 DOI: 10.1016/j.regpep.2013.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 12/28/2013] [Accepted: 12/31/2013] [Indexed: 10/25/2022]
Abstract
Hypersecretion of the incretin hormone glucose-dependent insulinotropic polypeptide (GIP) has been associated with obesity and glucose intolerance. This condition has been suggested to be linked to GIP resistance. Besides its insulinotropic effect, GIP also directly affects glucose uptake and lipid metabolism. This notwithstanding, effects of GIP on other circulating metabolites than glucose have not been thoroughly investigated. Here, we examined effects of infusion of various concentrations of GIP in normo- and hyperglycemic rats on serum metabolite profiles. We found that, despite a decrease in serum glucose levels (-26%, p<0.01), the serum metabolite profile was largely unaffected by GIP infusion in normoglycemic rats. Interestingly, levels of branched chain amino acids and the ketone body β-hydroxybutyrate were decreased by 21% (p<0.05) and 27% (p<0.001), respectively, in hyperglycemic rats infused with 60 ng/ml GIP. Hence, our data suggest that GIP provokes a decrease in BCAA levels and ketone body production. Increased concentrations of these metabolites have been associated with obesity and T2D.
Collapse
Affiliation(s)
- Peter Spégel
- Molecular Metabolism, Lund University Diabetes Centre, Malmö, Sweden.
| | | | - Monica Sandberg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Nils Wierup
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö Sweden
| |
Collapse
|
246
|
Thalacker-Mercer AE, Ingram KH, Guo F, Ilkayeva O, Newgard CB, Garvey WT. BMI, RQ, diabetes, and sex affect the relationships between amino acids and clamp measures of insulin action in humans. Diabetes 2014; 63:791-800. [PMID: 24130332 PMCID: PMC3900549 DOI: 10.2337/db13-0396] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Previous studies have used indirect measures of insulin sensitivity to link circulating amino acids with insulin resistance and identify potential biomarkers of diabetes risk. Using direct measures (i.e., hyperinsulinemic-euglycemic clamps), we examined the relationships between the metabolomic amino acid profile and insulin action (i.e., glucose disposal rate [GDR]). Relationships between GDR and serum amino acids were determined among insulin-sensitive, insulin-resistant, and type 2 diabetic (T2DM) individuals. In all subjects, glycine (Gly) had the strongest correlation with GDR (positive association), followed by leucine/isoleucine (Leu/Ile) (negative association). These relationships were dramatically influenced by BMI, the resting respiratory quotient (RQ), T2DM, and sex. Gly had a strong positive correlation with GDR regardless of BMI, RQ, or sex but became nonsignificant in T2DM. In contrast, Leu/Ile was negatively associated with GDR in nonobese and T2DM subjects. Increased resting fat metabolism (i.e., low RQ) and obesity were observed to independently promote and negate the association between Leu/Ile and insulin resistance, respectively. Additionally, the relationship between Leu/Ile and GDR was magnified in T2DM males. Future studies are needed to determine whether Gly has a mechanistic role in glucose homeostasis and whether dietary Gly enrichment may be an effective intervention in diseases characterized by insulin resistance.
Collapse
Affiliation(s)
- Anna E. Thalacker-Mercer
- Department of Cell, Developmental and Integrative Biology, University of Alabama, Birmingham, AL
- Nutrition Sciences, University of Alabama, Birmingham, AL
- Birmingham Veterans Affairs Medical Center, Birmingham, AL
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Katherine H. Ingram
- Nutrition Sciences, University of Alabama, Birmingham, AL
- Department of Exercise Science and Sport Management, Kennesaw State University, Kennesaw, GA
| | - Fangjian Guo
- Nutrition Sciences, University of Alabama, Birmingham, AL
| | | | - Christopher B. Newgard
- Department of Medicine, Duke University, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - W. Timothy Garvey
- Nutrition Sciences, University of Alabama, Birmingham, AL
- Birmingham Veterans Affairs Medical Center, Birmingham, AL
- Corresponding author: W. Timothy Garvey,
| |
Collapse
|
247
|
Metabolomics identifies changes in fatty acid and amino acid profiles in serum of overweight older adults following a weight loss intervention. J Physiol Biochem 2014; 70:593-602. [DOI: 10.1007/s13105-013-0311-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 12/22/2013] [Indexed: 12/13/2022]
|
248
|
Campbell C, Grapov D, Fiehn O, Chandler CJ, Burnett DJ, Souza EC, Casazza GA, Gustafson MB, Keim NL, Newman JW, Hunter GR, Fernandez JR, Garvey WT, Harper ME, Hoppel CL, Meissen JK, Take K, Adams SH. Improved metabolic health alters host metabolism in parallel with changes in systemic xeno-metabolites of gut origin. PLoS One 2014; 9:e84260. [PMID: 24416208 PMCID: PMC3885560 DOI: 10.1371/journal.pone.0084260] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/13/2013] [Indexed: 12/18/2022] Open
Abstract
Novel plasma metabolite patterns reflective of improved metabolic health (insulin sensitivity, fitness, reduced body weight) were identified before and after a 14–17 wk weight loss and exercise intervention in sedentary, obese insulin-resistant women. To control for potential confounding effects of diet- or microbiome-derived molecules on the systemic metabolome, sampling was during a tightly-controlled feeding test week paradigm. Pairwise and multivariate analysis revealed intervention- and insulin-sensitivity associated: (1) Changes in plasma xeno-metabolites (“non-self” metabolites of dietary or gut microbial origin) following an oral glucose tolerance test (e.g. higher post-OGTT propane-1,2,3-tricarboxylate [tricarballylic acid]) or in the overnight-fasted state (e.g., lower γ-tocopherol); (2) Increased indices of saturated very long chain fatty acid elongation capacity; (3) Increased post-OGTT α-ketoglutaric acid (α-KG), fasting α-KG inversely correlated with Matsuda index, and altered patterns of malate, pyruvate and glutamine hypothesized to stem from improved mitochondrial efficiency and more robust oxidation of glucose. The results support a working model in which improved metabolic health modifies host metabolism in parallel with altering systemic exposure to xeno-metabolites. This highlights that interpretations regarding the origins of peripheral blood or urinary “signatures” of insulin resistance and metabolic health must consider the potentially important contribution of gut-derived metabolites toward the host's metabolome.
Collapse
Affiliation(s)
- Caitlin Campbell
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Dmitry Grapov
- West Coast Metabolomics Center, University of California Davis, Davis, California, United States of America
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, California, United States of America
- Genome Center, University of California Davis, Davis, California, United States of America
- * E-mail: (SHA); (OF)
| | - Carol J. Chandler
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Dustin J. Burnett
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Elaine C. Souza
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Gretchen A. Casazza
- Sports Medicine Program, University of California, Davis School of Medicine, Sacramento, California, United States of America
| | - Mary B. Gustafson
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Nancy L. Keim
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
- Department of Nutrition, University of California Davis, Davis, California, United States of America
| | - John W. Newman
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
- Department of Nutrition, University of California Davis, Davis, California, United States of America
| | - Gary R. Hunter
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama, United States of America
- Human Studies Department, University of Alabama, Birmingham, Alabama, United States of America
| | - Jose R. Fernandez
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama, United States of America
| | - W. Timothy Garvey
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama, United States of America
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Charles L. Hoppel
- Pharmacology Department, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - John K. Meissen
- Genome Center, University of California Davis, Davis, California, United States of America
| | - Kohei Take
- Genome Center, University of California Davis, Davis, California, United States of America
| | - Sean H. Adams
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
- Department of Nutrition, University of California Davis, Davis, California, United States of America
- * E-mail: (SHA); (OF)
| |
Collapse
|
249
|
Scholtens DM, Muehlbauer MJ, Daya NR, Stevens RD, Dyer AR, Lowe LP, Metzger BE, Newgard CB, Bain JR, Lowe WL. Metabolomics reveals broad-scale metabolic perturbations in hyperglycemic mothers during pregnancy. Diabetes Care 2014; 37:158-66. [PMID: 23990511 PMCID: PMC3867997 DOI: 10.2337/dc13-0989] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To characterize metabolites across the range of maternal glucose by comparing metabolomic profiles of mothers with high and low fasting plasma glucose (FPG). RESEARCH DESIGN AND METHODS We compared fasting serum from an oral glucose tolerance test at ∼28 weeks' gestation from 67 Northern European ancestry mothers from the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study with high (>90th percentile) FPG with 50 mothers with low (<10th percentile) FPG but comparable BMI. Metabolic data from biochemical analyses of conventional clinical metabolites, targeted mass spectrometry (MS)-based measurement of amino acids, and nontargeted gas chromatography/MS were subjected to per-metabolite analyses and collective pathway analyses using Unipathway annotation. RESULTS High-FPG mothers had a metabolic profile consistent with insulin resistance including higher triglycerides, 3-hydroxybutyrate, and amino acids including alanine, proline, and branched-chain amino acids (false discovery rate [FDR]-adjusted P < 0.05). Lower 1,5-anhydroglucitol in high-FPG mothers suggested recent hyperglycemic excursions (FDR-adjusted P < 0.05). Pathway analyses indicated differences in amino acid degradation pathways for the two groups (FDR-adjusted P < 0.05), consistent with population-based findings in nonpregnant populations. Exploratory analyses with newborn outcomes indicated positive associations for maternal triglycerides with neonatal sum of skinfolds and cord C-peptide and a negative association between maternal glycine and cord C-peptide (P < 0.05). CONCLUSIONS Metabolomics reveals perturbations in metabolism of major macronutrients and amino acid degradation pathways in high- versus low-FPG mothers.
Collapse
|
250
|
Mullooly N, Vernon W, Smith DM, Newsholme P. Elevated levels of branched-chain amino acids have little effect on pancreatic islet cells, but L-arginine impairs function through activation of the endoplasmic reticulum stress response. Exp Physiol 2013; 99:538-51. [PMID: 24334335 DOI: 10.1113/expphysiol.2013.077495] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent metabolic profiling studies have identified a correlation between branched-chain amino acid levels, insulin resistance associated with prediabetes and susceptibility to type 2 diabetes. Glucose and lipids in chronic excess have been reported to induce toxic effects in pancreatic β-cells, but the effect of elevated amino acid concentrations on primary islet cell function has not been investigated to date. The aim of this study was to investigate the effect of chronic exposure to various amino acids on islet cell function in vitro. Isolated rat islets were incubated over periods of 48 h with a range of concentrations of individual amino acids (0.1 μm to 10 mm). After 48 h, islets were assessed for glucose-dependent insulin secretion capacity, proliferation or islet cell apoptosis. We report that elevated levels of branched-chain amino acids have little effect on pancreatic islet cell function or viability; however, increased levels of the amino acid l-arginine were found to be β-cell toxic, causing a dose-dependent decrease in insulin secretion accompanied by a decrease in islet cell proliferation and an increase in islet cell apoptosis. These effects were not due to l-arginine-dependent increases in production of nitric oxide but arose through elicitation of the islet cell endoplasmic reticulum stress response. This novel finding indicates, for the first time, that the l-arginine concentration in vitro may impact negatively on islet cell function, thus indicating further complexity in relationship to in vivo susceptibility of β-cells to nutrient-induced dysfunction.
Collapse
Affiliation(s)
- Niamh Mullooly
- * Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia.
| | | | | | | |
Collapse
|