1
|
Gladding M, Shen X, Snyder MP, Havel PJ, Adams SH. Interindividual Variability in Postprandial Plasma Fructose Patterns in Adults. Nutrients 2024; 16:3079. [PMID: 39339679 PMCID: PMC11435096 DOI: 10.3390/nu16183079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
High fructose consumption is associated with an increased risk of cardiometabolic disease, and fructose feeding dose-dependently induces markers reflective of poor metabolic health. However, unlike glucose, surprisingly little is known about person-to-person differences in postprandial plasma fructose patterns. Herein, we performed post hoc analyses of two published studies to address this question. In the first cohort, 16 participants' all-day plasma fructose concentration patterns (08:00-23:30) were determined (8 women and 8 men) while consuming mixed meals (breakfast, lunch, and dinner) with a fructose-sweetened beverage at each meal (30% of calories). Individually plotted results demonstrate remarkably disparate fructose patterns with respect to peak concentration and timing. A secondary study confirmed substantial interindividual variability in plasma fructose patterns over 240 min in 16 adults consuming Ensure®, a commercially available mixed macronutrient drink containing a low dose of fructose. The health ramifications of interindividual variations in postprandial fructose metabolism and the underlying physiological mechanisms driving differences in post-meal blood patterns remain to be explored. Future research is warranted to determine if interindividual variability in fructose digestion, metabolism, and postprandial blood concentration patterns is associated with cardiometabolic health phenotypes and disease risk.
Collapse
Affiliation(s)
- Mia Gladding
- Department of Food Science and Nutrition, California Polytechnic University, San Luis Obispo, CA 93407, USA
| | - Xiaotao Shen
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA 94306, USA
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Stanford, CA 94306, USA
| | - Michael P Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA 94306, USA
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Stanford, CA 94306, USA
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Sean H Adams
- Department of Surgery, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Center for Alimentary and Metabolic Science, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
2
|
李 雨, 王 瑗, 袁 泉. [Latest Findings on the Role of α-Ketoglutarate in Metabolic Syndrome]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:783-792. [PMID: 38948289 PMCID: PMC11211801 DOI: 10.12182/20240560302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Indexed: 07/02/2024]
Abstract
Alpha-ketoglutarate (α-KG), an endogenous intermediate of the tricarboxylic acid cycle, is involved in a variety of cellular metabolic pathways. It serves as an energy donor, a precursor of amino acid biosynthesis, and an epigenetic regulator. α-KG plays physiological functions in immune regulation, oxidative stress, and anti-aging as well. In recent years, it has been reported that the level of α-KG in the body is closely associated with metabolic syndrome, including obesity, hyperglycemia, and other pathological factors. Exogenous supplementation of α-KG improves obesity, blood glucose levels, and cardiovascular disease risks associated with metabolic syndrome. Furthermore, α-KG regulates the common pathological mechanisms of metabolic syndrome, suggesting the potential application prospect of α-KG in metabolic syndrome. In order to provide a theoretical basis for further exploration of the application of α-KG in metabolic syndrome, we focused on α-KG and metabolic syndrome in this article and summarized the latest research progress in the role of α-KG in improving the pathological condition and disease progression of metabolic syndrome. For the next step, researchers may focus on the co-pathogenesis of metabolic syndrome and investigate whether α-KG can be used to achieve the therapeutic goal of "homotherapy for heteropathy" in the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- 雨含 李
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 种植科 (成都 610041)State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Dental Implant, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 瑗 王
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 种植科 (成都 610041)State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Dental Implant, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 泉 袁
- 口腔疾病防治全国重点实验室 国家口腔医学中心 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 种植科 (成都 610041)State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Dental Implant, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Dhillon J, Pandey S, Newman JW, Fiehn O, Ortiz RM. Metabolic Responses to an Acute Glucose Challenge: The Differential Effects of Eight Weeks of Almond vs. Cracker Consumption in Young Adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.19.24307571. [PMID: 38826341 PMCID: PMC11142291 DOI: 10.1101/2024.05.19.24307571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
This study investigated the dynamic responses to an acute glucose challenge following chronic almond versus cracker consumption for 8 weeks (clinicaltrials.gov ID: NCT03084003). Seventy-three young adults (age: 18-19 years, BMI: 18-41 kg/m2) participated in an 8-week randomized, controlled, parallel-arm intervention and were randomly assigned to consume either almonds (2 oz/d, n=38) or an isocaloric control snack of graham crackers (325 kcal/d, n=35) daily for 8 weeks. Twenty participants from each group underwent a 2-hour oral glucose tolerance test (oGTT) at the end of the 8-week intervention. Metabolite abundances in the oGTT serum samples were quantified using untargeted metabolomics, and targeted analyses for free PUFAs, total fatty acids, oxylipins, and endocannabinoids. Multivariate, univariate, and chemical enrichment analyses were conducted to identify significant metabolic shifts. Findings exhibit a biphasic lipid response distinguished by higher levels of unsaturated triglycerides in the earlier periods of the oGTT followed by lower levels in the latter period in the almond versus cracker group (p-value<0.05, chemical enrichment analyses). Almond (vs. cracker) consumption was also associated with higher AUC120 min of aminomalonate, and oxylipins (p-value<0.05), but lower AUC120 min of L-cystine, N-acetylmannosamine, and isoheptadecanoic acid (p-value<0.05). Additionally, the Matsuda Index in the almond group correlated with AUC120 min of CE 22:6 (r=-0.46; p-value<0.05) and 12,13 DiHOME (r=0.45; p-value<0.05). Almond consumption for 8 weeks leads to dynamic, differential shifts in response to an acute glucose challenge, marked by alterations in lipid and amino acid mediators involved in metabolic and physiological pathways.
Collapse
Affiliation(s)
- Jaapna Dhillon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia
- Department of Molecular and Cell Biology, University of California, Merced
| | - Saurabh Pandey
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia
- Jaypee University of Information Technology, Waknaghat, India
| | - John W. Newman
- West Coast Metabolomics Center, University of California, Davis
- Department of Nutrition, University of California, Davis
- Obesity and Metabolism Research Unit, USDA Agricultural Research Service Western Human Nutrition Research Center, University of California, Davis
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis
| | - Rudy M. Ortiz
- Department of Molecular and Cell Biology, University of California, Merced
| |
Collapse
|
4
|
Kasperek MC, Mailing L, Piccolo BD, Moody B, Lan R, Gao X, Hernandez‐Saavedra D, Woods JA, Adams SH, Allen JM. Exercise training modifies xenometabolites in gut and circulation of lean and obese adults. Physiol Rep 2023; 11:e15638. [PMID: 36945966 PMCID: PMC10031301 DOI: 10.14814/phy2.15638] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/23/2023] Open
Abstract
Regular, moderate exercise modifies the gut microbiome and contributes to human metabolic and immune health. The microbiome may exert influence on host physiology through the microbial production and modification of metabolites (xenometabolites); however, this has not been extensively explored. We hypothesized that 6 weeks of supervised, aerobic exercise 3×/week (60%-75% heart rate reserve [HRR], 30-60 min) in previously sedentary, lean (n = 14) and obese (n = 10) adults would modify both the fecal and serum xenometabolome. Serum and fecal samples were collected pre- and post-6 week intervention and analyzed by liquid chromatography/tandem mass spectrometry (LC-MS/MS). Linear mixed models (LMMs) identified multiple fecal and serum xenometabolites responsive to exercise training. Further cluster and pathway analysis revealed that the most prominent xenometabolic shifts occurred within aromatic amino acid (ArAA) metabolic pathways. Fecal and serum ArAA derivatives correlated with body composition (lean mass), markers of insulin sensitivity (insulin, HOMA-IR) and cardiorespiratory fitness (V ̇ O 2 max $$ \dot{\mathrm{V}}{\mathrm{O}}_{2\max } $$ ), both at baseline and in response to exercise training. Two serum aromatic microbial-derived amino acid metabolites that were upregulated following the exercise intervention, indole-3-lactic acid (ILA: fold change: 1.2, FDR p < 0.05) and 4-hydroxyphenyllactic acid (4-HPLA: fold change: 1.3, FDR p < 0.05), share metabolic pathways within the microbiota and were associated with body composition and markers of insulin sensitivity at baseline and in response to training. These data provide evidence of physiologically relevant shifts in microbial metabolism that occur in response to exercise training, and reinforce the view that host metabolic health influences gut microbiota population and function. Future studies should consider the microbiome and xenometabolome when investigating the health benefits of exercise.
Collapse
Affiliation(s)
- Mikaela C. Kasperek
- Division of Nutritional SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Lucy Mailing
- Division of Nutritional SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Brian D. Piccolo
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Becky Moody
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Renny Lan
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Xiaotian Gao
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Diego Hernandez‐Saavedra
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Jeffrey A. Woods
- Division of Nutritional SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Sean H. Adams
- Department of SurgeryUniversity of California, Davis School of MedicineSacramentoCaliforniaUSA
- Center for Alimentary and Metabolic ScienceUniversity of California, DavisSacramentoCaliforniaUSA
| | - Jacob M. Allen
- Division of Nutritional SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Kinesiology and Community HealthUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
5
|
Metabolomic Signatures for the Effects of Weight Loss Interventions on Severe Obesity in Children and Adolescents. Metabolites 2021; 12:metabo12010027. [PMID: 35050149 PMCID: PMC8778282 DOI: 10.3390/metabo12010027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/25/2021] [Indexed: 01/04/2023] Open
Abstract
Childhood obesity has increased worldwide, and many clinical and public interventions have attempted to reduce morbidity. We aimed to determine the metabolomic signatures associated with weight control interventions in children with obesity. Forty children from the “Intervention for Children and Adolescent Obesity via Activity and Nutrition (ICAAN)” cohort were selected according to intervention responses. Based on changes in body mass index z-scores, 20 were responders and the remaining non-responders. Their serum metabolites were quantitatively analyzed using capillary electrophoresis time-of-flight mass spectrometry at baseline and after 6 and 18 months of intervention. After 18 months of intervention, the metabolite cluster changes in the responders and non-responders showed a difference on the heatmap, but significant metabolites were not clear. However, regardless of the responses, 13 and 49 metabolites were significant in the group of children with obesity intervention at 6 months and 18 months post-intervention compared to baseline. In addition, the top five metabolic pathways (D-glutamine and D-glutamate metabolism; arginine biosynthesis; alanine, aspartate, and glutamate metabolism; TCA cycle (tricarboxylic acid cycle); valine, leucine, and isoleucine biosynthesis) including several amino acids in the metabolites of obese children after 18 months were significantly changed. Our study showed significantly different metabolomic profiles based on time post obesity-related intervention. Through this study, we can better understand and predict childhood obesity through metabolite analysis and monitoring.
Collapse
|
6
|
Kistner S, Döring M, Krüger R, Rist MJ, Weinert CH, Bunzel D, Merz B, Radloff K, Neumann R, Härtel S, Bub A. Sex-Specific Relationship between the Cardiorespiratory Fitness and Plasma Metabolite Patterns in Healthy Humans-Results of the KarMeN Study. Metabolites 2021; 11:463. [PMID: 34357357 PMCID: PMC8303204 DOI: 10.3390/metabo11070463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 11/25/2022] Open
Abstract
Cardiorespiratory fitness (CRF) represents a strong predictor of all-cause mortality and is strongly influenced by regular physical activity (PA). However, the biological mechanisms involved in the body's adaptation to PA remain to be fully elucidated. The aim of this study was to systematically examine the relationship between CRF and plasma metabolite patterns in 252 healthy adults from the cross-sectional Karlsruhe Metabolomics and Nutrition (KarMeN) study. CRF was determined by measuring the peak oxygen uptake during incremental exercise. Fasting plasma samples were analyzed by nuclear magnetic resonance spectroscopy and mass spectrometry coupled to one- or two-dimensional gas chromatography or liquid chromatography. Based on this multi-platform metabolomics approach, 427 plasma analytes were detected. Bi- and multivariate association analyses, adjusted for age and menopausal status, showed that CRF was linked to specific sets of metabolites primarily indicative of lipid metabolism. However, CRF-related metabolite patterns largely differed between sexes. While several phosphatidylcholines were linked to CRF in females, single lyso-phosphatidylcholines and sphingomyelins were associated with CRF in males. When controlling for further assessed clinical and phenotypical parameters, sex-specific CRF tended to be correlated with a smaller number of metabolites linked to lipid, amino acid, or xenobiotics-related metabolism. Interestingly, sex-specific CRF explanation models could be improved when including selected plasma analytes in addition to clinical and phenotypical variables. In summary, this study revealed sex-related differences in CRF-associated plasma metabolite patterns and proved known associations between CRF and risk factors for cardiometabolic diseases such as fat mass, visceral adipose tissue mass, or blood triglycerides in metabolically healthy individuals. Our findings indicate that covariates like sex and, especially, body composition have to be considered when studying blood metabolic markers related to CRF.
Collapse
Affiliation(s)
- Sina Kistner
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany; (M.D.); (R.K.); (M.J.R.); (B.M.); (K.R.); (A.B.)
| | - Maik Döring
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany; (M.D.); (R.K.); (M.J.R.); (B.M.); (K.R.); (A.B.)
| | - Ralf Krüger
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany; (M.D.); (R.K.); (M.J.R.); (B.M.); (K.R.); (A.B.)
| | - Manuela J. Rist
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany; (M.D.); (R.K.); (M.J.R.); (B.M.); (K.R.); (A.B.)
| | - Christoph H. Weinert
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, 76131 Karlsruhe, Germany; (C.H.W.); (D.B.)
| | - Diana Bunzel
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, 76131 Karlsruhe, Germany; (C.H.W.); (D.B.)
| | - Benedikt Merz
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany; (M.D.); (R.K.); (M.J.R.); (B.M.); (K.R.); (A.B.)
| | - Katrin Radloff
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany; (M.D.); (R.K.); (M.J.R.); (B.M.); (K.R.); (A.B.)
| | - Rainer Neumann
- Institute of Sports and Sports Science, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany; (R.N.); (S.H.)
| | - Sascha Härtel
- Institute of Sports and Sports Science, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany; (R.N.); (S.H.)
| | - Achim Bub
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany; (M.D.); (R.K.); (M.J.R.); (B.M.); (K.R.); (A.B.)
- Institute of Sports and Sports Science, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany; (R.N.); (S.H.)
| |
Collapse
|
7
|
Mercer KE, Maurer A, Pack LM, Ono-Moore K, Spray BJ, Campbell C, Chandler CJ, Burnett D, Souza E, Casazza G, Keim N, Newman J, Hunter G, Fernadez J, Garvey WT, Harper ME, Hoppel C, Adams SH, Thyfault J. Exercise training and diet-induced weight loss increase markers of hepatic bile acid (BA) synthesis and reduce serum total BA concentrations in obese women. Am J Physiol Endocrinol Metab 2021; 320:E864-E873. [PMID: 33645254 PMCID: PMC8238126 DOI: 10.1152/ajpendo.00644.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Regular exercise has profound metabolic influence on the liver, but effects on bile acid (BA) metabolism are less well known. BAs are synthesized exclusively in the liver from cholesterol via the rate-limiting enzyme cholesterol 7 alpha-hydroxylase (CYP7A1). BAs contribute to the solubilization and absorption of lipids and serve as important signaling molecules, capable of systemic endocrine function. Circulating BAs increase with obesity and insulin resistance, but effects following exercise and diet-induced weight loss are unknown. To test if improvements in fitness and weight loss as a result of exercise training enhance BA metabolism, we measured serum concentrations of total BAs (conjugated and unconjugated primary and secondary BAs) in sedentary, obese, insulin-resistant women (N = 11) before (PRE) and after (POST) a ∼14-wk exercise and diet-induced weight loss intervention. BAs were measured in serum collected after an overnight fast and during an oral glucose tolerance test (OGTT). Serum fibroblast growth factor 19 (FGF19; a regulator of BA synthesis) and 7-alpha-hydroxy-cholesten-3-one (C4, a marker of CYP7A1 enzymatic activity) also were measured. Using linear mixed-model analyses and the change in V̇O2peak (mL/min/kg) as a covariate, we observed that exercise and weight loss intervention decreased total fasting serum BA by ∼30% (P = 0.001) and increased fasting serum C4 concentrations by 55% (P = 0.004). C4 was significantly correlated with serum total BAs only in the POST condition, whereas serum FGF19 was unchanged. These data indicate that a fitness and weight loss intervention modifies BA metabolism in obese women and suggest that improved metabolic health associates with higher postabsorptive (fasting) BA synthesis. Furthermore, pre- vs. postintervention patterns of serum C4 following an OGTT support the hypothesis that responsiveness of BA synthesis to postprandial inhibition is improved after exercise and weight loss.NEW & NOTEWORTHY Exercise and weight loss in previously sedentary, insulin-resistant women facilitates a significant improvement in insulin sensitivity and fitness that may be linked to changes in bile acid metabolism. Diet-induced weight loss plus exercise-induced increases in fitness promote greater postabsorptive bile acid synthesis while also sensitizing the bile acid metabolic system to feedback inhibition during a glucose challenge when glucose and insulin are elevated.
Collapse
Affiliation(s)
- Kelly E Mercer
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Adrianna Maurer
- Departments of Molecular and Integrative Physiology and Internal Medicine, Kansas Medical Center, Kansas City, Kansas
| | - Lindsay M Pack
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
| | | | - Beverly J Spray
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Caitlin Campbell
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Carol J Chandler
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Dustin Burnett
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Elaine Souza
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Gretchen Casazza
- Sports Medicine Program, University of California, Davis School of Medicine, Sacramento, California
| | - Nancy Keim
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - John Newman
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Gary Hunter
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama
| | - Jose Fernadez
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Charles Hoppel
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Sean H Adams
- Department of Surgery, University of California, Davis School of Medicine, Sacramento, California
- Center for Alimentary and Metabolic Science, University of California, Davis School of Medicine, Sacramento, California
| | - John Thyfault
- Departments of Molecular and Integrative Physiology and Internal Medicine, Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
8
|
Grapov D, Fiehn O, Campbell C, Chandler CJ, Burnett DJ, Souza EC, Casazza GA, Keim NL, Hunter GR, Fernandez JR, Garvey WT, Hoppel CL, Harper M, Newman JW, Adams SH. Impact of a weight loss and fitness intervention on exercise-associated plasma oxylipin patterns in obese, insulin-resistant, sedentary women. Physiol Rep 2020; 8:e14547. [PMID: 32869956 PMCID: PMC7460071 DOI: 10.14814/phy2.14547] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022] Open
Abstract
Very little is known about how metabolic health status, insulin resistance or metabolic challenges modulate the endocannabinoid (eCB) or polyunsaturated fatty acid (PUFA)-derived oxylipin (OxL) lipid classes. To address these questions, plasma eCB and OxL concentrations were determined at rest, 10 and 20 min during an acute exercise bout (30 min total, ~45% of preintervention V̇O2peak , ~63 W), and following 20 min recovery in overnight-fasted sedentary, obese, insulin-resistant women under controlled diet conditions. We hypothesized that increased fitness and insulin sensitivity following a ~14-week training and weight loss intervention would lead to significant changes in lipid signatures using an identical acute exercise protocol to preintervention. In the first 10 min of exercise, concentrations of a suite of OxL diols and hydroxyeicosatetraenoic acid (HETE) metabolites dropped significantly. There was no increase in 12,13-DiHOME, previously reported to increase with exercise and proposed to activate muscle fatty acid uptake and tissue metabolism. Following weight loss intervention, exercise-associated reductions were more pronounced for several linoleate and alpha-linolenate metabolites including DiHOMEs, DiHODEs, KODEs, and EpODEs, and fasting concentrations of 9,10-DiHODE, 12,13-DiHODE, and 9,10-DiHOME were reduced. These findings suggest that improved metabolic health modifies soluble epoxide hydrolase, cytochrome P450 epoxygenase (CYP), and lipoxygenase (LOX) systems. Acute exercise led to reductions for most eCB metabolites, with no evidence for concentration increases even at recovery. It is proposed that during submaximal aerobic exercise, nonoxidative fates of long-chain saturated, monounsaturated, and PUFAs are attenuated in tissues that are important contributors to the blood OxL and eCB pools.
Collapse
Affiliation(s)
| | - Oliver Fiehn
- West Coast Metabolomics CenterUniversity of CaliforniaDavisCAUSA
| | - Caitlin Campbell
- United States Department of Agriculture‐Agricultural Research Service Western Human Nutrition Research CenterDavisCAUSA
| | - Carol J. Chandler
- United States Department of Agriculture‐Agricultural Research Service Western Human Nutrition Research CenterDavisCAUSA
| | - Dustin J. Burnett
- United States Department of Agriculture‐Agricultural Research Service Western Human Nutrition Research CenterDavisCAUSA
| | - Elaine C. Souza
- United States Department of Agriculture‐Agricultural Research Service Western Human Nutrition Research CenterDavisCAUSA
| | | | - Nancy L. Keim
- United States Department of Agriculture‐Agricultural Research Service Western Human Nutrition Research CenterDavisCAUSA
- Department of NutritionUniversity of CaliforniaDavisCAUSA
| | - Gary R. Hunter
- Department of Nutrition SciencesUniversity of AlabamaBirminghamALUSA
- Human Studies DepartmentUniversity of AlabamaBirminghamALUSA
| | - Jose R. Fernandez
- Department of Nutrition SciencesUniversity of AlabamaBirminghamALUSA
| | - W. Timothy Garvey
- Department of Nutrition SciencesUniversity of AlabamaBirminghamALUSA
| | - Charles L. Hoppel
- Pharmacology DepartmentCase Western Reserve UniversityClevelandOHUSA
| | - Mary‐Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems BiologyUniversity of OttawaOttawaONCanada
| | - John W. Newman
- United States Department of Agriculture‐Agricultural Research Service Western Human Nutrition Research CenterDavisCAUSA
- Department of NutritionUniversity of CaliforniaDavisCAUSA
| | - Sean H. Adams
- Arkansas Children’s Nutrition CenterLittle RockARUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
| |
Collapse
|
9
|
Mercer KE, Yeruva L, Pack L, Graham JL, Stanhope KL, Chintapalli SV, Wankhade UD, Shankar K, Havel PJ, Adams SH, Piccolo BD. Xenometabolite signatures in the UC Davis type 2 diabetes mellitus rat model revealed using a metabolomics platform enriched with microbe-derived metabolites. Am J Physiol Gastrointest Liver Physiol 2020; 319:G157-G169. [PMID: 32508155 PMCID: PMC7500265 DOI: 10.1152/ajpgi.00105.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gut microbiome has the potential to create or modify xenometabolites (i.e., nonhost-derived metabolites) through de novo synthesis or modification of exogenous and endogenous compounds. While there are isolated examples of xenometabolites influencing host health and disease, wide-scale characterization of these metabolites remains limited. We developed a metabolomics platform ("XenoScan") using liquid chromatography-mass spectrometry to characterize a range of known and suspected xenometabolites and their derivatives. This assay currently applies authentic standards for 190 molecules, enriched for metabolites of microbial origin. As a proof-of-principle, we characterized the cecal content xenometabolomics profile in adult male lean Sprague-Dawley (LSD) and University of California, Davis type 2 diabetes mellitus (UCD-T2DM) rats at different stages of diabetes. These results were correlated to specific bacterial species generated via shotgun metagenomic sequencing. UCD-T2DM rats had a unique xenometabolite profile compared with LSD rats, regardless of diabetes status, suggesting that at least some of the variation is associated with host genetics. Furthermore, modeling approaches revealed that several xenometabolites discriminated UCD-T2DM rats at early stages of diabetes versus those at 3 mo postdiabetes onset. Several xenometabolite hubs correlated with specific bacterial species in both LSD and UCD-T2DM rats. For example, indole-3-propionic acid negatively correlated with species within the Oscillibacter genus in UCD-T2DM rats considered to be prediabetic or recently diagnosed diabetic, in contrast to gluconic acid and trimethylamine, which were positively correlated with Oscillibacter species. The application of a xenometabolite-enriched metabolomics assay in relevant milieus will enable rapid identification of a wide variety of gut-derived metabolites, their derivatives, and their potential biochemical origins of xenometabolites in relationship to host gastrointestinal microbial ecology.NEW & NOTEWORTHY We debut a liquid chromatography-mass spectrometry (LC/MS) platform called the XenoScan, which is a metabolomics platform for xenometabolites (nonself-originating metabolites). This assay has 190 in-house standards with the majority enriched for microbe-derived metabolites. As a proof-of-principle, we used the XenoScan to discriminate genetic differences from cecal samples associated with different rat lineages, in addition to characterizing diabetes progression in rat model of type 2 diabetes. Complementing microbial sequencing data with xenometabolites uncovered novel microbial metabolism in targeted organisms.
Collapse
Affiliation(s)
- Kelly E. Mercer
- 1Arkansas Children’s Nutrition
Center, Little Rock, Arkansas,2Department of Pediatrics, University of
Arkansas for Medical Sciences, Little Rock,
Arkansas
| | - Laxmi Yeruva
- 1Arkansas Children’s Nutrition
Center, Little Rock, Arkansas,2Department of Pediatrics, University of
Arkansas for Medical Sciences, Little Rock,
Arkansas,3Arkansas Children’s Research
Institute, Little Rock, Arkansas
| | - Lindsay Pack
- 1Arkansas Children’s Nutrition
Center, Little Rock, Arkansas
| | - James L. Graham
- 4Department of Nutrition, University of
California of California, Davis,
California,5Department of Molecular Biosciences, School of Veterinary
Medicine, University of California, Davis,
California
| | - Kimber L. Stanhope
- 4Department of Nutrition, University of
California of California, Davis,
California,5Department of Molecular Biosciences, School of Veterinary
Medicine, University of California, Davis,
California
| | - Sree V. Chintapalli
- 1Arkansas Children’s Nutrition
Center, Little Rock, Arkansas,2Department of Pediatrics, University of
Arkansas for Medical Sciences, Little Rock,
Arkansas
| | - Umesh D. Wankhade
- 1Arkansas Children’s Nutrition
Center, Little Rock, Arkansas,2Department of Pediatrics, University of
Arkansas for Medical Sciences, Little Rock,
Arkansas
| | - Kartik Shankar
- 6Department of Pediatrics, University of
Colorado Anschutz Medical Campus, Aurora,
Colorado
| | - Peter J. Havel
- 4Department of Nutrition, University of
California of California, Davis,
California,5Department of Molecular Biosciences, School of Veterinary
Medicine, University of California, Davis,
California
| | - Sean H. Adams
- 1Arkansas Children’s Nutrition
Center, Little Rock, Arkansas,2Department of Pediatrics, University of
Arkansas for Medical Sciences, Little Rock,
Arkansas
| | - Brian D. Piccolo
- 1Arkansas Children’s Nutrition
Center, Little Rock, Arkansas,2Department of Pediatrics, University of
Arkansas for Medical Sciences, Little Rock,
Arkansas
| |
Collapse
|
10
|
Galvin VC, Yang ST, Paspalas CD, Yang Y, Jin LE, Datta D, Morozov YM, Lightbourne TC, Lowet AS, Rakic P, Arnsten AFT, Wang M. Muscarinic M1 Receptors Modulate Working Memory Performance and Activity via KCNQ Potassium Channels in the Primate Prefrontal Cortex. Neuron 2020; 106:649-661.e4. [PMID: 32197063 DOI: 10.1016/j.neuron.2020.02.030] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/21/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023]
Abstract
Working memory relies on the dorsolateral prefrontal cortex (dlPFC), where microcircuits of pyramidal neurons enable persistent firing in the absence of sensory input, maintaining information through recurrent excitation. This activity relies on acetylcholine, although the molecular mechanisms for this dependence are not thoroughly understood. This study investigated the role of muscarinic M1 receptors (M1Rs) in the dlPFC using iontophoresis coupled with single-unit recordings from aging monkeys with naturally occurring cholinergic depletion. We found that M1R stimulation produced an inverted-U dose response on cell firing and behavioral performance when given systemically to aged monkeys. Immunoelectron microscopy localized KCNQ isoforms (Kv7.2, Kv7.3, and Kv7.5) on layer III dendrites and spines, similar to M1Rs. Iontophoretic manipulation of KCNQ channels altered cell firing and reversed the effects of M1R compounds, suggesting that KCNQ channels are one mechanism for M1R actions in the dlPFC. These results indicate that M1Rs may be an appropriate target to treat cognitive disorders with cholinergic alterations.
Collapse
Affiliation(s)
- Veronica C Galvin
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sheng Tao Yang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | - Yang Yang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lu E Jin
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yury M Morozov
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Taber C Lightbourne
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Adam S Lowet
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Pasko Rakic
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
11
|
Grapov D, Fiehn O, Campbell C, Chandler CJ, Burnett DJ, Souza EC, Casazza GA, Keim NL, Newman JW, Hunter GR, Fernandez JR, Garvey WT, Hoppel CL, Harper ME, Adams SH. Exercise plasma metabolomics and xenometabolomics in obese, sedentary, insulin-resistant women: impact of a fitness and weight loss intervention. Am J Physiol Endocrinol Metab 2019; 317:E999-E1014. [PMID: 31526287 PMCID: PMC6962502 DOI: 10.1152/ajpendo.00091.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Insulin resistance has wide-ranging effects on metabolism, but there are knowledge gaps regarding the tissue origins of systemic metabolite patterns and how patterns are altered by fitness and metabolic health. To address these questions, plasma metabolite patterns were determined every 5 min during exercise (30 min, ∼45% of V̇o2peak, ∼63 W) and recovery in overnight-fasted sedentary, obese, insulin-resistant women under controlled conditions of diet and physical activity. We hypothesized that improved fitness and insulin sensitivity following a ∼14-wk training and weight loss intervention would lead to fixed workload plasma metabolomics signatures reflective of metabolic health and muscle metabolism. Pattern analysis over the first 15 min of exercise, regardless of pre- versus postintervention status, highlighted anticipated increases in fatty acid tissue uptake and oxidation (e.g., reduced long-chain fatty acids), diminution of nonoxidative fates of glucose [e.g., lowered sorbitol-pathway metabolites and glycerol-3-galactoside (possible glycerolipid synthesis metabolite)], and enhanced tissue amino acid use (e.g., drops in amino acids; modest increase in urea). A novel observation was that exercise significantly increased several xenometabolites ("non-self" molecules, from microbes or foods), including benzoic acid-salicylic acid-salicylaldehyde, hexadecanol-octadecanol-dodecanol, and chlorogenic acid. In addition, many nonannotated metabolites changed with exercise. Although exercise itself strongly impacted the global metabolome, there were surprisingly few intervention-associated differences despite marked improvements in insulin sensitivity, fitness, and adiposity. These results and previously reported plasma acylcarnitine profiles support the principle that most metabolic changes during submaximal aerobic exercise are closely tethered to absolute ATP turnover rate (workload), regardless of fitness or metabolic health status.
Collapse
Affiliation(s)
| | - Oliver Fiehn
- West Coast Metabolomics Center, Genome Center, University of California, Davis, California
| | - Caitlin Campbell
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Carol J Chandler
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Dustin J Burnett
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Elaine C Souza
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Gretchen A Casazza
- Sports Medicine Program, School of Medicine, University of California, Davis, California
| | - Nancy L Keim
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
- Department of Nutrition, University of California, Davis, California
| | - John W Newman
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
- Department of Nutrition, University of California, Davis, California
| | - Gary R Hunter
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama
- Human Studies Department, University of Alabama, Birmingham, Alabama
| | - Jose R Fernandez
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama
| | - Charles L Hoppel
- Pharmacology Department, Case Western Reserve University, Cleveland, Ohio
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
12
|
Ribot J, Arreguín A, Kuda O, Kopecky J, Palou A, Bonet ML. Novel Markers of the Metabolic Impact of Exogenous Retinoic Acid with A Focus on Acylcarnitines and Amino Acids. Int J Mol Sci 2019; 20:E3640. [PMID: 31349613 PMCID: PMC6696161 DOI: 10.3390/ijms20153640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/21/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023] Open
Abstract
Treatment with all-trans retinoic acid (ATRA), the carboxylic form of vitamin A, lowers body weight in rodents by promoting oxidative metabolism in multiple tissues including white and brown adipose tissues. We aimed to identify novel markers of the metabolic impact of ATRA through targeted blood metabolomics analyses, with a focus on acylcarnitines and amino acids. Blood was obtained from mice treated with a high ATRA dose (50 mg/kg body weight/day, subcutaneous injection) or placebo (controls) during the 4 days preceding collection. LC-MS/MS analyses with a focus on acylcarnitines and amino acids were conducted on plasma and PBMC. Main results showed that, relative to controls, ATRA-treated mice had in plasma: increased levels of carnitine, acetylcarnitine, and longer acylcarnitine species; decreased levels of citrulline, and increased global arginine bioavailability ratio for nitric oxide synthesis; increased levels of creatine, taurine and docosahexaenoic acid; and a decreased n-6/n-3 polyunsaturated fatty acids ratio. While some of these features likely reflect the stimulation of lipid mobilization and oxidation promoted by ATRA treatment systemically, other may also play a causal role underlying ATRA actions. The results connect ATRA to specific nutrition-modulated biochemical pathways, and suggest novel mechanisms of action of vitamin A-derived retinoic acid on metabolic health.
Collapse
Affiliation(s)
- Joan Ribot
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain.
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain.
| | - Andrea Arreguín
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
| | - Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Jan Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Andreu Palou
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Maria Luisa Bonet
- Grup de Recerca Nutrigenòmica i Obesitat, Laboratori de Biologia Molecular, Nutrició i Biotecnologia (LBNB), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| |
Collapse
|
13
|
Liu R, Zhao J, Guo J, Liu X, Yu J, Wang H, Li Y, Sun C, Liu L. Postprandial metabolomics: GC-MS analysis reveals differences in organic acid profiles of impaired fasting glucose individuals in response to highland barley loads. Food Funct 2019; 10:1552-1562. [DOI: 10.1039/c8fo02321b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The HB load producing low postprandial glucose and insulin responses brings about several alterations in organic acids.
Collapse
Affiliation(s)
- Rui Liu
- National Key Discipline Laboratory
- Department of Nutrition and Food Hygiene
- School of Public Health
- Harbin Medical University
- Harbin
| | - Jinhui Zhao
- National Key Discipline Laboratory
- Department of Nutrition and Food Hygiene
- School of Public Health
- Harbin Medical University
- Harbin
| | - Jing Guo
- National Key Discipline Laboratory
- Department of Nutrition and Food Hygiene
- School of Public Health
- Harbin Medical University
- Harbin
| | - Xiaowei Liu
- National Key Discipline Laboratory
- Department of Nutrition and Food Hygiene
- School of Public Health
- Harbin Medical University
- Harbin
| | - Jiaying Yu
- National Key Discipline Laboratory
- Department of Nutrition and Food Hygiene
- School of Public Health
- Harbin Medical University
- Harbin
| | - Hanming Wang
- Department of Infectious Diseases
- Harbin Children's Hospital
- Harbin
- P. R. China
| | - Ying Li
- National Key Discipline Laboratory
- Department of Nutrition and Food Hygiene
- School of Public Health
- Harbin Medical University
- Harbin
| | - Changhao Sun
- National Key Discipline Laboratory
- Department of Nutrition and Food Hygiene
- School of Public Health
- Harbin Medical University
- Harbin
| | - Liyan Liu
- National Key Discipline Laboratory
- Department of Nutrition and Food Hygiene
- School of Public Health
- Harbin Medical University
- Harbin
| |
Collapse
|
14
|
Krishnan S, Adams SH, Allen LH, Laugero KD, Newman JW, Stephensen CB, Burnett DJ, Witbracht M, Welch LC, Que ES, Keim NL. A randomized controlled-feeding trial based on the Dietary Guidelines for Americans on cardiometabolic health indexes. Am J Clin Nutr 2018; 108:266-278. [PMID: 30101333 DOI: 10.1093/ajcn/nqy113] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 05/04/2018] [Indexed: 12/13/2022] Open
Abstract
Background The 2010 Dietary Guidelines for Americans (DGA) recommend nutrient needs be met by increasing fruit, vegetable, and whole-grain intake with the use of low-fat or fat-free dairy products and by reducing sodium, solid fats, and added sugars. However, the DGA, as a dietary pattern, have not been tested in an intervention trial. Objective The aim of this study was to evaluate the impact of a DGA-based diet compared with a representative typical American diet (TAD) on glucose homeostasis and fasting lipids in individuals at risk of cardiometabolic disease. Design A randomized, double-blind, controlled 8-wk intervention was conducted in overweight and obese women selected according to indexes of insulin resistance or dyslipidemia. Women were randomly assigned to the DGA or TAD group (n = 28 DGA and 24 TAD). The TAD diet was based on average adult intake from the NHANES 2009-2010. The DGA and TAD diets had respective Healthy Eating Index scores of 98 and 62. All foods and beverages were provided during the intervention. Oral-glucose tolerance and fasting lipids were evaluated at 0, 2, and 8 wk of the intervention. Insulin resistance and sensitivity were estimated with the use of surrogates (e.g., homeostasis model assessment of insulin resistance). Results By design, volunteers maintained their weight during the intervention. Fasting insulin, glucose, triglycerides, oral-glucose tolerance, and indexes of insulin resistance were not affected by either of the diets. Systolic blood pressure decreased in the DGA group (∼-9 mm Hg; P < 0.05). Total and HDL cholesterol also decreased in both groups (P < 0.05). Exploratory analysis comparing volunteers entering the study with insulin resistance and dyslipidemia with those with only dyslipidemia did not show an effect of pre-existing conditions on glucose tolerance or fasting lipid outcomes. Conclusions The consumption of a DGA dietary pattern for 8 wk without weight loss reduced systolic blood pressure. There were no differences between the DGA and TAD diets in fasting insulin, glucose, indexes of insulin resistance, or fasting lipids. This trial was registered at www.clinicaltrials.gov as NCT02298725.
Collapse
Affiliation(s)
- Sridevi Krishnan
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| | - Sean H Adams
- Arkansas Children's Nutrition Center.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Lindsay H Allen
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| | - Kevin D Laugero
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| | - John W Newman
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| | - Charles B Stephensen
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| | - Dustin J Burnett
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| | - Megan Witbracht
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| | - Lucas C Welch
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| | - Excel S Que
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA
| | - Nancy L Keim
- Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA.,Department of Nutrition, University of California-Davis, Davis, CA
| |
Collapse
|
15
|
Nowak C, Hetty S, Salihovic S, Castillejo-Lopez C, Ganna A, Cook NL, Broeckling CD, Prenni JE, Shen X, Giedraitis V, Ärnlöv J, Lind L, Berne C, Sundström J, Fall T, Ingelsson E. Glucose challenge metabolomics implicates medium-chain acylcarnitines in insulin resistance. Sci Rep 2018; 8:8691. [PMID: 29875472 PMCID: PMC5989236 DOI: 10.1038/s41598-018-26701-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/17/2018] [Indexed: 12/27/2022] Open
Abstract
Insulin resistance (IR) predisposes to type 2 diabetes and cardiovascular disease but its causes are incompletely understood. Metabolic challenges like the oral glucose tolerance test (OGTT) can reveal pathogenic mechanisms. We aimed to discover associations of IR with metabolite trajectories during OGTT. In 470 non-diabetic men (age 70.6 ± 0.6 years), plasma samples obtained at 0, 30 and 120 minutes during an OGTT were analyzed by untargeted liquid chromatography-mass spectrometry metabolomics. IR was assessed with the hyperinsulinemic-euglycemic clamp method. We applied age-adjusted linear regression to identify metabolites whose concentration change was related to IR. Nine trajectories, including monounsaturated fatty acids, lysophosphatidylethanolamines and a bile acid, were significantly associated with IR, with the strongest associations observed for medium-chain acylcarnitines C10 and C12, and no associations with L-carnitine or C2-, C8-, C14- or C16-carnitine. Concentrations of C10- and C12-carnitine decreased during OGTT with a blunted decline in participants with worse insulin resistance. Associations persisted after adjustment for obesity, fasting insulin and fasting glucose. In mouse 3T3-L1 adipocytes exposed to different acylcarnitines, we observed blunted insulin-stimulated glucose uptake after treatment with C10- or C12-carnitine. In conclusion, our results identify medium-chain acylcarnitines as possible contributors to IR.
Collapse
Affiliation(s)
- Christoph Nowak
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Susanne Hetty
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Samira Salihovic
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Casimiro Castillejo-Lopez
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andrea Ganna
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, United States of America
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Naomi L Cook
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Corey D Broeckling
- Proteomics and Metabolomics Facility, Colorado State University, Fort Collins, CO, United States of America
| | - Jessica E Prenni
- Proteomics and Metabolomics Facility, Colorado State University, Fort Collins, CO, United States of America
| | - Xia Shen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- School of Health and Social Studies, Dalarna University, Falun, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Christian Berne
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, Uppsala, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States of America.
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
16
|
Calder PC, Carding SR, Christopher G, Kuh D, Langley-Evans SC, McNulty H. A holistic approach to healthy ageing: how can people live longer, healthier lives? J Hum Nutr Diet 2018; 31:439-450. [PMID: 29862589 DOI: 10.1111/jhn.12566] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Although lifespan is increasing, there is no evidence to suggest that older people are experiencing better health in their later years than previous generations. Nutrition, at all stages of life, plays an important role in determining health and wellbeing. METHODS A roundtable meeting of UK experts on nutrition and ageing considered key aspects of the diet-ageing relationship and developed a consensus position on the main priorities for research and public health actions that are required to help people live healthier lives as they age. RESULTS The group consensus highlighted the requirement for a life course approach, recognising the multifactorial nature of the impact of ageing. Environmental and lifestyle influences at any life stage are modified by genetic factors and early development. The response to the environment at each stage of life can determine the impact of lifestyle later on. There are no key factors that act in isolation to determine patterns of ageing and it is a combination of environmental and social factors that drives healthy or unhealthy ageing. Too little is known about how contemporary dietary patterns and sedentary lifestyles will impact upon healthy ageing in future generations and this is a priority for future research. CONCLUSIONS There is good evidence to support change to lifestyle (i.e. diet, nutrition and physical) activity in relation to maintaining or improving body composition, cognitive health and emotional intelligence, immune function and vascular health. Lifestyle change at any stage of life may extend healthy lifespan, although the impact of early changes appears to be greatest.
Collapse
Affiliation(s)
- P C Calder
- Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - S R Carding
- Quadram Institute Bioscience and Norwich Medical School, University of East Anglia, Norwich, UK
| | - G Christopher
- Faculty of Health and Applied Sciences, University of the West of England, Bristol, UK
| | - D Kuh
- Medical Research Council Unit for Lifelong Health and Ageing, University College London, London, UK
| | - S C Langley-Evans
- School of Biosciences, University of Nottingham, Sutton Bonnington, UK
| | - H McNulty
- Nutrition Innovation Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, UK
| |
Collapse
|
17
|
Stefanaki C, Peppa M, Mastorakos G, Chrousos GP. Examining the gut bacteriome, virome, and mycobiome in glucose metabolism disorders: Are we on the right track? Metabolism 2017; 73:52-66. [PMID: 28732571 DOI: 10.1016/j.metabol.2017.04.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 04/21/2017] [Accepted: 04/28/2017] [Indexed: 12/19/2022]
Abstract
Human gut microbiome is defined as the gene complement of the gut microbial community, measured via laboratory metagenomic techniques. It includes bacteriome, virome and mycobiome, which represent, respectively, the assemblages of bacteria, viruses and fungi, living in the human gut. Gut microbiota function as a living "organ" that interacts with the gastro-intestinal environment, provides nutrients and vitamins to the organism and transduces hormonal messages, essentially influencing the main metabolic pathways, including drug metabolism. A clear association between gut, and glucose metabolism disorders has recently emerged. Medications acting on glucose absorption in the gut, or enhancing gut hormone activity are already extensively employed in the therapy of diabetes. Moreover, the gut is characterized by immune, and autonomous neuronal features, which play a critical role in maintaining glucose metabolism homeostasis. Gut microbes respond to neuroendocrine, and immune biochemical messages, affecting the health, and behavior of the host. There is vast heterogeneity in the studies included in this review, hence a meta-analysis, or a systematic review were not applicable. In this article, we attempt to reveal the interplay between human gut microbiota physiology, and hyperglycemic states, synthesizing, and interpreting findings from human studies.
Collapse
Affiliation(s)
- Charikleia Stefanaki
- 1st Department of Pediatrics, Choremeio Research Laboratory, Athens University Medical School, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| | - Melpomeni Peppa
- Endocrine Unit, 2nd Department of Internal Medicine Propaedeutic, Research Institute and Diabetes Center, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - George Mastorakos
- Department of Endocrinology, Metabolism and Diabetes, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - George P Chrousos
- 1st Department of Pediatrics, Choremeio Research Laboratory, Athens University Medical School, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| |
Collapse
|
18
|
Pantophlet AJ, Wopereis S, Eelderink C, Vonk RJ, Stroeve JH, Bijlsma S, van Stee L, Bobeldijk I, Priebe MG. Metabolic Profiling Reveals Differences in Plasma Concentrations of Arabinose and Xylose after Consumption of Fiber-Rich Pasta and Wheat Bread with Differential Rates of Systemic Appearance of Exogenous Glucose in Healthy Men. J Nutr 2017; 147:152-160. [PMID: 27927976 DOI: 10.3945/jn.116.237404] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/01/2016] [Accepted: 11/01/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The consumption of products rich in cereal fiber and with a low glycemic index is implicated in a lower risk of metabolic diseases. Previously, we showed that the consumption of fiber-rich pasta compared with bread resulted in a lower rate of appearance of exogenous glucose and a lower glucose clearance rate quantified with a dual-isotope technique, which was in accordance with a lower insulin and glucose-dependent insulinotropic polypeptide response. OBJECTIVE To gain more insight into the acute metabolic consequences of the consumption of products resulting in differential glucose kinetics, postprandial metabolic profiles were determined. METHODS In a crossover study, 9 healthy men [mean ± SEM age: 21 ± 0.5 y; mean ± SEM body mass index (kg/m2): 22 ± 0.5] consumed wheat bread (132 g) and fresh pasta (119 g uncooked) enriched with wheat bran (10%) meals. A total of 134 different metabolites in postprandial plasma samples (at -5, 30, 60, 90, 120, and 180 min) were quantified by using a gas chromatography-mass spectrometry-based metabolomics approach (secondary outcomes). Two-factor ANOVA and advanced multivariate statistical analysis (partial least squares) were applied to detect differences between both food products. RESULTS Forty-two different postprandial metabolite profiles were identified, primarily representing pathways related to protein and energy metabolism, which were on average 8% and 7% lower after the men consumed pasta rather than bread, whereas concentrations of arabinose and xylose were 58% and 53% higher, respectively. Arabinose and xylose are derived from arabinoxylans, which are important components of wheat bran. The higher bioavailability of arabinose and xylose after pasta intake coincided with a lower rate of appearance of glucose and amino acids. We speculate that this higher bioavailability is due to higher degradation of arabinoxylans by small intestinal microbiota, facilitated by the higher viscosity of arabinoxylans after pasta intake than after bread intake. CONCLUSION This study suggests that wheat bran, depending on the method of processing, can increase the viscosity of the meal bolus in the small intestine and interfere with macronutrient absorption in healthy men, thereby influencing postprandial glucose and insulin responses. This trial was registered at www.controlled-trials.com as ISRCTN42106325.
Collapse
Affiliation(s)
- Andre J Pantophlet
- Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, and
| | - Suzan Wopereis
- Netherlands Organization for Applied Scientific Research (TNO), Food and Nutrition, Zeist, Netherlands
| | - Coby Eelderink
- Center for Medical Biomics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands; and
| | - Roel J Vonk
- Center for Medical Biomics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands; and
| | - Johanna H Stroeve
- Netherlands Organization for Applied Scientific Research (TNO), Food and Nutrition, Zeist, Netherlands
| | - Sabina Bijlsma
- Netherlands Organization for Applied Scientific Research (TNO), Food and Nutrition, Zeist, Netherlands
| | - Leo van Stee
- Netherlands Organization for Applied Scientific Research (TNO), Food and Nutrition, Zeist, Netherlands
| | - Ivana Bobeldijk
- Netherlands Organization for Applied Scientific Research (TNO), Food and Nutrition, Zeist, Netherlands
| | - Marion G Priebe
- Center for Medical Biomics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands; and
| |
Collapse
|
19
|
Siopi A, Deda O, Manou V, Kellis S, Kosmidis I, Komninou D, Raikos N, Christoulas K, Theodoridis GA, Mougios V. Effects of Different Exercise Modes on the Urinary Metabolic Fingerprint of Men with and without Metabolic Syndrome. Metabolites 2017; 7:metabo7010005. [PMID: 28134772 PMCID: PMC5372208 DOI: 10.3390/metabo7010005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 01/20/2017] [Accepted: 01/24/2017] [Indexed: 01/21/2023] Open
Abstract
Exercise is important in the prevention and treatment of the metabolic syndrome (MetS), a cluster of risk factors that raises morbidity. Metabolomics can facilitate the optimization of exercise prescription. This study aimed to investigate whether the response of the human urinary metabolic fingerprint to exercise depends on the presence of MetS or exercise mode. Twenty-three sedentary men (MetS, n = 9, and Healthy, n = 14) completed four trials: resting, high-intensity interval exercise (HIIE), continuous moderate-intensity exercise (CME), and resistance exercise (RE). Urine samples were collected pre-exercise and at 2, 4, and 24 h for targeted analysis by liquid chromatography-mass spectrometry. Time exerted the strongest differentiating effect, followed by exercise mode and health status. The greatest changes were observed in the first post-exercise samples, with a gradual return to baseline at 24 h. RE caused the greatest responses overall, followed by HIIE, while CME had minimal effect. The metabolic fingerprints of the two groups were separated at 2 h, after HIIE and RE; and at 4 h, after HIIE, with evidence of blunted response to exercise in MetS. Our findings show diverse responses of the urinary metabolic fingerprint to different exercise modes in men with and without metabolic syndrome.
Collapse
Affiliation(s)
- Aikaterina Siopi
- School of Physical Education and Sport Science at Thessaloniki, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Olga Deda
- School of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Vasiliki Manou
- School of Physical Education and Sport Science at Thessaloniki, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Spyros Kellis
- School of Physical Education and Sport Science at Thessaloniki, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Ioannis Kosmidis
- School of Physical Education and Sport Science at Thessaloniki, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Despina Komninou
- Department of Nutrition and Dietetics, Alexander Technological Educational Institute of Thessaloniki, 57400 Thessaloniki, Greece.
| | - Nikolaos Raikos
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Kosmas Christoulas
- School of Physical Education and Sport Science at Thessaloniki, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | | | - Vassilis Mougios
- School of Physical Education and Sport Science at Thessaloniki, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| |
Collapse
|
20
|
Geidenstam N, Magnusson M, Danielsson APH, Gerszten RE, Wang TJ, Reinius LE, Mulder H, Melander O, Ridderstråle M. Amino Acid Signatures to Evaluate the Beneficial Effects of Weight Loss. Int J Endocrinol 2017; 2017:6490473. [PMID: 28484491 PMCID: PMC5412138 DOI: 10.1155/2017/6490473] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/10/2017] [Accepted: 02/27/2017] [Indexed: 11/17/2022] Open
Abstract
Aims. We investigated the relationship between circulating amino acid levels and obesity; to what extent weight loss followed by weight maintenance can correct amino acid abnormalities; and whether amino acids are related to weight loss. Methods. Amino acids associated with waist circumference (WC) and BMI were studied in 804 participants from the Malmö Diet and Cancer Cardiovascular Cohort (MDC-CC). Changes in amino acid levels were analyzed after weight loss and weight maintenance in 12 obese subjects and evaluated in a replication cohort (n = 83). Results. Out of the eight identified BMI-associated amino acids from the MDC-CC, alanine, isoleucine, tyrosine, phenylalanine, and glutamate decreased after weight loss, while asparagine increased after weight maintenance. These changes were validated in the replication cohort. Scores that were constructed based on obesity-associated amino acids and known risk factors decreased in the ≥10% weight loss group with an associated change in BMI (R2 = 0.16-0.22, p < 0.002), whereas the scores increased in the <10% weight loss group (p < 0.0004). Conclusions. Weight loss followed by weight maintenance leads to differential changes in amino acid levels associated with obesity. Treatment modifiable scores based on epidemiological and interventional data may be used to evaluate the potential metabolic benefit of weight loss.
Collapse
Affiliation(s)
- Nina Geidenstam
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
- *Nina Geidenstam:
| | - Martin Magnusson
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Anders P. H. Danielsson
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Robert E. Gerszten
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas J. Wang
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lovisa E. Reinius
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hindrik Mulder
- Department of Clinical Sciences Malmö, Molecular Metabolism, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
- Center of Emergency Medicine, Skåne University Hospital, Malmö, Sweden
| | - Martin Ridderstråle
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
- Steno Diabetes Center A/S, Gentofte, Denmark
| |
Collapse
|
21
|
Zhang J, Light AR, Hoppel CL, Campbell C, Chandler CJ, Burnett DJ, Souza EC, Casazza GA, Hughen RW, Keim NL, Newman JW, Hunter GR, Fernandez JR, Garvey WT, Harper ME, Fiehn O, Adams SH. Acylcarnitines as markers of exercise-associated fuel partitioning, xenometabolism, and potential signals to muscle afferent neurons. Exp Physiol 2016; 102:48-69. [PMID: 27730694 DOI: 10.1113/ep086019] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 10/07/2016] [Indexed: 01/18/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does improved metabolic health and insulin sensitivity following a weight-loss and fitness intervention in sedentary, obese women alter exercise-associated fuel metabolism and incomplete mitochondrial fatty acid oxidation (FAO), as tracked by blood acylcarnitine patterns? What is the main finding and its importance? Despite improved fitness and blood sugar control, indices of incomplete mitochondrial FAO increased in a similar manner in response to a fixed load acute exercise bout; this indicates that intramitochondrial muscle FAO is inherently inefficient and is tethered directly to ATP turnover. With insulin resistance or type 2 diabetes mellitus, mismatches between mitochondrial fatty acid fuel delivery and oxidative phosphorylation/tricarboxylic acid cycle activity may contribute to inordinate accumulation of short- or medium-chain acylcarnitine fatty acid derivatives [markers of incomplete long-chain fatty acid oxidation (FAO)]. We reasoned that incomplete FAO in muscle would be ameliorated concurrent with improved insulin sensitivity and fitness following a ∼14 week training and weight-loss intervention in obese, sedentary, insulin-resistant women. Contrary to this hypothesis, overnight-fasted and exercise-induced plasma C4-C14 acylcarnitines did not differ between pre- and postintervention phases. These metabolites all increased robustly with exercise (∼45% of pre-intervention peak oxygen consumption) and decreased during a 20 min cool-down. This supports the idea that, regardless of insulin sensitivity and fitness, intramitochondrial muscle β-oxidation and attendant incomplete FAO are closely tethered to absolute ATP turnover rate. Acute exercise also led to branched-chain amino acid acylcarnitine derivative patterns suggestive of rapid and transient diminution of branched-chain amino acid flux through the mitochondrial branched-chain ketoacid dehydrogenase complex. We confirmed our prior novel observation that a weight-loss/fitness intervention alters plasma xenometabolites [i.e. cis-3,4-methylene-heptanoylcarnitine and γ-butyrobetaine (a co-metabolite possibly derived in part from gut bacteria)], suggesting that host metabolic health regulated gut microbe metabolism. Finally, we considered whether acylcarnitine metabolites signal to muscle-innervating afferents; palmitoylcarnitine at concentrations as low as 1-10 μm activated a subset (∼2.5-5%) of these neurons ex vivo. This supports the hypothesis that in addition to tracking exercise-associated shifts in fuel metabolism, muscle acylcarnitines act as signals of exertion to short-loop somatosensory-motor circuits or to the brain.
Collapse
Affiliation(s)
- Jie Zhang
- Anesthesiology Department, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Alan R Light
- Anesthesiology Department, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Charles L Hoppel
- Pharmacology Department, Case Western Reserve University, Cleveland, OH, USA
| | - Caitlin Campbell
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA
| | - Carol J Chandler
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA
| | - Dustin J Burnett
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA
| | - Elaine C Souza
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA
| | - Gretchen A Casazza
- Sports Medicine Program, School of Medicine, University of California, Davis, CA, USA
| | - Ronald W Hughen
- Anesthesiology Department, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Nancy L Keim
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA.,Department of Nutrition, University of California, Davis, CA, USA
| | - John W Newman
- United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA.,Department of Nutrition, University of California, Davis, CA, USA
| | - Gary R Hunter
- Department of Nutrition Sciences, University of Alabama, Birmingham, AL, USA.,Human Studies Department, University of Alabama, Birmingham, AL, USA
| | - Jose R Fernandez
- Department of Nutrition Sciences, University of Alabama, Birmingham, AL, USA
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama, Birmingham, AL, USA
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Oliver Fiehn
- Genome Center and West Coast Metabolomics Center, University of California, Davis, CA, USA.,Biochemistry Department, King Abdulaziz University, Jeddah, Saudi-Arabia
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
22
|
Tulipani S, Griffin J, Palau-Rodriguez M, Mora-Cubillos X, Bernal-Lopez RM, Tinahones FJ, Corkey BE, Andres-Lacueva C. Metabolomics-guided insights on bariatric surgery versus behavioral interventions for weight loss. Obesity (Silver Spring) 2016; 24:2451-2466. [PMID: 27891833 DOI: 10.1002/oby.21686] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 08/30/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To review the metabolomic studies carried out so far to identify metabolic markers associated with surgical and dietary treatments for weight loss in subjects with obesity. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. RESULTS Thirty-two studies successfully met the eligibility criteria. The metabolic adaptations shared by surgical and dietary interventions mirrored a state of starvation ketoacidosis (increase of circulating ketone bodies), an increase of acylcarnitines and fatty acid β-oxidation, a decrease of specific amino acids including branched-chain amino acids (BCAA) and (lyso)glycerophospholipids previously associated with obesity, and adipose tissue expansion. The metabolic footprint of bariatric procedures was specifically characterized by an increase of bile acid circulating pools and a decrease of ceramide levels, a greater perioperative decline in BCAA, and the rise of circulating serine and glycine, mirroring glycemic control and inflammation improvement. In one study, 3-hydroxybutyrate was particularly identified as an early metabolic marker of long-term prognosis after surgery and proposed to increase current prognostic modalities and contribute to personalized treatment. CONCLUSIONS Metabolomics helped in deciphering the metabolic response to weight loss treatments. Moving from association to causation is the next challenge to move to a further level of clinical application.
Collapse
Affiliation(s)
- Sara Tulipani
- Department of Nutrition, Food Sciences and Gastronomy, Biomarkers & Nutrimetabolomic Lab, XaRTA, INSA, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga, Malaga, Spain
| | - Jules Griffin
- MRC Human Nutrition Research, Elsie Widdowson Laboratory, Cambridge, UK
- Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Magali Palau-Rodriguez
- Department of Nutrition, Food Sciences and Gastronomy, Biomarkers & Nutrimetabolomic Lab, XaRTA, INSA, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Ximena Mora-Cubillos
- Department of Nutrition, Food Sciences and Gastronomy, Biomarkers & Nutrimetabolomic Lab, XaRTA, INSA, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Rosa M Bernal-Lopez
- Biomedical Research Institute (IBIMA), Service of Internal Medicine, Malaga Hospital Complex (Hospital Regional Universitario de Malaga), University of Malaga, Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Francisco J Tinahones
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga, Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Barbara E Corkey
- School of Medicine, Obesity Research Center, Boston University, Boston, Massachusetts, USA
| | - Cristina Andres-Lacueva
- Department of Nutrition, Food Sciences and Gastronomy, Biomarkers & Nutrimetabolomic Lab, XaRTA, INSA, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| |
Collapse
|
23
|
Fecal Microbiota and Metabolome in a Mouse Model of Spontaneous Chronic Colitis: Relevance to Human Inflammatory Bowel Disease. Inflamm Bowel Dis 2016; 22:2767-2787. [PMID: 27824648 DOI: 10.1097/mib.0000000000000970] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dysbiosis of the gut microbiota may be involved in the pathogenesis of inflammatory bowel disease (IBD). However, the mechanisms underlying the role of the intestinal microbiome and metabolome in IBD onset and its alteration during active treatment and recovery remain unknown. Animal models of chronic intestinal inflammation with similar microbial and metabolomic profiles would enable investigation of these mechanisms and development of more effective treatments. Recently, the Winnie mouse model of colitis closely representing the clinical symptoms and characteristics of human IBD has been developed. In this study, we have analyzed fecal microbial and metabolomic profiles in Winnie mice and discussed their relevance to human IBD. METHODS The 16S rRNA gene was sequenced from fecal DNA of Winnie and C57BL/6 mice to define operational taxonomic units at ≥97% similarity threshold. Metabolomic profiling of the same fecal samples was performed by gas chromatography-mass spectrometry. RESULTS Composition of the dominant microbiota was disturbed, and prominent differences were evident at all levels of the intestinal microbiome in fecal samples from Winnie mice, similar to observations in patients with IBD. Metabolomic profiling revealed that chronic colitis in Winnie mice upregulated production of metabolites and altered several metabolic pathways, mostly affecting amino acid synthesis and breakdown of monosaccharides to short chain fatty acids. CONCLUSIONS Significant dysbiosis in the Winnie mouse gut replicates many changes observed in patients with IBD. These results provide justification for the suitability of this model to investigate mechanisms underlying the role of intestinal microbiota and metabolome in the pathophysiology of IBD.
Collapse
|
24
|
Chen M, Li Y, Sun Q, Pan A, Manson JE, Rexrode KM, Willett WC, Rimm EB, Hu FB. Dairy fat and risk of cardiovascular disease in 3 cohorts of US adults. Am J Clin Nutr 2016; 104:1209-1217. [PMID: 27557656 PMCID: PMC5081717 DOI: 10.3945/ajcn.116.134460] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/28/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Few prospective studies have examined dairy fat in relation to cardiovascular disease (CVD). OBJECTIVE We aimed to evaluate the association between dairy fat and incident CVD in US adults. DESIGN We followed 43,652 men in the Health Professionals Follow-Up Study (1986-2010), 87,907 women in the Nurses' Health Study (1980-2012), and 90,675 women in the Nurses' Health Study II (1991-2011). Dairy fat and other fat intakes were assessed every 4 y with the use of validated food-frequency questionnaires. RESULTS During 5,158,337 person-years of follow-up, we documented 14,815 incident CVD cases including 8974 coronary heart disease cases (nonfatal myocardial infarction or fatal coronary disease) and 5841 stroke cases. In multivariate analyses, compared with an equivalent amount of energy from carbohydrates (excluding fruit and vegetables), dairy fat intake was not significantly related to risk of total CVD (for a 5% increase in energy from dairy fat, the RR was 1.02; 95% CI: 0.98, 1.05), coronary heart disease (RR: 1.03; 95% CI: 0.98, 1.09), or stroke (RR: 0.99; 95% CI: 0.93, 1.05) (P > 0.05 for all). In models in which we estimated the effects of exchanging different fat sources, the replacement of 5% of energy intake from dairy fat with equivalent energy intake from polyunsaturated fatty acid (PUFA) or vegetable fat was associated with 24% (RR: 0.76; 95% CI: 0.71, 0.81) and 10% (RR: 0.90; 95% CI: 0.87, 0.93) lower risk of CVD, respectively, whereas the 5% energy intake substitution of other animal fat with dairy fat was associated with 6% increased CVD risk (RR: 1.06; 95% CI: 1.02, 1.09). CONCLUSIONS The replacement of animal fats, including dairy fat, with vegetable sources of fats and PUFAs may reduce risk of CVD. Whether the food matrix may modify the effect of dairy fat on health outcomes warrants further investigation.
Collapse
Affiliation(s)
- Mu Chen
- Departments of Nutrition and.,Epidemiology, Harvard TH Chan School of Public Health, Boston, MA
| | | | - Qi Sun
- Departments of Nutrition and.,Channing Division of Network Medicine and
| | - An Pan
- Department of Epidemiology and Biostatistics, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - JoAnn E Manson
- Epidemiology, Harvard TH Chan School of Public Health, Boston, MA.,Epidemiology, Harvard TH Chan School of Public Health, Boston, MA.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; and
| | - Kathryn M Rexrode
- Epidemiology, Harvard TH Chan School of Public Health, Boston, MA.,Epidemiology, Harvard TH Chan School of Public Health, Boston, MA.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; and
| | - Walter C Willett
- Departments of Nutrition and.,Epidemiology, Harvard TH Chan School of Public Health, Boston, MA.,Channing Division of Network Medicine and
| | - Eric B Rimm
- Departments of Nutrition and.,Epidemiology, Harvard TH Chan School of Public Health, Boston, MA.,Channing Division of Network Medicine and
| | - Frank B Hu
- Departments of Nutrition and .,Epidemiology, Harvard TH Chan School of Public Health, Boston, MA.,Channing Division of Network Medicine and
| |
Collapse
|
25
|
Kieffer DA, Piccolo BD, Marco ML, Kim EB, Goodson ML, Keenan MJ, Dunn TN, Knudsen KEB, Adams SH, Martin RJ. Obese Mice Fed a Diet Supplemented with Enzyme-Treated Wheat Bran Display Marked Shifts in the Liver Metabolome Concurrent with Altered Gut Bacteria. J Nutr 2016; 146:2445-2460. [PMID: 27798344 DOI: 10.3945/jn.116.238923] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/11/2016] [Accepted: 09/09/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Enzyme-treated wheat bran (ETWB) contains a fermentable dietary fiber previously shown to decrease liver triglycerides (TGs) and modify the gut microbiome in mice. It is not clear which mechanisms explain how ETWB feeding affects hepatic metabolism, but factors (i.e., xenometabolites) associated with specific microbes may be involved. OBJECTIVE The objective of this study was to characterize ETWB-driven shifts in the cecal microbiome and to identify correlates between microbial changes and diet-related differences in liver metabolism in diet-induced obese mice that typically display steatosis. METHODS Five-week-old male C57BL/6J mice fed a 45%-lard-based fat diet supplemented with ETWB (20% wt:wt) or rapidly digestible starch (control) (n = 15/group) for 10 wk were characterized by using a multi-omics approach. Multivariate statistical analysis was used to identify variables that were strong discriminators between the ETWB and control groups. RESULTS Body weight and liver TGs were decreased by ETWB feeding (by 10% and 25%, respectively; P < 0.001), and an index of liver reactive oxygen species was increased (by 29%; P < 0.01). The cecal microbiome showed an increase in Bacteroidetes (by 42%; P < 0.05) and a decrease in Firmicutes (by 16%; P < 0.05). Metabolites that were strong discriminators between the ETWB and control groups included decreased liver antioxidants (glutathione and α-tocopherol); decreased liver carbohydrate metabolites, including glucose; lower hepatic arachidonic acid; and increased liver and plasma β-hydroxybutyrate. Liver transcriptomics revealed key metabolic pathways affected by ETWB, especially those related to lipid metabolism and some fed- or fasting-regulated genes. CONCLUSIONS Together, these changes indicate that dietary fibers such as ETWB regulate hepatic metabolism concurrently with specific gut bacteria community shifts in C57BL/6J mice. It is proposed that these changes may elicit gut-derived signals that reach the liver via enterohepatic circulation, ultimately affecting host liver metabolism in a manner that mimics, in part, the fasting state.
Collapse
Affiliation(s)
- Dorothy A Kieffer
- Graduate Group in Nutritional Biology and.,Department of Nutrition.,Obesity and Metabolism Research Unit, USDA-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center and.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | | | - Eun Bae Kim
- Food Science and Technology Department, and.,Department of Animal Life Science, College of Animal Life Sciences, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | | | | | - Tamara N Dunn
- Graduate Group in Nutritional Biology and.,Department of Nutrition.,Obesity and Metabolism Research Unit, USDA-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA
| | | | - Sean H Adams
- Graduate Group in Nutritional Biology and .,Department of Nutrition.,Arkansas Children's Nutrition Center and.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Roy J Martin
- Graduate Group in Nutritional Biology and .,Department of Nutrition.,Obesity and Metabolism Research Unit, USDA-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA
| |
Collapse
|
26
|
Abstract
An investigation of human biological variation in urinary organic acids, including pyroglutamic acid along with 39 other compounds, was previously reported in which levels were determined for 8 weeks in healthy adult subjects. Here, unique, 4‐week‐long physiological trends for one of those compounds, pyroglutamic acid (PGA), are reported. When PGA levels for an individual rose above 40 μg/mg creatinine, 4‐week downward progressions occurred until levels reached values near 15 μg/mg creatinine and the pattern was reversed when levels for an individual were below that level in the early weeks of the study. The pattern was especially prominent among 8 of the 13 menstruating female subjects suggesting a possible association with metabolic stress of the menstrual cycle. However, it also appeared in 3 of the 8 male subjects where other sources of metabolic stress may be present. The menstrual association is consistent with estrogen‐mediated increase in oxidative stress. Since PGA is linked to glutathione turnover, the consistency of extreme values across all individuals displaying the pattern indicates that 15 and 40 μg/mg creatinine may represent limits that trigger shifts in sulfur amino acid metabolism. This is the first observation of approximate month‐long cyclic responses in a glutathione‐related urinary marker in humans.
Collapse
|
27
|
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
|
28
|
Aragonès G, Auguet T, Berlanga A, Guiu-Jurado E, Martinez S, Armengol S, Sabench F, Ras R, Hernandez M, Aguilar C, Colom J, Sirvent JJ, Del Castillo D, Richart C. Increased Circulating Levels of Alpha-Ketoglutarate in Morbidly Obese Women with Non-Alcoholic Fatty Liver Disease. PLoS One 2016; 11:e0154601. [PMID: 27123846 PMCID: PMC4849715 DOI: 10.1371/journal.pone.0154601] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/16/2016] [Indexed: 01/06/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) causes a wide spectrum of liver damage, ranging from simple steatosis to cirrhosis. However, simple steatosis (SS) and steatohepatitis (NASH) cannot yet be distinguished by clinical or laboratory features. The aim of this study was to assess the relationship between alpha-ketoglutarate and the degrees of NAFLD in morbidly obese patients. Materials and Methods We used a gas chromatography-quadruple time-of-flight-mass spectrometry analysis to quantify alpha-ketoglutarate in serum from normal-weight subjects (n = 30) and morbidly obese women (n = 97) with or without NAFLD. Results We found that serum levels of alpha-ketoglutarate were significantly higher in morbidly obese women than in normal-weight women. We showed that circulating levels of alpha-ketoglutarate were lower in lean controls and morbidly obese patients without NAFLD. We also found that alpha-ketoglutarate serum levels were higher in both SS and NASH than in normal liver of morbidly obese patients. However, there was no difference between SS and NASH. Moreover, we observed that circulating levels of alpha-ketoglutarate were associated with glucose metabolism parameters, lipid profile, hepatic enzymes and steatosis degree. In addition, diagnostic performance of alpha-ketoglutarate has been analyzed in NAFLD patients. The AUROC curves from patients with liver steatosis exhibited an acceptable clinical utility. Finally, we showed that the combination of biomarkers (AST, ALT and alpha-ketoglutarate) had the highest accuracy in diagnosing liver steatosis. Conclusion These findings suggest that alpha-ketoglutarate can determine the presence of non-alcoholic fatty liver in morbidly obese patients but it is not valid a biomarker for NASH.
Collapse
Affiliation(s)
- Gemma Aragonès
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Teresa Auguet
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
- Servei Medicina Interna, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Alba Berlanga
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Esther Guiu-Jurado
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Salomé Martinez
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Sandra Armengol
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Fàtima Sabench
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Rosa Ras
- Group of Research on Omic Methodologies (GROM). Centre for Omic Sciences (COS), Reus, Spain
| | - Mercè Hernandez
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Carmen Aguilar
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Josep Colom
- Servei Medicina Interna, Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Joan Josep Sirvent
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Daniel Del Castillo
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Cristóbal Richart
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
- Servei Medicina Interna, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
- * E-mail:
| |
Collapse
|
29
|
Ho JE, Larson MG, Ghorbani A, Cheng S, Chen MH, Keyes M, Rhee EP, Clish CB, Vasan RS, Gerszten RE, Wang TJ. Metabolomic Profiles of Body Mass Index in the Framingham Heart Study Reveal Distinct Cardiometabolic Phenotypes. PLoS One 2016; 11:e0148361. [PMID: 26863521 PMCID: PMC4749349 DOI: 10.1371/journal.pone.0148361] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
Background Although obesity and cardiometabolic traits commonly overlap, underlying pathways remain incompletely defined. The association of metabolite profiles across multiple cardiometabolic traits may lend insights into the interaction of obesity and metabolic health. We sought to investigate metabolic signatures of obesity and related cardiometabolic traits in the community using broad-based metabolomic profiling. Methods and Results We evaluated the association of 217 assayed metabolites and cross-sectional as well as longitudinal changes in cardiometabolic traits among 2,383 Framingham Offspring cohort participants. Body mass index (BMI) was associated with 69 of 217 metabolites (P<0.00023 for all), including aromatic (tyrosine, phenylalanine) and branched chain amino acids (valine, isoleucine, leucine). Additional metabolic pathways associated with BMI included the citric acid cycle (isocitrate, alpha-ketoglutarate, aconitate), the tryptophan pathway (kynurenine, kynurenic acid), and the urea cycle. There was considerable overlap in metabolite profiles between BMI, abdominal adiposity, insulin resistance [IR] and dyslipidemia, modest overlap of metabolite profiles between BMI and hyperglycemia, and little overlap with fasting glucose or elevated blood pressure. Metabolite profiles were associated with longitudinal changes in fasting glucose, but the involved metabolites (ornithine, 5-HIAA, aminoadipic acid, isoleucine, cotinine) were distinct from those associated with baseline glucose or other traits. Obesity status appeared to “modify” the association of 9 metabolites with IR. For example, bile acid metabolites were strongly associated with IR among obese but not lean individuals, whereas isoleucine had a stronger association with IR in lean individuals. Conclusions In this large-scale metabolite profiling study, body mass index was associated with a broad range of metabolic alterations. Metabolite profiling highlighted considerable overlap with abdominal adiposity, insulin resistance, and dyslipidemia, but not with fasting glucose or blood pressure traits.
Collapse
Affiliation(s)
- Jennifer E. Ho
- Framingham Heart Study of the National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, United States of America
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| | - Martin G. Larson
- Framingham Heart Study of the National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, United States of America
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts, United States of America
| | - Anahita Ghorbani
- Mount Auburn Hospital, Cambridge, Massachusetts, United States of America
| | - Susan Cheng
- Framingham Heart Study of the National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, United States of America
- Division of Cardiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ming-Huei Chen
- Framingham Heart Study of the National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, United States of America
| | - Michelle Keyes
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Eugene P. Rhee
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Renal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Ramachandran S. Vasan
- Framingham Heart Study of the National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, United States of America
- Division of Cardiology and Preventive Medicine, Department of Medicine, Boston University, Boston, Massachusetts, United States of America
| | - Robert E. Gerszten
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Thomas J. Wang
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
30
|
Kieffer DA, Piccolo BD, Vaziri ND, Liu S, Lau WL, Khazaeli M, Nazertehrani S, Moore ME, Marco ML, Martin RJ, Adams SH. Resistant starch alters gut microbiome and metabolomic profiles concurrent with amelioration of chronic kidney disease in rats. Am J Physiol Renal Physiol 2016; 310:F857-71. [PMID: 26841824 DOI: 10.1152/ajprenal.00513.2015] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023] Open
Abstract
Patients and animals with chronic kidney disease (CKD) exhibit profound alterations in the gut environment including shifts in microbial composition, increased fecal pH, and increased blood levels of gut microbe-derived metabolites (xenometabolites). The fermentable dietary fiber high amylose maize-resistant starch type 2 (HAMRS2) has been shown to alter the gut milieu and in CKD rat models leads to markedly improved kidney function. The aim of the present study was to identify specific cecal bacteria and cecal, blood, and urinary metabolites that associate with changes in kidney function to identify potential mechanisms involved with CKD amelioration in response to dietary resistant starch. Male Sprague-Dawley rats with adenine-induced CKD were fed a semipurified low-fiber diet or a high-fiber diet [59% (wt/wt) HAMRS2] for 3 wk (n = 9 rats/group). The cecal microbiome was characterized, and cecal contents, serum, and urine metabolites were analyzed. HAMRS2-fed rats displayed decreased cecal pH, decreased microbial diversity, and an increased Bacteroidetes-to-Firmicutes ratio. Several uremic retention solutes were altered in the cecal contents, serum, and urine, many of which had strong correlations with specific gut bacteria abundances, i.e., serum and urine indoxyl sulfate were reduced by 36% and 66%, respectively, in HAMRS2-fed rats and urine p-cresol was reduced by 47% in HAMRS2-fed rats. Outcomes from this study were coincident with improvements in kidney function indexes and amelioration of CKD outcomes previously reported for these rats, suggesting an important role for microbial-derived factors and gut microbe metabolism in regulating host kidney function.
Collapse
Affiliation(s)
- Dorothy A Kieffer
- Obesity and Metabolism Research Unit, United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California; Graduate Group in Nutritional Biology and Department of Nutrition, University of California, Davis, California
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Shuman Liu
- Division of Nephrology, University of California, Irvine, California
| | - Wei L Lau
- Division of Nephrology, University of California, Irvine, California
| | - Mahyar Khazaeli
- Division of Nephrology, University of California, Irvine, California
| | | | - Mary E Moore
- Department of Food Science and Technology, University of California, Davis, California; and
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, California; and
| | - Roy J Martin
- Obesity and Metabolism Research Unit, United States Department of Agriculture-Agricultural Research Service Western Human Nutrition Research Center, Davis, California
| | - Sean H Adams
- Graduate Group in Nutritional Biology and Department of Nutrition, University of California, Davis, California; Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
31
|
Palau-Rodriguez M, Tulipani S, Isabel Queipo-Ortuño M, Urpi-Sarda M, Tinahones FJ, Andres-Lacueva C. Metabolomic insights into the intricate gut microbial-host interaction in the development of obesity and type 2 diabetes. Front Microbiol 2015; 6:1151. [PMID: 26579078 PMCID: PMC4621279 DOI: 10.3389/fmicb.2015.01151] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
Gut microbiota has recently been proposed as a crucial environmental factor in the development of metabolic diseases such as obesity and type 2 diabetes, mainly due to its contribution in the modulation of several processes including host energy metabolism, gut epithelial permeability, gut peptide hormone secretion, and host inflammatory state. Since the symbiotic interaction between the gut microbiota and the host is essentially reflected in specific metabolic signatures, much expectation is placed on the application of metabolomic approaches to unveil the key mechanisms linking the gut microbiota composition and activity with disease development. The present review aims to summarize the gut microbial-host co-metabolites identified so far by targeted and untargeted metabolomic studies in humans, in association with impaired glucose homeostasis and/or obesity. An alteration of the co-metabolism of bile acids, branched fatty acids, choline, vitamins (i.e., niacin), purines, and phenolic compounds has been associated so far with the obese or diabese phenotype, in respect to healthy controls. Furthermore, anti-diabetic treatments such as metformin and sulfonylurea have been observed to modulate the gut microbiota or at least their metabolic profiles, thereby potentially affecting insulin resistance through indirect mechanisms still unknown. Despite the scarcity of the metabolomic studies currently available on the microbial-host crosstalk, the data-driven results largely confirmed findings independently obtained from in vitro and animal model studies, putting forward the mechanisms underlying the implication of a dysfunctional gut microbiota in the development of metabolic disorders.
Collapse
Affiliation(s)
- Magali Palau-Rodriguez
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| | - Sara Tulipani
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain ; Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain
| | - Maria Isabel Queipo-Ortuño
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain ; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII) Madrid, Spain
| | - Mireia Urpi-Sarda
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| | - Francisco J Tinahones
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain ; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII) Madrid, Spain
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| |
Collapse
|
32
|
Modeling and Classification of Kinetic Patterns of Dynamic Metabolic Biomarkers in Physical Activity. PLoS Comput Biol 2015; 11:e1004454. [PMID: 26317529 PMCID: PMC4552566 DOI: 10.1371/journal.pcbi.1004454] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/09/2015] [Indexed: 11/19/2022] Open
Abstract
The objectives of this work were the classification of dynamic metabolic biomarker candidates and the modeling and characterization of kinetic regulatory mechanisms in human metabolism with response to external perturbations by physical activity. Longitudinal metabolic concentration data of 47 individuals from 4 different groups were examined, obtained from a cycle ergometry cohort study. In total, 110 metabolites (within the classes of acylcarnitines, amino acids, and sugars) were measured through a targeted metabolomics approach, combining tandem mass spectrometry (MS/MS) with the concept of stable isotope dilution (SID) for metabolite quantitation. Biomarker candidates were selected by combined analysis of maximum fold changes (MFCs) in concentrations and P-values resulting from statistical hypothesis testing. Characteristic kinetic signatures were identified through a mathematical modeling approach utilizing polynomial fitting. Modeled kinetic signatures were analyzed for groups with similar behavior by applying hierarchical cluster analysis. Kinetic shape templates were characterized, defining different forms of basic kinetic response patterns, such as sustained, early, late, and other forms, that can be used for metabolite classification. Acetylcarnitine (C2), showing a late response pattern and having the highest values in MFC and statistical significance, was classified as late marker and ranked as strong predictor (MFC = 1.97, P < 0.001). In the class of amino acids, highest values were shown for alanine (MFC = 1.42, P < 0.001), classified as late marker and strong predictor. Glucose yields a delayed response pattern, similar to a hockey stick function, being classified as delayed marker and ranked as moderate predictor (MFC = 1.32, P < 0.001). These findings coincide with existing knowledge on central metabolic pathways affected in exercise physiology, such as β-oxidation of fatty acids, glycolysis, and glycogenolysis. The presented modeling approach demonstrates high potential for dynamic biomarker identification and the investigation of kinetic mechanisms in disease or pharmacodynamics studies using MS data from longitudinal cohort studies. Human metabolism is controlled through basic kinetic regulatory mechanisms, where the overall system aims to maintain a state of homeostasis. In response to external perturbations, such as environmental influences, nutrition or physical exercise, circulating metabolites show specific kinetic response patterns, which can be computationally modeled. In this work, we searched for dynamic metabolic biomarker candidates and analyzed specific kinetic mechanisms from longitudinal metabolic concentration data, obtained through a cycle ergometry stress test. In total, 110 metabolites measured from blood samples of 47 individuals were analyzed using tandem mass spectrometry (MS/MS). Dynamic biomarker candidates could be selected based on the amplitudes of changes in metabolite concentrations and the significance of statistical hypothesis testing. We were able to characterize specific kinetic patterns for groups of similarly behaving metabolites. Kinetic shape templates were identified, defining basic kinetic response patterns to physical exercise, such as sustained, early, late and other shape forms. The presented approach contributes to a better understanding of (patho)physiological biochemical mechanisms in human health, disease or during drug therapy, by offering tools for classifying dynamic biomarker candidates and for modeling and characterizing kinetic regulatory mechanisms from longitudinal experimental data.
Collapse
|
33
|
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: 48] [Impact Index Per Article: 4.8] [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
|
34
|
Allen JM, Berg Miller ME, Pence BD, Whitlock K, Nehra V, Gaskins HR, White BA, Fryer JD, Woods JA. Voluntary and forced exercise differentially alters the gut microbiome in C57BL/6J mice. J Appl Physiol (1985) 2015; 118:1059-66. [PMID: 25678701 DOI: 10.1152/japplphysiol.01077.2014] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/08/2015] [Indexed: 12/18/2022] Open
Abstract
We have previously shown that voluntary wheel running (VWR) attenuates, whereas forced treadmill running (FTR) exacerbates, intestinal inflammation and clinical outcomes in a mouse model of colitis. As the gut microbiome is implicated in colitis, we hypothesized that VWR and FTR would differentially affect the gut microbiome. Mice (9-10/treatment) were randomly assigned to VWR, FTR, or sedentary home cage control (SED) for 6 wk. VWR were given running wheel access, whereas FTR ran on a treadmill for 40 min/day at 8-12 m/min, 5% grade. Forty-eight hours after the last exercise session, DNA was isolated from the fecal pellets and cecal contents, and the conserved bacterial 16S rRNA gene was amplified and sequenced using the Illumina Miseq platform. Permutational multivariate analysis of variance based on weighted UniFrac distance matrix revealed different bacterial clusters between feces and cecal contents in all groups (P < 0.01). Interestingly, the community structures of the three treatment groups clustered separately from each other in both gut regions (P < 0.05). Contrary to our hypothesis, the α-diversity metric, Chao1, indicated that VWR led to reduced bacterial richness compared with FTR or SED (P < 0.05). Taxonomic evaluation revealed that both VWR and FTR altered many individual bacterial taxa. Of particular interest, Turicibacter spp., which has been strongly associated with immune function and bowel disease, was significantly lower in VWR vs. SED/FTR. These data indicate that VWR and FTR differentially alter the intestinal microbiome of mice. These effects were observed in both the feces and cecum despite vastly different community structures between each intestinal region.
Collapse
Affiliation(s)
- Jacob M Allen
- Department of Kinesiology and Community Health, Mayo Clinic, Rochester, Minnesota; Integrative Immunology and Behavior Program, Mayo Clinic, Rochester, Minnesota
| | - Margret E Berg Miller
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Brandt D Pence
- Department of Kinesiology and Community Health, Mayo Clinic, Rochester, Minnesota; Integrative Immunology and Behavior Program, Mayo Clinic, Rochester, Minnesota
| | - Keith Whitlock
- Department of Kinesiology and Community Health, Mayo Clinic, Rochester, Minnesota
| | - Vandana Nehra
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Bryan A White
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Jeffrey A Woods
- Department of Kinesiology and Community Health, Mayo Clinic, Rochester, Minnesota; Integrative Immunology and Behavior Program, Mayo Clinic, Rochester, Minnesota; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois;
| |
Collapse
|
35
|
Abstract
Metabolic pathways are tightly regulated in a tissue-specific manner to maintain whole-body homeostasis. Nutrients and hormones control these pathways at the level of transcription, translation, and/or post-translation. Genomic and proteomic tools have been predominantly used to understand metabolic regulation, and only a few studies used metabolomics approaches. Metabolomics is a powerful, unbiased approach that allows comprehensive metabolic analysis of physiologic measurements and energy balance. Thus, nutrimetabolomics can expedite our ability to identify metabolic diseases that are influenced by nutrients and to develop targeted diet-based treatments. Presentations at this symposium reviewed current resources and platforms for metabolic profiling along with statistical and bioinformatics tools for data and pathway analyses. Specific applications of metabolomics were illustrated in nutritional and disease conditions, including polycystic ovary syndrome, diabetes, and obesity and in host-gut microbiome interactions.
Collapse
Affiliation(s)
| | | | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and the Obesity Research Cluster, College of Human Sciences, Texas Tech University, Lubbock, TX
| |
Collapse
|
36
|
Kuehnbaum NL, Gillen JB, Gibala MJ, Britz-McKibbin P. Personalized metabolomics for predicting glucose tolerance changes in sedentary women after high-intensity interval training. Sci Rep 2014; 4:6166. [PMID: 25164777 PMCID: PMC4147371 DOI: 10.1038/srep06166] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/04/2014] [Indexed: 12/16/2022] Open
Abstract
High-intensity interval training (HIIT) offers a practical approach for enhancing cardiorespiratory fitness, however its role in improving glucose regulation among sedentary yet normoglycemic women remains unclear. Herein, multi-segment injection capillary electrophoresis-mass spectrometry is used as a high-throughput platform in metabolomics to assess dynamic responses of overweight/obese women (BMI > 25, n = 11) to standardized oral glucose tolerance tests (OGTTs) performed before and after a 6-week HIIT intervention. Various statistical methods were used to classify plasma metabolic signatures associated with post-prandial glucose and/or training status when using a repeated measures/cross-over study design. Branched-chain/aromatic amino acids and other intermediates of urea cycle and carnitine metabolism decreased over time in plasma after oral glucose loading. Adaptive exercise-induced changes to plasma thiol redox and orthinine status were measured for trained subjects while at rest in a fasting state. A multi-linear regression model was developed to predict changes in glucose tolerance based on a panel of plasma metabolites measured for naïve subjects in their untrained state. Since treatment outcomes to physical activity are variable between-subjects, prognostic markers offer a novel approach to screen for potential negative responders while designing lifestyle modifications that maximize the salutary benefits of exercise for diabetes prevention on an individual level.
Collapse
Affiliation(s)
- Naomi L Kuehnbaum
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada
| | - Jenna B Gillen
- Department of Kinesiology, McMaster University, Hamilton, Canada
| | - Martin J Gibala
- Department of Kinesiology, McMaster University, Hamilton, Canada
| | | |
Collapse
|