101
|
Regan JA, Shah SH. Obesity Genomics and Metabolomics: a Nexus of Cardiometabolic Risk. Curr Cardiol Rep 2020; 22:174. [PMID: 33040225 DOI: 10.1007/s11886-020-01422-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Obesity is a significant international public health epidemic with major downstream consequences on morbidity and mortality. While lifestyle factors contribute, there is an evolving understanding of genomic and metabolomic pathways involved with obesity and its relationship with cardiometabolic risk. This review will provide an overview of some of these important findings from both a biologic and clinical perspective. RECENT FINDINGS Recent studies have identified polygenic risk scores and metabolomic biomarkers of obesity and related outcomes, which have also highlighted biological pathways, such as the branched-chain amino acid (BCAA) pathway that is dysregulated in this disease. These biomarkers may help in personalizing obesity interventions and for mitigation of future cardiometabolic risk. A multifaceted approach is necessary to impact the growing epidemic of obesity and related diseases. This will likely include incorporating precision medicine approaches with genomic and metabolomic biomarkers to personalize interventions and improve risk prediction.
Collapse
Affiliation(s)
- Jessica A Regan
- Department of Medicine, Duke University, Durham, NC, USA.,Duke Molecular Physiology Institute, Duke University, 300 N. Duke Street, DUMC, Box 104775, Durham, NC, 27701, USA
| | - Svati H Shah
- Department of Medicine, Duke University, Durham, NC, USA. .,Duke Molecular Physiology Institute, Duke University, 300 N. Duke Street, DUMC, Box 104775, Durham, NC, 27701, USA.
| |
Collapse
|
102
|
Khambule L, Snyman T, Norris SA, Crowther NJ, George JA. Branched-chain and aromatic amino acids and cardiometabolic risk in Black African and Asian Indian populations. Metabolomics 2020; 16:108. [PMID: 33033875 DOI: 10.1007/s11306-020-01734-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Studies have shown that systemic levels of branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs) are elevated in cardiometabolic diseases (CMDs) in populations resident in high income countries. However, little is known about the association of BCAAs and AAAs with metabolic syndrome and its components in Asian Indian (AI) and Black African (BA) populations. OBJECTIVE The aim of this study was to describe the association of BCAAs and AAAs with the metabolic syndrome, its individual components and insulin resistance in AI and BA populations. METHODS Serum samples collected from AI (n = 349) and BA (n = 369) subjects were used to measure levels of BCAAs and AAAs by ultra-pressure liquid chromatography tandem mass spectrometry (UPLC-MS/MS). Anthropometric, demographic and cardiometabolic variables were measured in all subjects. RESULTS The sum of BCAAs and AAAs was higher in AIs compared to BAs. The BCAAs and AAAs were positively associated with insulin resistance, metabolic syndrome and its individual components. This was particularly the case for AI subjects, in unadjusted regression models. However, these associations were non-significant after adjusting for co-variates, particularly visceral adipose tissue (VAT). Triglyceride levels were significantly associated with valine and leucine levels in BAs even after adjustment for co-variates. Lastly, we found that fasting circulatory BCAA and AAA levels are strongly correlated with VAT in both populations. CONCLUSION This study identified specific associations of serum valine and leucine levels with triglycerides in BAs. The association of amino acids with CMDs was observed in AIs, but was found to be the result of confounding by VAT. Further studies are required to determine whether BCAAs and AAAs are aetiological factors in CMDs and how VAT modulates their serum levels.
Collapse
Affiliation(s)
- Lungile Khambule
- Department of Chemical Pathology, National Health Laboratory Service and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Tracy Snyman
- Department of Chemical Pathology, National Health Laboratory Service and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shane A Norris
- Medical Research Council/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nigel J Crowther
- Department of Chemical Pathology, National Health Laboratory Service and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jaya A George
- Department of Chemical Pathology, National Health Laboratory Service and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
103
|
Sayda MH, Phillips BE, Williams JP, Greenhaff PL, Wilkinson DJ, Smith K, Atherton PJ. Associations between Plasma Branched Chain Amino Acids and Health Biomarkers in Response to Resistance Exercise Training Across Age. Nutrients 2020; 12:nu12103029. [PMID: 33023275 PMCID: PMC7601782 DOI: 10.3390/nu12103029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 09/24/2020] [Accepted: 09/30/2020] [Indexed: 01/10/2023] Open
Abstract
Leucine, isoleucine and valine (i.e., the branched chain amino acids, BCAA) play a key role in the support and regulation of tissue protein regulation and also as energy substrates. However, positive relationships exist between elevated levels of BCAA and insulin resistance (IR). Thus, we sought to investigate the links between fasting plasma BCAA following a progressive resistance exercise training (RET) programme, an intervention known to improve metabolic health. Fasting plasma BCAA were quantified in adults (young: 18-28 y, n = 8; middle-aged: 45-55 y, n = 9; older: 65-75 y, n = 15; BMI: 23-28 kg/m2, both males and females (~50:50), in a cross-sectional, intervention study. Participants underwent 20-weeks whole-body RET. Measurements of body composition, muscle strength (1-RM) and metabolic health biomarkers (e.g., HOMA-IR) were made pre- and post-RET. BCAA concentrations were determined by gas-chromatography mass spectrometry (GC-MS). No associations were observed across age with BCAA; however, RET elicited (p < 0.05) increases in plasma BCAA (all age-groups), while HOMA-IR scores reduced (p < 0.05) following RET. After RET, positive correlations in lean body mass (p = 0.007) and strength gains (p = 0.001) with fasting BCAA levels were observed. Elevated BCAA are not a robust marker of ageing nor IR in those with a healthy BMI; rather, despite decreasing IR, RET was associated with increased BCAA.
Collapse
Affiliation(s)
- Mariwan H. Sayda
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham NG7 2RD, UK; (M.H.S.); (B.E.P.); (J.P.W.); (P.L.G.); (D.J.W.); (K.S.)
- The National Centre for Sport and Exercise Medicine—East Midlands, University of Nottingham, Nottingham NG7 2UH, UK
| | - Bethan E. Phillips
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham NG7 2RD, UK; (M.H.S.); (B.E.P.); (J.P.W.); (P.L.G.); (D.J.W.); (K.S.)
- The National Centre for Sport and Exercise Medicine—East Midlands, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - John P. Williams
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham NG7 2RD, UK; (M.H.S.); (B.E.P.); (J.P.W.); (P.L.G.); (D.J.W.); (K.S.)
| | - Paul L. Greenhaff
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham NG7 2RD, UK; (M.H.S.); (B.E.P.); (J.P.W.); (P.L.G.); (D.J.W.); (K.S.)
- The National Centre for Sport and Exercise Medicine—East Midlands, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Daniel J. Wilkinson
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham NG7 2RD, UK; (M.H.S.); (B.E.P.); (J.P.W.); (P.L.G.); (D.J.W.); (K.S.)
- The National Centre for Sport and Exercise Medicine—East Midlands, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Ken Smith
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham NG7 2RD, UK; (M.H.S.); (B.E.P.); (J.P.W.); (P.L.G.); (D.J.W.); (K.S.)
- The National Centre for Sport and Exercise Medicine—East Midlands, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Philip J. Atherton
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham NG7 2RD, UK; (M.H.S.); (B.E.P.); (J.P.W.); (P.L.G.); (D.J.W.); (K.S.)
- The National Centre for Sport and Exercise Medicine—East Midlands, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK
- Correspondence: ; Tel.: +01-332-724-725
| |
Collapse
|
104
|
Méndez-Rodríguez KB, Figueroa-Vega N, Ilizaliturri-Hernandez CA, Cardona-Alvarado M, Borjas-García JA, Kornhauser C, Malacara JM, Flores-Ramírez R, Pérez-Vázquez FJ. Identification of metabolic markers in patients with type 2 Diabetes by Ultrafast gas chromatography coupled to electronic nose. A pilot study. Biomed Chromatogr 2020; 34:e4956. [PMID: 32706910 DOI: 10.1002/bmc.4956] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/27/2022]
Abstract
Metabolomics is a potential tool for the discovery of new biomarkers in the early diagnosis of diseases. An ultra-fast gas chromatography system equipped to an electronic nose detector (FGC eNose) was used to identify the metabolomic profile of Volatile Organic Compounds (VOCs) in type 2 diabetes (T2D) urine from Mexican population. A cross-sectional, comparative, and clinical study with translational approach was performed. We recruited twenty T2D patients and twenty-one healthy subjects. Urine samples were taken and analyzed by FGC eNose. Eighty-eight compounds were identified through Kovats's indexes. A natural variation of 30% between the metabolites, expressed by study groups, was observed in Principal Component 1 and 2 with a significant difference (p < 0.001). The model, performed through a Canonical Analysis of Principal coordinated (CAP), allowed a correct classification of 84.6% between healthy and T2D patients, with a 15.4% error. The metabolites 2-propenal, 2-propanol, butane- 2,3-dione and 2-methylpropanal, were increased in patients with T2D, and they were strongly correlated with discrimination between clinically healthy people and T2D patients. This study identified metabolites in urine through FGC eNose that can be used as biomarkers in the identification of T2D patients. However, more studies are needed for its implementation in clinical practice.
Collapse
Affiliation(s)
- Karen Beatriz Méndez-Rodríguez
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACyT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, S.L.P., Mexico
| | - Nicté Figueroa-Vega
- Department of Medical Sciences, University of Guanajuato, León, Gto., Mexico
| | - César Arturo Ilizaliturri-Hernandez
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACyT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, S.L.P., Mexico
| | | | | | - Carlos Kornhauser
- Department of Medical Sciences, University of Guanajuato, León, Gto., Mexico
| | | | - Rogelio Flores-Ramírez
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACyT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, S.L.P., Mexico.,CONACYT Research Fellow, Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, S.L.P., Mexico
| | - Francisco Javier Pérez-Vázquez
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACyT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, S.L.P., Mexico.,CONACYT Research Fellow, Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT), Universidad Autónoma de San Luis Potosí, San Luis Potosí, S.L.P., Mexico
| |
Collapse
|
105
|
Sjøberg KA, Schmoll D, Piper MDW, Kiens B, Rose AJ. Effects of Short-Term Dietary Protein Restriction on Blood Amino Acid Levels in Young Men. Nutrients 2020; 12:nu12082195. [PMID: 32717986 PMCID: PMC7468950 DOI: 10.3390/nu12082195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/27/2022] Open
Abstract
Pre-clinical studies show that dietary protein restriction (DPR) improves healthspan and retards many age-related diseases such as type 2 diabetes. While mouse studies have shown that restriction of certain essential amino acids is required for this response, less is known about which amino acids are affected by DPR in humans. Here, using a within-subjects diet design, we examined the effects of dietary protein restriction in the fasted state, as well as acutely after meal feeding, on blood plasma amino acid levels. While very few amino acids were affected by DPR in the fasted state, several proteinogenic AAs such as isoleucine, leucine, lysine, phenylalanine, threonine, tyrosine, and valine were lower in the meal-fed state with DPR. In addition, the non-proteinogenic AAs such as 1- and 3-methyl-histidine were also lower with meal feeding during DPR. Lastly, using in silico predictions of the most limiting essential AAs compared with human exome AA usage, we demonstrate that leucine, methionine, and threonine are potentially the most limiting essential AAs with DPR. In summary, acute meal feeding allows more accurate determination of which AAs are affected by dietary interventions, with most essential AAs lowered by DPR.
Collapse
Affiliation(s)
- Kim A. Sjøberg
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2100 Copenhagen, Denmark; (K.A.S.); (B.K.)
| | - Dieter Schmoll
- Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, 65926 Frankfurt am Main, Germany;
| | - Matthew D. W. Piper
- School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia;
| | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2100 Copenhagen, Denmark; (K.A.S.); (B.K.)
| | - Adam J. Rose
- Nutrient Metabolism & Signalling Laboratory, Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Australia
- Correspondence:
| |
Collapse
|
106
|
Lu J, Gu Y, Liu H, Wang L, Li W, Li W, Leng J, Zhang S, Qi L, Yang X, Hu G. Daily Branched-Chain Amino Acid Intake and Risks of Obesity and Insulin Resistance in Children: A Cross-Sectional Study. Obesity (Silver Spring) 2020; 28:1310-1316. [PMID: 32510827 PMCID: PMC7311291 DOI: 10.1002/oby.22834] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 01/05/2023]
Abstract
OBJECTIVE This study aimed to investigate the association of daily branched-chain amino acid (BCAA) intake with the risks of obesity and insulin resistance in children of mothers with gestational diabetes mellitus (GDM). METHODS Daily BCAA intake was calculated by using a validated food frequency questionnaire in 996 children of mothers with GDM. The odds ratios (ORs) (95% CI) of childhood obesity and insulin resistance were obtained using logistic regression models. RESULTS The multivariable-adjusted ORs for overweight and insulin resistance increased across quartiles of daily BCAA intake (P < 0.05 for trend). Multivariable-adjusted ORs for each 1-SD increase in BCAA intake were 1.37 (1.16-1.62) for overweight and 1.19 (1.02-1.38) for insulin resistance. After additional adjustment of children's daily total energy intake, the OR was still significant for overweight risk but no longer significant for insulin resistance. There were positive associations of daily leucine, isoleucine, and valine intake with the risks for overweight and insulin resistance. CONCLUSIONS Daily BCAA intake was associated with increased risks for overweight and insulin resistance in children of mothers with GDM, but this association was not fully independent of children's daily energy intake. Restriction in dietary BCAA may help prevent childhood obesity and insulin resistance.
Collapse
Affiliation(s)
- Jun Lu
- Department of Endocrinology and Metabolism, Fengxian Hospital affiliated to Southern Medical University, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital South Campus, Shanghai University of Medicine and Health Sciences Affiliated Shanghai Sixth People’s Hospital South Campus, Shanghai, China
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Yuying Gu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
- Shanghai Business School, Shanghai, China
| | - Huikun Liu
- Tianjin Women’s and Children’s Health Center, Tianjin, China
| | - Leishen Wang
- Tianjin Women’s and Children’s Health Center, Tianjin, China
| | - Wei Li
- Tianjin Women’s and Children’s Health Center, Tianjin, China
| | - Weiqin Li
- Tianjin Women’s and Children’s Health Center, Tianjin, China
| | - Junhong Leng
- Tianjin Women’s and Children’s Health Center, Tianjin, China
| | - Shuang Zhang
- Tianjin Women’s and Children’s Health Center, Tianjin, China
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Xilin Yang
- Department of Epidemiology, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
107
|
Armstrong SC, Windom M, Bihlmeyer NA, Li JS, Shah SH, Story M, Zucker N, Kraus WE, Pagidipati N, Peterson E, Wong C, Wiedemeier M, Sibley L, Berchuck SI, Merrill P, Zizzi A, Sarria C, Dressman HK, Rawls JF, Skinner AC. Rationale and design of "Hearts & Parks": study protocol for a pragmatic randomized clinical trial of an integrated clinic-community intervention to treat pediatric obesity. BMC Pediatr 2020; 20:308. [PMID: 32590958 PMCID: PMC7318397 DOI: 10.1186/s12887-020-02190-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/02/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The prevalence of child and adolescent obesity and severe obesity continues to increase despite decades of policy and research aimed at prevention. Obesity strongly predicts cardiovascular and metabolic disease risk; both begin in childhood. Children who receive intensive behavioral interventions can reduce body mass index (BMI) and reverse disease risk. However, delivering these interventions with fidelity at scale remains a challenge. Clinic-community partnerships offer a promising strategy to provide high-quality clinical care and deliver behavioral treatment in local park and recreation settings. The Hearts & Parks study has three broad objectives: (1) evaluate the effectiveness of the clinic-community model for the treatment of child obesity, (2) define microbiome and metabolomic signatures of obesity and response to lifestyle change, and (3) inform the implementation of similar models in clinical systems. METHODS Methods are designed for a pragmatic randomized, controlled clinical trial (n = 270) to test the effectiveness of an integrated clinic-community child obesity intervention as compared with usual care. We are powered to detect a difference in body mass index (BMI) between groups at 6 months, with follow up to 12 months. Secondary outcomes include changes in biomarkers for cardiovascular disease, psychosocial risk, and quality of life. Through collection of biospecimens (serum and stool), additional exploratory outcomes include microbiome and metabolomics biomarkers of response to lifestyle modification. DISCUSSION We present the study design, enrollment strategy, and intervention details for a randomized clinical trial to measure the effectiveness of a clinic-community child obesity treatment intervention. This study will inform a critical area in child obesity and cardiovascular risk research-defining outcomes, implementation feasibility, and identifying potential molecular mechanisms of treatment response. CLINICAL TRIAL REGISTRATION NCT03339440 .
Collapse
Affiliation(s)
- Sarah C. Armstrong
- Department of Pediatrics, Duke University, Durham, NC 27710 USA
- Duke Clinical Research Institute, Duke University, Durham, NC 27710 USA
| | | | | | - Jennifer S. Li
- Department of Pediatrics, Duke University, Durham, NC 27710 USA
| | - Svati H. Shah
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710 USA
- Department of Medicine, Duke University, Durham, NC 27710 USA
| | - Mary Story
- Department of Family Medicine and Community Health, Duke University, Durham, NC 27710 USA
| | - Nancy Zucker
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27710 USA
| | | | - Neha Pagidipati
- Duke Clinical Research Institute, Duke University, Durham, NC 27710 USA
- Department of Medicine, Duke University, Durham, NC 27710 USA
| | - Eric Peterson
- Duke Clinical Research Institute, Duke University, Durham, NC 27710 USA
- Department of Medicine, Duke University, Durham, NC 27710 USA
| | - Charlene Wong
- Department of Pediatrics, Duke University, Durham, NC 27710 USA
- Duke Clinical Research Institute, Duke University, Durham, NC 27710 USA
| | | | - Lauren Sibley
- University of North Carolina School of Medicine, Chapel Hill, NC 27516 USA
| | - Samuel I. Berchuck
- Department of Statistical Science, Duke University, Durham, NC 27710 USA
| | - Peter Merrill
- Duke Clinical Research Institute, Duke University, Durham, NC 27710 USA
| | - Alexandra Zizzi
- Department of Pediatrics, Duke University, Durham, NC 27710 USA
| | - Charles Sarria
- Department of Pediatrics, Duke University, Durham, NC 27710 USA
| | - Holly K. Dressman
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708 USA
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708 USA
| | - Asheley C. Skinner
- Duke Clinical Research Institute, Duke University, Durham, NC 27710 USA
- Department of Population Health Sciences, Duke University, 215 Morris Street, Suite 210, Durham, NC 27701 USA
| |
Collapse
|
108
|
Chatterjee R, Davenport CA, Kwee L, D'Alessio D, Svetkey LP, Lin PH, Slentz CA, Ilkayeva O, Johnson J, Edelman D, Shah SH. Preliminary evidence of effects of potassium chloride on a metabolomic path to diabetes and cardiovascular disease. Metabolomics 2020; 16:75. [PMID: 32556595 PMCID: PMC8053254 DOI: 10.1007/s11306-020-01696-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/11/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Low potassium intake can affect cardiovascular disease (CVD) risk and cardiometabolic risk factors. OBJECTIVE We hypothesize that potassium chloride (KCl) supplementation can improve cardiovascular risk metabolomic profile. METHODS In this secondary analysis of a pilot randomized clinical trial (RCT) of 26 participants with prediabetes randomized to KCl or placebo, we performed targeted mass-spectrometry-based metabolomic profiling on baseline and 12-week (end-of-study) plasma samples. Principal component analysis (PCA) was used to reduce the many correlated metabolites into fewer, independent factors that retain most of the information in the original data. RESULTS Those taking KCl had significant reductions (corresponding to lower cardiovascular risk) in the branched-chain amino acids (BCAA) factor (P = 0.004) and in valine levels (P = 0.02); and non-significant reductions in short-chain acylcarnitines (SCA) factor (P = 0.11). CONCLUSIONS KCl supplementation may improve circulating BCAA levels, which may reflect improvements in overall cardiometabolic risk profile. CLINICAL TRIALS REGISTRY Clinicaltrials.gov identifier: NCT02236598; https://clinicaltrials.gov/ct2/show/NCT02236598.
Collapse
Affiliation(s)
- Ranee Chatterjee
- Department of Medicine, Duke University, 200 Morris Street, 3rd Floor, Durham, NC, 27701, USA.
| | - Clemontina A Davenport
- Department of Medicine, Duke University, 200 Morris Street, 3rd Floor, Durham, NC, 27701, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Lydia Kwee
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - David D'Alessio
- Department of Medicine, Duke University, 200 Morris Street, 3rd Floor, Durham, NC, 27701, USA
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Laura P Svetkey
- Department of Medicine, Duke University, 200 Morris Street, 3rd Floor, Durham, NC, 27701, USA
| | - Pao-Hwa Lin
- Department of Medicine, Duke University, 200 Morris Street, 3rd Floor, Durham, NC, 27701, USA
| | - Cris A Slentz
- Department of Medicine, Duke University, 200 Morris Street, 3rd Floor, Durham, NC, 27701, USA
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Johanna Johnson
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - David Edelman
- Department of Medicine, Duke University, 200 Morris Street, 3rd Floor, Durham, NC, 27701, USA
| | - Svati H Shah
- Department of Medicine, Duke University, 200 Morris Street, 3rd Floor, Durham, NC, 27701, USA
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| |
Collapse
|
109
|
Amino acid and lipid metabolism in post-gestational diabetes and progression to type 2 diabetes: A metabolic profiling study. PLoS Med 2020; 17:e1003112. [PMID: 32433647 PMCID: PMC7239388 DOI: 10.1371/journal.pmed.1003112] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/20/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Women with a history of gestational diabetes mellitus (GDM) have a 7-fold higher risk of developing type 2 diabetes (T2D) during midlife and an elevated risk of developing hypertension and cardiovascular disease. Glucose tolerance reclassification after delivery is recommended, but fewer than 40% of women with GDM are tested. Thus, improved risk stratification methods are needed, as is a deeper understanding of the pathology underlying the transition from GDM to T2D. We hypothesize that metabolites during the early postpartum period accurately distinguish risk of progression from GDM to T2D and that metabolite changes signify underlying pathophysiology for future disease development. METHODS AND FINDINGS The study utilized fasting plasma samples collected from a well-characterized prospective research study of 1,035 women diagnosed with GDM. The cohort included racially/ethnically diverse pregnant women (aged 20-45 years-33% primiparous, 37% biparous, 30% multiparous) who delivered at Kaiser Permanente Northern California hospitals from 2008 to 2011. Participants attended in-person research visits including 2-hour 75-g oral glucose tolerance tests (OGTTs) at study baseline (6-9 weeks postpartum) and annually thereafter for 2 years, and we retrieved diabetes diagnoses from electronic medical records for 8 years. In a nested case-control study design, we collected fasting plasma samples among women without diabetes at baseline (n = 1,010) to measure metabolites among those who later progressed to incident T2D or did not develop T2D (non-T2D). We studied 173 incident T2D cases and 485 controls (pair-matched on BMI, age, and race/ethnicity) to discover metabolites associated with new onset of T2D. Up to 2 years post-baseline, we analyzed samples from 98 T2D cases with 239 controls to reveal T2D-associated metabolic changes. The longitudinal analysis tracked metabolic changes within individuals from baseline to 2 years of follow-up as the trajectory of T2D progression. By building prediction models, we discovered a distinct metabolic signature in the early postpartum period that predicted future T2D with a median discriminating power area under the receiver operating characteristic curve of 0.883 (95% CI 0.820-0.945, p < 0.001). At baseline, the most striking finding was an overall increase in amino acids (AAs) as well as diacyl-glycerophospholipids and a decrease in sphingolipids and acyl-alkyl-glycerophospholipids among women with incident T2D. Pathway analysis revealed up-regulated AA metabolism, arginine/proline metabolism, and branched-chain AA (BCAA) metabolism at baseline. At follow-up after the onset of T2D, up-regulation of AAs and down-regulation of sphingolipids and acyl-alkyl-glycerophospholipids were sustained or strengthened. Notably, longitudinal analyses revealed only 10 metabolites associated with progression to T2D, implicating AA and phospholipid metabolism. A study limitation is that all of the analyses were performed with the same cohort. It would be ideal to validate our findings in an independent longitudinal cohort of women with GDM who had glucose tolerance tested during the early postpartum period. CONCLUSIONS In this study, we discovered a metabolic signature predicting the transition from GDM to T2D in the early postpartum period that was superior to clinical parameters (fasting plasma glucose, 2-hour plasma glucose). The findings suggest that metabolic dysregulation, particularly AA dysmetabolism, is present years prior to diabetes onset, and is revealed during the early postpartum period, preceding progression to T2D, among women with GDM. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01967030.
Collapse
|
110
|
Noerman S, Kolehmainen M, Hanhineva K. Profiling of Endogenous and Gut Microbial Metabolites to Indicate Metabotype-Specific Dietary Responses: A Systematic Review. Adv Nutr 2020; 11:1237-1254. [PMID: 32271864 PMCID: PMC7490160 DOI: 10.1093/advances/nmaa031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/21/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022] Open
Abstract
Upon dietary exposure, the endogenous metabolism responds to the diet-derived nutrients and bioactive compounds, such as phytochemicals. However, the responses vary remarkably due to the interplay with other dietary components, lifestyle exposures, and intrinsic factors, which lead to differences in endogenous regulatory metabolism. These physiological processes are evidenced as a signature profile composed of various metabolites constituting metabolic phenotypes, or metabotypes. The metabolic profiling of biological samples following dietary intake hence would provide information about diet-that is, as the intake biomarkers and the ongoing physiological reactions triggered by this intake-thereby enable evaluation of the metabolic basis required to distinguish the different metabotypes. The capacity of nontargeted metabolomics to also encompass the unprecedented metabolite species has enabled the profiling of multiple metabolites and the corresponding metabotypes with a single analysis, decoding the complex interplay between diet, other relevant factors, and health. In this systematic review, we screened 345 articles published in English in January 2007-July 2018, which applied the metabolomics approach to profile the changes of endogenous metabolites in the blood related to dietary interventions, either derived by metabolism of gut microbiota or the human host. We excluded all the compounds that were directly derived from diet, and also the dietary interventions focusing on supplementation with individual compounds. After the removal of less relevant studies and assessment of eligibility, 49 articles were included in this review. First, we mention the contribution of individual factors, either modifiable or nonmodifiable factors, in shaping metabolic profile. Then, how different aspects of the diet would affect the metabolic profiles are disentangled. Next, the classes of endogenous metabolites altered following included dietary interventions are listed. We also discuss the current challenges in the field, along with future research opportunities.
Collapse
Affiliation(s)
- Stefania Noerman
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland,Address correspondence to SN (e-mail: )
| | - Marjukka Kolehmainen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Kati Hanhineva
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland,Address correspondence to KH ()
| |
Collapse
|
111
|
Effect of Lifestyle Intervention in the Concentration of Adipoquines and Branched Chain Amino Acids in Subjects with High Risk of Developing Type 2 Diabetes: Feel4Diabetes Study. Cells 2020; 9:cells9030693. [PMID: 32178221 PMCID: PMC7140606 DOI: 10.3390/cells9030693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
Introduction: The global prevalence of type 2 diabetes (T2D) is increasing rapidly, especially in low- and middle-income countries and has a high number of associated comorbidities. Plasmatic concentrations of branched chain amino acids (BCAA) and retinol-binding protein 4 (RBP4) have been shown to be elevated in T2D subjects in cross-sectional studies. However, the effect of lifestyle community-based interventions on BCAA and RBP4 concentrations has not yet been analyzed. Material and methods: The Feel4Diabetes study is a school and community-based intervention that identified 360 European families with a high risk of developing T2D according to the FINDRISC questionnaire. Families were randomized in control and intervention groups were followed-up from 2016 to 2018. In the Spanish families, the concentration of BCAA and RBP4 was determined in 266 subjects (115 control and 151 intervention group) that attended the three time-point assessments by colorimetric and ELISA reaction, respectively. Results: Baseline BCAA levels showed positive correlations with the FINDRISC score and glucose impairment (baseline glucose, insulin, and glycated hemoglobin), body mass index, and body weight. The participants receiving the community-based intervention showed a significant decrease in glycated hemoglobin and BCAA levels compared to the control group (p = 0.011 and p < 0.001, respectively). However, baseline RBP4 did not show significant correlations with anthropometric and glycemic parameters, and no significant change was observed in anthropometric parameters and RBP4 concentrations throughout the follow-up. Conclusion: A community-based intervention on lifestyle led to a significant reduction in BCAA levels regardless of weight loss. These findings suggest that this interventional approach could be promising in T2D prevention.
Collapse
|
112
|
Hosseinpour-Niazi S, Tahmasebinejad Z, Esfandiar Z, Bakhshi B, Mirmiran P, Azizi F. Weight gain, but not macronutrient intake, modifies the effect of dietary branch chain amino acids on the risk of metabolic syndrome. Diabetes Res Clin Pract 2020; 161:108039. [PMID: 32007512 DOI: 10.1016/j.diabres.2020.108039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 01/20/2020] [Accepted: 01/27/2020] [Indexed: 12/28/2022]
Abstract
AIMS The aim of this study was to investigate whether both weight change and the background intakes of macronutrient modulate the association between dietary branch chain amino acids (BCAAs) and the risk of metabolic syndrome (MetS). METHODS This prospective study was conducted within the framework of theTehranLipidand Glucose Study. BCAA intakes were collected using a valid and reliable semi-quantitative food frequency questionnaire. MetS components were defined according to the modified national Cholesterol Education Program Adult Treatment Panel III. Weight change was categorized as weight gain (≥ or <7% over 8.9 year follow-up). Dietary fat and carbohydrate intake were categorized as above/below the median intake. RESULTS Among participants with weight gain ≥ 7% during follow-up, intakes of both dietary BCAAs and its various sources (below or above the median intake) were associated with higher risk of MetS, compared with subjects with lower intakes of BCAAs and weight change ≤ 7%. Background dietary fat and carbohydrate did not modify the association of dietary BCAAs and its various sources with the risk of MetS. CONCLUSIONS Weight change, but not dietary macronutrient intake, modulates the association between dietary BCAAs and risk of MetS among adults.
Collapse
Affiliation(s)
- Somayeh Hosseinpour-Niazi
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zhale Tahmasebinejad
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohre Esfandiar
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahar Bakhshi
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
113
|
Satheesh G, Ramachandran S, Jaleel A. Metabolomics-Based Prospective Studies and Prediction of Type 2 Diabetes Mellitus Risks. Metab Syndr Relat Disord 2019; 18:1-9. [PMID: 31634052 DOI: 10.1089/met.2019.0047] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The preceding decade has witnessed an intense upsurge in the diabetic population across the world making type 2 diabetes mellitus (T2DM) more of an epidemic than a lifestyle disease. Metabolic disorders are often latent for a while before becoming clinically evident, thus reinforcing the pursuit of early biomarkers of metabolic alterations. A prospective study along with metabolic profiling is the most appropriate way to detect the early pathophysiological changes in metabolic diseases such as T2DM. The aim of this review was to summarize the different potential biomarkers of T2DM identified in prospective studies, which used tools of metabolomics. The review also demonstrates on how metabolomic profiling-based prospective studies can be used to address a concern like population-specific disease mechanism. We performed a literature search on metabolomics-based prospective studies on T2DM using the key words "metabolomics," "Type 2 diabetes," "diabetes mellitus", "metabolite profiling," "prospective study," "metabolism," and "biomarker." Additional articles that were obtained from the reference lists of the articles obtained using the above key words were also examined. Articles on dietary intake, type 1 diabetes mellitus, and gestational diabetes were excluded. The review revealed that many studies showed a direct association of branched-chain amino acids and an inverse association of glycine with T2DM. Majority of the prospective studies conducted were targeted metabolomics-based, with Caucasians as their study cohort. The whole disease risk in populations, including Asians, could therefore not be identified. This review proposes the utility of prospective studies in conjunction with metabolomics platform to unravel the altered metabolic pathways that contribute to the risk of T2DM.
Collapse
Affiliation(s)
- Gopika Satheesh
- Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | - Abdul Jaleel
- Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
114
|
Wang J, Liu Y, Lian K, Shentu X, Fang J, Shao J, Chen M, Wang Y, Zhou M, Sun H. BCAA Catabolic Defect Alters Glucose Metabolism in Lean Mice. Front Physiol 2019; 10:1140. [PMID: 31551816 PMCID: PMC6738029 DOI: 10.3389/fphys.2019.01140] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/20/2019] [Indexed: 01/01/2023] Open
Abstract
Recent studies show branched-chain amino acid (BCAA) catabolic pathway is defective in obese animals and humans, contributing to the pathogenesis of insulin resistance and diabetes. However, in the context of obesity, various processes including the dysfunctional lipid metabolism can affect insulin sensitivity and glycemic regulation. It remains unclear how BCAA catabolic defect may exert direct impacts on glucose metabolism without the disturbance of obesity. The current study characterized the glucose metabolism in lean mice in which the genetic deletion of PP2Cm leads to moderate BCAA catabolic defect. Interestingly, compared to the wildtype control, lean PP2Cm deficient mice showed enhanced insulin sensitivity and glucose tolerance, lower body weight, and the preference for carbohydrate over lipids utilization. Metabolomics profiling of plasma and tissues revealed significantly different metabolic patterns in the PP2Cm deficient mice, featured by the marked alterations in glucose metabolic processes, including gluconeogenesis/glycolysis, glycogen metabolism, and tricarboxylic acid cycle. The metabolic changes of glucose were predominantly observed in liver but not skeletal muscle or white adipose tissue. The elevated branched-chain keto acids (BCKAs) resulted from the BCAA catabolic defect may play a critical role in regulating the expression of key regulators of glucose metabolic processes and the activity of respiratory Complex II/succinate dehydrogenase in TCA cycle. Together, these results show BCAA catabolic defect significantly alters glucose metabolism in lean mice with some impacts different or even opposite from those in obese mice, highlighting the critical role of BCAA catabolism in glycemic regulation and the complex interplay between macronutrients in lean and obese animals.
Collapse
Affiliation(s)
- Ji Wang
- Department of Pathophysiology, Hongqiao International Institute of Medicine, Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunxia Liu
- Department of Pathophysiology, Hongqiao International Institute of Medicine, Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Lian
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinyi Shentu
- Department of Pathophysiology, Hongqiao International Institute of Medicine, Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junwei Fang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Shao
- Department of Pathophysiology, Hongqiao International Institute of Medicine, Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengping Chen
- Department of Pathophysiology, Hongqiao International Institute of Medicine, Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yibin Wang
- Departments of Anesthesiology, Medicine and Physiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Meiyi Zhou
- Department of Pathophysiology, Hongqiao International Institute of Medicine, Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haipeng Sun
- Department of Pathophysiology, Hongqiao International Institute of Medicine, Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
115
|
Zhou M, Shao J, Wu CY, Shu L, Dong W, Liu Y, Chen M, Wynn RM, Wang J, Wang J, Gui WJ, Qi X, Lusis AJ, Li Z, Wang W, Ning G, Yang X, Chuang DT, Wang Y, Sun H. Targeting BCAA Catabolism to Treat Obesity-Associated Insulin Resistance. Diabetes 2019; 68:1730-1746. [PMID: 31167878 PMCID: PMC6702639 DOI: 10.2337/db18-0927] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 05/29/2019] [Indexed: 12/12/2022]
Abstract
Recent studies implicate a strong association between elevated plasma branched-chain amino acids (BCAAs) and insulin resistance (IR). However, a causal relationship and whether interrupted BCAA homeostasis can serve as a therapeutic target for diabetes remain to be established experimentally. In this study, unbiased integrative pathway analyses identified a unique genetic link between obesity-associated IR and BCAA catabolic gene expression at the pathway level in human and mouse populations. In genetically obese (ob/ob) mice, rate-limiting branched-chain α-keto acid (BCKA) dehydrogenase deficiency (i.e., BCAA and BCKA accumulation), a metabolic feature, accompanied the systemic suppression of BCAA catabolic genes. Restoring BCAA catabolic flux with a pharmacological inhibitor of BCKA dehydrogenase kinase (BCKDK) ( a suppressor of BCKA dehydrogenase) reduced the abundance of BCAA and BCKA and markedly attenuated IR in ob/ob mice. Similar outcomes were achieved by reducing protein (and thus BCAA) intake, whereas increasing BCAA intake did the opposite; this corroborates the pathogenic roles of BCAAs and BCKAs in IR in ob/ob mice. Like BCAAs, BCKAs also suppressed insulin signaling via activation of mammalian target of rapamycin complex 1. Finally, the small-molecule BCKDK inhibitor significantly attenuated IR in high-fat diet-induced obese mice. Collectively, these data demonstrate a pivotal causal role of a BCAA catabolic defect and elevated abundance of BCAAs and BCKAs in obesity-associated IR and provide proof-of-concept evidence for the therapeutic validity of manipulating BCAA metabolism for treating diabetes.
Collapse
Affiliation(s)
- Meiyi Zhou
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Shao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-Yang Wu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Le Shu
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, CA
| | - Weibing Dong
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunxia Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengping Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - R Max Wynn
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji Wang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Jun Gui
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Xiangbing Qi
- Chemistry Center, National Institute of Biological Science, Beijing, China
| | - Aldons J Lusis
- Departments of Medicine, Microbiology, and Human Genetics, University of California at Los Angeles, Los Angeles, CA
| | - Zhaoping Li
- Department of Clinical Nutrition, University of California at Los Angeles, Los Angeles, CA
| | - Weiqing Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, CA
| | - David T Chuang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yibin Wang
- Departments of Anesthesiology, Medicine, and Physiology, University of California at Los Angeles, Los Angeles, CA
| | - Haipeng Sun
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Departments of Anesthesiology, Medicine, and Physiology, University of California at Los Angeles, Los Angeles, CA
| |
Collapse
|
116
|
Chen Z, Newgard CB, Kim JS, IIkayeva O, Alderete TL, Thomas DC, Berhane K, Breton C, Chatzi L, Bastain TM, McConnell R, Avol E, Lurmann F, Muehlbauer MJ, Hauser ER, Gilliland FD. Near-roadway air pollution exposure and altered fatty acid oxidation among adolescents and young adults - The interplay with obesity. ENVIRONMENT INTERNATIONAL 2019; 130:104935. [PMID: 31238265 PMCID: PMC6679991 DOI: 10.1016/j.envint.2019.104935] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/06/2019] [Accepted: 06/14/2019] [Indexed: 05/22/2023]
Abstract
BACKGROUND Air pollution exposure has been shown to increase the risk of obesity and metabolic dysfunction in animal models and human studies. However, the metabolic pathways altered by air pollution exposure are unclear, especially in adolescents and young adults who are at a critical period in the development of cardio-metabolic diseases. OBJECTIVES The aim of this study was to examine the associations between air pollution exposure and indices of fatty acid and amino acid metabolism. METHODS A total of 173 young adults (18-23 years) from eight Children's Health Study (CHS) Southern California communities were examined from 2014 to 2018. Near-roadway air pollution (NRAP) exposure (freeway and non-freeway) and regional air pollution exposure (nitrogen dioxide, ozone and particulate matter) during one year before the study visit were estimated based on participants' residential addresses. Serum concentrations of 64 targeted metabolites including amino acids, acylcarnitines, non-esterified fatty acid (NEFA) and glycerol were measured in fasting serum samples. Principal component analysis of metabolites was performed to identify metabolite clusters that represent key metabolic pathways. Mixed effects models were used to analyze the associations of air pollution exposure with metabolomic principal component (PC) scores and individual metabolite concentrations adjusting for potential confounders. RESULTS Higher lagged one-year averaged non-freeway NRAP exposure was associated with higher concentrations of NEFA oxidation byproducts and higher NEFA-related PC score (all p's ≤ 0.038). The effect sizes were larger among obese individuals (interaction p = 0.047). Among females, higher freeway NRAP exposure was also associated with a higher NEFA-related PC score (p = 0.042). Among all participants, higher freeway NRAP exposure was associated with a lower PC score for lower concentrations of short- and median-chain acylcarnitines (p = 0.044). CONCLUSIONS Results of this study indicate that NRAP exposure is associated with altered fatty acid metabolism, which could contribute to the metabolic perturbation in obese youth.
Collapse
Affiliation(s)
- Zhanghua Chen
- Division of Environmental Health, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| | - Christopher B Newgard
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center Durham, NC, USA
| | - Jeniffer S Kim
- Division of Environmental Health, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Olga IIkayeva
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center Durham, NC, USA
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA
| | - Duncan C Thomas
- Division of Biostatistics, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Kiros Berhane
- Division of Biostatistics, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Carrie Breton
- Division of Environmental Health, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Leda Chatzi
- Division of Environmental Health, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Theresa M Bastain
- Division of Environmental Health, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Rob McConnell
- Division of Environmental Health, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Edward Avol
- Division of Environmental Health, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | | | - Michael J Muehlbauer
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center Durham, NC, USA
| | - Elizabeth R Hauser
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center Durham, NC, USA
| | - Frank D Gilliland
- Division of Environmental Health, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
117
|
Vallianou N, Stratigou T, Christodoulatos GS, Dalamaga M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Obesity and Obesity-Associated Metabolic Disorders: Current Evidence and Perspectives. Curr Obes Rep 2019; 8:317-332. [PMID: 31175629 DOI: 10.1007/s13679-019-00352-2] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE In this review, we summarize current evidence on the gut microbiome and microbial metabolites in relation to obesity and obesity-associated metabolic disorders. Special emphasis is given on mechanisms interconnecting gut microbiome and microbial metabolites with metabolic disorders as well as on potential preventive and therapeutic perspectives with a "bench to bedside" approach. RECENT FINDINGS Recent data have highlighted the role of gut dysbiosis in the etiology and pathogenesis of metabolic disorders, including obesity, metabolic syndrome, type 2 diabetes mellitus, and non-alcoholic fatty liver disease. Overall, most studies have demonstrated a reduction in gut microbiome diversity and richness in obese subjects, but there is still much debate on the exact microbial signature of a healthy or an obese gut microbiome. Despite the controversial role of an altered gut microbiome as a cause or consequence of obesity in human studies, numerous animal studies and certain human studies suggest beneficial metabolic effects of certain microbial intestinal metabolites, such as butyrate, that could be used in the prevention and treatment of obesity and its comorbidities. More randomized controlled trials and larger prospective studies including well-defined cohorts as well as a multi-omics approach are warranted to better identify the associations between the gut microbiome, microbial metabolites, and obesity and its metabolic complications.
Collapse
Affiliation(s)
- Natalia Vallianou
- Department of Endocrinology, Evangelismos General Hospital of Athens, 45-47 Ypsilantou street, 10676, Athens, Greece
| | - Theodora Stratigou
- Department of Endocrinology, Evangelismos General Hospital of Athens, 45-47 Ypsilantou street, 10676, Athens, Greece
| | - Gerasimos Socrates Christodoulatos
- Laboratory of Microbiology, KAT Hospital, 2 Nikis, Kifisia, 14561, Athens, Greece
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias #27, 75 Mikras Asias, Goudi, 11527, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias #27, 75 Mikras Asias, Goudi, 11527, Athens, Greece.
| |
Collapse
|
118
|
Abstract
The cause of insulin resistance in obesity and type 2 diabetes mellitus (T2DM) is not limited to impaired insulin signalling but also involves the complex interplay of multiple metabolic pathways. The analysis of large data sets generated by metabolomics and lipidomics has shed new light on the roles of metabolites such as lipids, amino acids and bile acids in modulating insulin sensitivity. Metabolites can regulate insulin sensitivity directly by modulating components of the insulin signalling pathway, such as insulin receptor substrates (IRSs) and AKT, and indirectly by altering the flux of substrates through multiple metabolic pathways, including lipogenesis, lipid oxidation, protein synthesis and degradation and hepatic gluconeogenesis. Moreover, the post-translational modification of proteins by metabolites and lipids, including acetylation and palmitoylation, can alter protein function. Furthermore, the role of the microbiota in regulating substrate metabolism and insulin sensitivity is unfolding. In this Review, we discuss the emerging roles of metabolites in the pathogenesis of insulin resistance and T2DM. A comprehensive understanding of the metabolic adaptations involved in insulin resistance may enable the identification of novel targets for improving insulin sensitivity and preventing, and treating, T2DM.
Collapse
|
119
|
Ribeiro RV, Solon-Biet SM, Pulpitel T, Senior AM, Cogger VC, Clark X, O'Sullivan J, Koay YC, Hirani V, Blyth FM, Seibel MJ, Waite LM, Naganathan V, Cumming RG, Handelsman DJ, Simpson SJ, Le Couteur DG. Of Older Mice and Men: Branched-Chain Amino Acids and Body Composition. Nutrients 2019; 11:E1882. [PMID: 31412601 PMCID: PMC6723310 DOI: 10.3390/nu11081882] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/06/2019] [Accepted: 08/06/2019] [Indexed: 01/02/2023] Open
Abstract
Protein and branched-chain amino acid (BCAA) intake are associated with changes in circulating BCAAs and influence metabolic health in humans and rodents. However, the relationship between BCAAs and body composition in both species is unclear, with many studies questioning the translatability of preclinical findings to humans. Here, we assessed and directly compared the relationship between circulating BCAAs, body composition, and intake in older mice and men. Body weight and body fat were positively associated with circulating BCAA levels in both mouse and human, which remained significant after adjustments for age, physical activity, number of morbidities, smoking status, and source of income in the human cohort. Macronutrient intakes were similarly associated with circulating BCAA levels; however, the relationship between protein intake and BCAAs were more pronounced in the mice. These findings indicate that the relationship between circulating BCAAs, body composition, and intakes are comparable in both species, suggesting that the mouse is an effective model for examining the effects of BCAAs on body composition in older humans.
Collapse
Affiliation(s)
- Rosilene V Ribeiro
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney 2006, Australia.
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia.
| | - Samantha M Solon-Biet
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney 2006, Australia.
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia.
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney 2006, Australia.
| | - Tamara Pulpitel
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney 2006, Australia
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
| | - Alistair M Senior
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney 2006, Australia
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney 2006, Australia
| | - Ximonie Clark
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney 2006, Australia
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
| | - John O'Sullivan
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
- Heart Research Institute, The University of Sydney, Sydney 2006, Australia
| | - Yen Chin Koay
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
- Heart Research Institute, The University of Sydney, Sydney 2006, Australia
| | - Vasant Hirani
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney 2006, Australia
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
- ARC Centre of Excellence in Population Ageing Research (CEPAR), Kensington 2033, Australia
| | - Fiona M Blyth
- Concord Clinical School, Faculty of Health and Medicine, The University of Sydney, Concord 2139, Australia
| | - Markus J Seibel
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney 2006, Australia
- Concord Clinical School, Faculty of Health and Medicine, The University of Sydney, Concord 2139, Australia
- ANZAC Research Institute, The University of Sydney, Concord 2139, Australia
| | - Louise M Waite
- ARC Centre of Excellence in Population Ageing Research (CEPAR), Kensington 2033, Australia
| | - Vasi Naganathan
- Concord Clinical School, Faculty of Health and Medicine, The University of Sydney, Concord 2139, Australia
| | - Robert G Cumming
- School of Public Health, University of Sydney, Sydney 2006, Australia
- ARC Centre of Excellence in Population Ageing Research (CEPAR), Kensington 2033, Australia
| | - David J Handelsman
- ANZAC Research Institute, The University of Sydney, Concord 2139, Australia
| | - Stephen J Simpson
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney 2006, Australia
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
| | - David G Le Couteur
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney 2006, Australia
- Charles Perkins Centre, Camperdown, The University of Sydney, Sydney 2006, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney 2006, Australia
- Ageing and Alzheimers Institute, Concord Hospital, University of Sydney, Concord 2139, Australia
| |
Collapse
|
120
|
Herring Milt Protein Hydrolysate Improves Insulin Resistance in High-Fat-Diet-Induced Obese Male C57BL/6J Mice. Mar Drugs 2019; 17:md17080456. [PMID: 31382619 PMCID: PMC6724050 DOI: 10.3390/md17080456] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/27/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022] Open
Abstract
Protein consumption influences glucose homeostasis, but the effect depends on the type and origin of proteins ingested. The present study was designed to determine the effect of herring milt protein hydrolysate (HPH) on insulin function and glucose metabolism in a mouse model of diet-induced obesity. Male C57BL/6J mice were pretreated with a low-fat diet or a high-fat diet for 6 weeks. Mice on the high-fat diet were divided into four groups where one group continued on the high-fat diet and the other three groups were fed a modified high-fat diet where 15%, 35%, and 70%, respectively, of casein was replaced with an equal percentage of protein derived from HPH. After 10 weeks, mice that continued on the high-fat diet showed significant increases in body weight, blood glucose, insulin, and leptin levels and exhibited impaired oral glucose tolerance, insulin resistance, and pancreatic β-cell dysfunction. Compared to mice fed the high-fat diet, the 70% replacement of dietary casein with HPH protein reduced body weight, semi-fasting blood glucose, fasting blood glucose, insulin, leptin, and cholesterol levels and improved glucose tolerance, homeostasis model assessment of insulin resistance (HOMA-IR), and homeostasis model assessment of β-cell function (HOMA-β) indices. The 35% replacement of dietary casein with HPH protein showed moderate effects, while the 15% replacement of dietary casein with HPH protein had no effects. This is the first study demonstrating that replacing dietary casein with the same amount of protein derived from HPH can prevent high-fat-diet-induced obesity and insulin resistance.
Collapse
|
121
|
Gene-Environment Interactions on Body Fat Distribution. Int J Mol Sci 2019; 20:ijms20153690. [PMID: 31357654 PMCID: PMC6696304 DOI: 10.3390/ijms20153690] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 02/08/2023] Open
Abstract
The prevalence of obesity has been increasing markedly in the U.S. and worldwide in the past decades; and notably, the obese populations are signified by not only the overall elevated adiposity but also particularly harmful accumulation of body fat in the central region of the body, namely, abdominal obesity. The profound shift from “traditional” to “obesogenic” environments, principally featured by the abundance of palatable, energy-dense diet, reduced physical activity, and prolonged sedentary time, promotes the obesity epidemics and detrimental body fat distribution. Recent advances in genomics studies shed light on the genetic basis of obesity and body fat distribution. In addition, growing evidence from investigations in large cohorts and clinical trials has lent support to interactions between genetic variations and environmental factors, e.g., diet and lifestyle factors, in relation to obesity and body fat distribution. This review summarizes the recent discoveries from observational studies and randomized clinical trials on the gene–environment interactions on obesity and body fat distribution.
Collapse
|
122
|
Zhuang Y, Xing C, Cao H, Zhang C, Luo J, Guo X, Hu G. Insulin resistance and metabonomics analysis of fatty liver haemorrhagic syndrome in laying hens induced by a high-energy low-protein diet. Sci Rep 2019; 9:10141. [PMID: 31300671 PMCID: PMC6626135 DOI: 10.1038/s41598-019-46183-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/24/2019] [Indexed: 12/31/2022] Open
Abstract
Fatty liver haemorrhagic syndrome (FLHS) is a widespread metabolic disease in laying hens that causes a decrease in egg production and even death. Insulin resistance is a major contributor to the pathogenesis of nonalcoholic fatty liver disease. However, the relationship between FLHS and the insulin resistance mechanisms underlying FLHS is not well elucidated. Therefore, we established an FLHS model induced by feeding a high-energy low-protein diet. In the current study, we found that the fasting glucose and insulin concentrations were elevated in the FLHS group compared with the control group during the experimental period. The results of the oral glucose tolerance test (OGTT) and insulin sensitivity test (IST) showed a high level of insulin resistance in the FLHS model. InsR, 4EBP-1, Glut-1 and Glut-3 mRNA expression were decreased, and TOR, S6K1, and FOXO1 were elevated (P < 0.05). Metabolomic analysis with GC/MS identified 46 differentially expressed metabolites between these two groups, and of these, 14 kinds of metabolism molecules and 32 kinds of small metabolism molecules were decreased (P < 0.05). Further investigation showed that glucose, lipid and amino acid metabolism blocks in the progression of FLHS by GO functional and pathway analysis. Overall, these results suggest that insulin resistance participated in FLHS; comprehensively, metabolites participated in the dysregulated biological process.
Collapse
Affiliation(s)
- Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Chenghong Xing
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Junrong Luo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, 330045, Jiangxi, P.R. China.
| |
Collapse
|
123
|
Pre-meal protein intake alters postprandial plasma metabolome in subjects with metabolic syndrome. Eur J Nutr 2019; 59:1881-1894. [PMID: 31280343 DOI: 10.1007/s00394-019-02039-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/29/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE We examined the effect on the postprandial plasma metabolome of protein pre-meals before a fat-rich main meal. METHODS Two randomized, cross-over meal studies were conducted to test the dose-response effect (0 g, 10 g, 20 g) of a pre-meal with whey protein (WP) (PREMEAL I), and the effect of protein quality (10 g WP, casein, or gluten) and timing (- 15 min vs - 30 min) of the pre-meal (PREMEAL II). Participants with metabolic syndrome received one of the test meals on each test day, - 15 min (or - 30 min) prior to a standardized fat-rich breakfast. Plasma samples were collected at - 15 min (or - 30 min), 0, 120, 240 a nd 360 min and analyzed using liquid chromatography-mass spectrometry with an untargeted method. RESULTS Pre-meal WP intake elevated plasma branched-chain amino acids (BCAA), aromatic amino acids and methionine and decreased plasma LPC (16:0) and PC (32:1) levels before the main meal. Early (- 15 to 0 min) aromatic amino acids and BCAA in response to pre-meal WP partially predict the glucose and insulin response after the main meal. A pre-meal with WP altered the postprandial plasma metabolic pattern of acyl-carnitines, specific PCs, LPCs and LPEs, betaine, citric acid, linoleic acid, and β-hydroxypalmitic acid compared to no pre-meal. The casein and WP pre-meals exhibited similar postprandial amino acid responses whereas a pre-meal with gluten resulted in lower levels of plasma amino acids and its metabolites. CONCLUSION A pre-meal with protein affects the postprandial metabolic pattern indicating facilitated glucose and lipid disposal from plasma in participants with metabolic syndrome.
Collapse
|
124
|
Arneth B, Arneth R, Shams M. Metabolomics of Type 1 and Type 2 Diabetes. Int J Mol Sci 2019; 20:ijms20102467. [PMID: 31109071 PMCID: PMC6566263 DOI: 10.3390/ijms20102467] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Type 1 and type 2 diabetes mellitus (DM) are chronic diseases that affect nearly 425 million people worldwide, leading to poor health outcomes and high health care costs. High-throughput metabolomics screening can provide vital insight into the pathophysiological pathways of DM and help in managing its effects. The primary aim of this study was to contribute to the understanding and management of DM by providing reliable evidence of the relationships between metabolites and type 1 diabetes (T1D) and metabolites and type 2 diabetes (T2D). Information for the study was obtained from the PubMed, MEDLINE, and EMBASE databases, and leads to additional articles that were obtained from the reference lists of the studies examined. The results from the selected studies were used to assess the relationships between diabetes (T1D and/or T2D) and metabolite markers—such as glutamine, glycine, and aromatic amino acids—in patients. Seventy studies were selected from the three databases and from the reference lists in the records retrieved. All studies explored associations between various metabolites and T1D or T2D. This review identified several plasma metabolites associated with T2D prediabetes and/or T1D and/or T2D in humans. The evidence shows that metabolites such as glucose, fructose, amino acids, and lipids are typically altered in individuals with T1D and T2D. These metabolites exhibit significant predictive associations with T2D prediabetes, T1D, and/or T2D. The current review suggests that changes in plasma metabolites can be identified by metabolomic techniques and used to identify and analyze T1D and T2D biomarkers. The results of the metabolomic studies can be used to help create effective interventions for managing these diseases.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany.
| | - Rebekka Arneth
- Clinics for Internal Medicine 2, University Hospital of the Universities of Giessen and Marburg UKGM, Justus Liebig University. Giessen, 35392 Giessen, Germany.
| | - Mohamed Shams
- Department of Pharmacy Practice, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
125
|
Biswas D, Duffley L, Pulinilkunnil T. Role of branched‐chain amino acid–catabolizing enzymes in intertissue signaling, metabolic remodeling, and energy homeostasis. FASEB J 2019; 33:8711-8731. [DOI: 10.1096/fj.201802842rr] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Dipsikha Biswas
- Department of Biochemistry and Molecular Biology Faculty of Medicine Dalhousie Medicine New Brunswick Dalhousie University Saint John New Brunswick Canada
| | - Luke Duffley
- Department of Biochemistry and Molecular Biology Faculty of Medicine Dalhousie Medicine New Brunswick Dalhousie University Saint John New Brunswick Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology Faculty of Medicine Dalhousie Medicine New Brunswick Dalhousie University Saint John New Brunswick Canada
| |
Collapse
|
126
|
Guasch-Ferré M, Ruiz-Canela M, Li J, Zheng Y, Bulló M, Wang DD, Toledo E, Clish C, Corella D, Estruch R, Ros E, Fitó M, Arós F, Fiol M, Lapetra J, Serra-Majem L, Liang L, Papandreou C, Dennis C, Martínez-González MA, Hu FB, Salas-Salvadó J. Plasma Acylcarnitines and Risk of Type 2 Diabetes in a Mediterranean Population at High Cardiovascular Risk. J Clin Endocrinol Metab 2019; 104:1508-1519. [PMID: 30423132 PMCID: PMC6435097 DOI: 10.1210/jc.2018-01000] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/08/2018] [Indexed: 01/14/2023]
Abstract
CONTEXT The potential associations between acylcarnitine profiles and incidence of type 2 diabetes (T2D) and whether acylcarnitines can be used to improve diabetes prediction remain unclear. OBJECTIVE To evaluate the associations between baseline and 1-year changes in acylcarnitines and their diabetes predictive ability beyond traditional risk factors. DESIGN, SETTING, AND PARTICIPANTS We designed a case-cohort study within the PREDIMED Study including all incident cases of T2D (n = 251) and 694 randomly selected participants at baseline (follow-up, 3.8 years). Plasma acylcarnitines were measured using a targeted approach by liquid chromatography-tandem mass spectrometry. We tested the associations between baseline and 1-year changes in individual acylcarnitines and T2D risk using weighted Cox regression models. We used elastic net regressions to select acylcarnitines for T2D prediction and compute a weighted score using a cross-validation approach. RESULTS An acylcarnitine profile, especially including short- and long-chain acylcarnitines, was significantly associated with a higher risk of T2D independent of traditional risk factors. The relative risks of T2D per SD increment of the predictive model scores were 4.03 (95% CI, 3.00 to 5.42; P < 0.001) for the conventional model and 4.85 (95% CI, 3.65 to 6.45; P < 0.001) for the model including acylcarnitines, with a hazard ratio of 1.33 (95% CI, 1.08 to 1.63; P < 0.001) attributed to the acylcarnitines. Including the acylcarnitines into the model did not significantly improve the area under the receiver operator characteristic curve (0.86 to 0.88, P = 0.61). A 1-year increase in C4OH-carnitine was associated with higher risk of T2D [per SD increment, 1.44 (1.03 to 2.01)]. CONCLUSIONS An acylcarnitine profile, mainly including short- and long-chain acylcarnitines, was significantly associated with higher T2D risk in participants at high cardiovascular risk. The inclusion of acylcarnitines into the model did not significantly improve the T2D prediction C-statistics beyond traditional risk factors, including fasting glucose.
Collapse
Affiliation(s)
- Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Miguel Ruiz-Canela
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- University of Navarra, Department of Preventive Medicine and Public Health, Pamplona, Spain
- Health Research Institute of Navarra, Pamplona, Spain
| | - Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Mònica Bulló
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Estefanía Toledo
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- University of Navarra, Department of Preventive Medicine and Public Health, Pamplona, Spain
- Health Research Institute of Navarra, Pamplona, Spain
| | - Clary Clish
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | - Dolores Corella
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Ramon Estruch
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Hospital Clinic, August Pi Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Emilio Ros
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Montserrat Fitó
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular and Nutrition Research Group (REGICOR Study Group), Hospital del Mar Research Institute, Barcelona, Spain
| | - Fernando Arós
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Cardiology, Organización Sanitaria Integrada (OSI) ARABA, Universidad del País Vasco/Euskal Herriko Univertsitatea (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Miquel Fiol
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Institute of Health Sciences (Institut Universitari d’Investigació en Ciències de la Salut-IUNICS), University of Balearic Islands and Hospital Son Espases, Palma de Mallorca, Spain
| | - José Lapetra
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, Sevilla, Spain
| | - Lluís Serra-Majem
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Research Institute of Biomedical and Health Sciences, University of Las Palmas de Gran Canaria and Service of Preventive Medicine, Complejo Hospitalario Universitario Insular Materno Infantil (CHUIMI), Canary Health Service, Las Palmas de Gran Canaria, Spain
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Christopher Papandreou
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Courtney Dennis
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | - Miguel A Martínez-González
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- University of Navarra, Department of Preventive Medicine and Public Health, Pamplona, Spain
- Health Research Institute of Navarra, Pamplona, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jordi Salas-Salvadó
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
127
|
Solon-Biet SM, Cogger VC, Pulpitel T, Wahl D, Clark X, Bagley E, Gregoriou GC, Senior AM, Wang QP, Brandon AE, Perks R, O’Sullivan J, Koay YC, Bell-Anderson K, Kebede M, Yau B, Atkinson C, Svineng G, Dodgson T, Wali JA, Piper MDW, Juricic P, Partridge L, Rose AJ, Raubenheimer D, Cooney GJ, Le Couteur DG, Simpson SJ. Branched chain amino acids impact health and lifespan indirectly via amino acid balance and appetite control. Nat Metab 2019; 1:532-545. [PMID: 31656947 PMCID: PMC6814438 DOI: 10.1038/s42255-019-0059-2] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/22/2019] [Indexed: 12/11/2022]
Abstract
Elevated branched chain amino acids (BCAAs) are associated with obesity and insulin resistance. How long-term dietary BCAAs impact late-life health and lifespan is unknown. Here, we show that when dietary BCAAs are varied against a fixed, isocaloric macronutrient background, long-term exposure to high BCAA diets leads to hyperphagia, obesity and reduced lifespan. These effects are not due to elevated BCAA per se or hepatic mTOR activation, but rather due to a shift in the relative quantity of dietary BCAAs and other AAs, notably tryptophan and threonine. Increasing the ratio of BCAAs to these AAs resulted in hyperphagia and is associated with central serotonin depletion. Preventing hyperphagia by calorie restriction or pair-feeding averts the health costs of a high BCAA diet. Our data highlight a role for amino acid quality in energy balance and show that health costs of chronic high BCAA intakes need not be due to intrinsic toxicity but, rather, a consequence of hyperphagia driven by AA imbalance.
Collapse
Affiliation(s)
- Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
- Ageing and Alzheimers Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord NSW, Australia
- ANZAC Research Institute, The University of Sydney NSW, Australia
| | - Tamara Pulpitel
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - Devin Wahl
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
- Ageing and Alzheimers Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord NSW, Australia
| | - Ximonie Clark
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Elena Bagley
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Medical Sciences, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - Gabrielle C Gregoriou
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Medical Sciences, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Qiao-Ping Wang
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Guangzhou 510275, China
| | - Amanda E Brandon
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - Ruth Perks
- Charles Perkins Centre, The University of Sydney NSW, Australia
| | - John O’Sullivan
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Heart Research Institute, The University of Sydney, NSW, Australia
| | - Yen Chin Koay
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Heart Research Institute, The University of Sydney, NSW, Australia
| | - Kim Bell-Anderson
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Melkam Kebede
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Belinda Yau
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Clare Atkinson
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | | | - Timothy Dodgson
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Jibran A Wali
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | | | - Paula Juricic
- Max Planck Institute for Biology of Aging, Cologne, Germany
| | | | - Adam J Rose
- Monash Biomedicine Discovery Institute, Monash University VIC, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Gregory J Cooney
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - David G Le Couteur
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
- Ageing and Alzheimers Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord NSW, Australia
- ANZAC Research Institute, The University of Sydney NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| |
Collapse
|
128
|
Zhou W, Wang Y, Yang F, Dong Q, Wang H, Hu N. Rapid Determination of Amino Acids of Nitraria tangutorum Bobr. from the Qinghai-Tibet Plateau Using HPLC-FLD-MS/MS and a Highly Selective and Sensitive Pre-Column Derivatization Method. Molecules 2019; 24:E1665. [PMID: 31035340 PMCID: PMC6539371 DOI: 10.3390/molecules24091665] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/22/2019] [Accepted: 04/26/2019] [Indexed: 11/16/2022] Open
Abstract
Amino acids are indispensable components of living organisms. The high amino acid content in Nitraria tangutorum Bobr. fruit distinguishes it from other berry plants and is of great significance to its nutritional value. Herein, using 10-ethyl-acridine-3-sulfonyl chloride as a fluorescent pre-column labeling reagent, a method for the efficient and rapid determination of amino acid content in N. tangutorum by pre-column fluorescence derivatization and on-line mass spectrometry was established and further validated. The limits of detection (signal-to-noise ratio = 3) were between 0.13 and 1.13 nmol/L, with a linear coefficient greater than 0.997 and a relative standard deviation between 1.37% and 2.64%. In addition, the method required a short analysis time, separating 19 amino acids within 20 min. Subsequently, the method was used to analyze the amino acid content of Nitraria tangutorum Bobr. from tissues retrieved from seven regions of the Qinghai-Tibet Plateau. Nitraria tangutorum Bobr. was shown to contain a large amount of amino acids, with the total content and main amino acid varying between the different tissues. This research supports the nutritional evaluation, quality control, and development and utilization of Nitraria tangutorum Bobr.
Collapse
Affiliation(s)
- Wu Zhou
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China.
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China.
| | - Yuwei Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China.
| | - Fang Yang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China.
| | - Qi Dong
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China.
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining 810008, China.
| | - Honglun Wang
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China.
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining 810008, China.
| | - Na Hu
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China.
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining 810008, China.
| |
Collapse
|
129
|
Metabolomics Identifies Novel Blood Biomarkers of Pulmonary Function and COPD in the General Population. Metabolites 2019; 9:metabo9040061. [PMID: 30939782 PMCID: PMC6523962 DOI: 10.3390/metabo9040061] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 12/26/2022] Open
Abstract
Determination of metabolomic signatures of pulmonary function and chronic obstructive pulmonary disease (COPD) in the general population could aid in identification and understanding of early disease processes. Metabolome measurements were performed on serum from 4742 individuals (2354 African-Americans and 1529 European-Americans from the Atherosclerosis Risk in Communities study and 859 Europeans from the Cooperative Health Research in the Region of Augsburg study). We examined 368 metabolites in relation to cross-sectional measures of forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC), their ratio (FEV1/FVC) and COPD using multivariable regression followed by meta-analysis. At a false discovery rate of 0.05, 95 metabolites were associated with FEV1 and 100 with FVC (73 overlapping), including inverse associations with branched-chain amino acids and positive associations with glutamine. Ten metabolites were associated with FEV1/FVC and seventeen with COPD (393 cases). Enriched pathways of amino acid metabolism were identified. Associations with FEV1 and FVC were not driven by individuals with COPD. We identified novel metabolic signatures of pulmonary function and COPD in African and European ancestry populations. These may allow development of biomarkers in the general population of early disease pathogenesis, before pulmonary function has decreased to levels diagnostic for COPD.
Collapse
|
130
|
Amino acid transporters in the regulation of insulin secretion and signalling. Biochem Soc Trans 2019; 47:571-590. [PMID: 30936244 DOI: 10.1042/bst20180250] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 01/02/2023]
Abstract
Amino acids are increasingly recognised as modulators of nutrient disposal, including their role in regulating blood glucose through interactions with insulin signalling. More recently, cellular membrane transporters of amino acids have been shown to form a pivotal part of this regulation as they are primarily responsible for controlling cellular and circulating amino acid concentrations. The availability of amino acids regulated by transporters can amplify insulin secretion and modulate insulin signalling in various tissues. In addition, insulin itself can regulate the expression of numerous amino acid transporters. This review focuses on amino acid transporters linked to the regulation of insulin secretion and signalling with a focus on those of the small intestine, pancreatic β-islet cells and insulin-responsive tissues, liver and skeletal muscle. We summarise the role of the amino acid transporter B0AT1 (SLC6A19) and peptide transporter PEPT1 (SLC15A1) in the modulation of global insulin signalling via the liver-secreted hormone fibroblast growth factor 21 (FGF21). The role of vesicular vGLUT (SLC17) and mitochondrial SLC25 transporters in providing glutamate for the potentiation of insulin secretion is covered. We also survey the roles SNAT (SLC38) family and LAT1 (SLC7A5) amino acid transporters play in the regulation of and by insulin in numerous affective tissues. We hypothesise the small intestine amino acid transporter B0AT1 represents a crucial nexus between insulin, FGF21 and incretin hormone signalling pathways. The aim is to give an integrated overview of the important role amino acid transporters have been found to play in insulin-regulated nutrient signalling.
Collapse
|
131
|
Novin ZS, Ghavamzadeh S, Mehdizadeh A. The Weight Loss Effects of Branched Chain Amino Acids and Vitamin B6: A Randomized Controlled Trial on Obese and Overweight Women. INT J VITAM NUTR RES 2019; 88:80-89. [PMID: 30841823 DOI: 10.1024/0300-9831/a000511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Branched chain amino acids (BCAA), with vitamin B6 have been reported to improve fat metabolism and muscle synthesis. We hypothesized that supplementation with BCAA and vitamin B6 would result in more weight loss and improve body composition and blood markers related to cardiovascular diseases. Our aim was to determine whether the mentioned supplementation would affect weight loss, body composition, and cardiovascular risk factors during weight loss intervention. To this end, we performed a placebo-controlled randomized clinical trial in 42 overweight and obese women (BMI = 25-34.9 kg/m2). Taking a four-week moderate deficit calorie diet (-500 kcal/day), participants were randomized to receive BCAA (6 g/day) with vitamin B6 (40 mg/day) or placebo. Body composition variables measured with the use of bioelectrical impedance analysis, homeostatic model assessment, and plasma insulin, Low density lipoprotein, High density lipoprotein, Total Cholesterol, Triglyceride, and fasting blood sugar were measured. The result indicated that, weight loss was not significantly affected by BCAA and vitamin B6 supplementation (-2.43 ± 1.02 kg) or placebo (-1.64 ± 1.48 kg). However, significant time × treatment interactions in waist to hip ratio (P = 0.005), left leg lean (P = 0.004) and right leg lean (P = 0.023) were observed. Overall, supplementation with BCAA and vitamin B6 could preserve legs lean and also attenuated waist to hip ratio.
Collapse
|
132
|
Delplancke TDJ, Wu Y, Han TL, Joncer LR, Qi H, Tong C, Baker PN. Metabolomics of Pregnancy Complications: Emerging Application of Maternal Hair. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2815439. [PMID: 30662903 PMCID: PMC6312607 DOI: 10.1155/2018/2815439] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/18/2018] [Indexed: 02/01/2023]
Abstract
In recent years, the study of metabolomics has begun to receive increasing international attention, especially as it pertains to medical research. This is due in part to the potential for discovery of new biomarkers in the metabolome and to a new understanding of the "exposome", which refers to the endogenous and exogenous compounds that reflect external exposures. Consequently, metabolomics research into pregnancy-related issues has increased. Biomarkers discovered through metabolomics may shed some light on the etiology of certain pregnancy-related complications and their adverse effects on future maternal health and infant development and improve current clinical management. The discoveries and methods used in these studies will be compiled and summarized within the following paper. A further focus of this paper is the use of hair as a biological sample, which is gaining increasing attention across diverse fields due to its noninvasive sampling method and the metabolome stability. Its significance in exposome studies will be considered in this review, as well as the potential to associate exposures with adverse pregnancy outcomes. Currently, hair has been used in only two metabolomics studies relating to fetal growth restriction (FGR) and gestational diabetes mellitus (GDM).
Collapse
Affiliation(s)
- Thibaut D. J. Delplancke
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Yue Wu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Ting-Li Han
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Lingga R. Joncer
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Chao Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing Medical University, Chongqing 400016, China
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Philip N. Baker
- International Collaborative Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
- Liggins Institute, University of Auckland, Auckland, New Zealand
- College of Medicine, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
133
|
Flores-Guerrero JL, Osté MCJ, Kieneker LM, Gruppen EG, Wolak-Dinsmore J, Otvos JD, Connelly MA, Bakker SJL, Dullaart RPF. Plasma Branched-Chain Amino Acids and Risk of Incident Type 2 Diabetes: Results from the PREVEND Prospective Cohort Study. J Clin Med 2018; 7:jcm7120513. [PMID: 30518023 PMCID: PMC6306832 DOI: 10.3390/jcm7120513] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/21/2018] [Accepted: 11/27/2018] [Indexed: 01/06/2023] Open
Abstract
Plasma branched-chain amino acids (BCAAs) are linked to metabolic disease, but their relevance for prediction of type 2 diabetes development is unclear. We determined the association of plasma BCAAs with type 2 diabetes risk in the prevention of renal and vascular end-stage disease (PREVEND) cohort. The BCAAs were measured by means of nuclear magnetic resonance spectroscopy. We evaluated the prospective associations of BCAAs with type 2 diabetes in 6244 subjects. The BCAAs were positively associated with HOMA-IR after multivariable adjustment (p < 0.0001). During median follow-up for 7.5 years, 301 cases of type 2 diabetes were ascertained. The Kaplan-Meier plot demonstrated that patients in the highest BCAA quartile presented a higher risk (p log-rank < 0.001). Cox regression analyses revealed a positive association between BCAA and type 2 diabetes; the hazard ratio (HR) for the highest quartile was 6.15 (95% CI: 4.08, 9.24, p < 0.0001). After adjustment for multiple clinical and laboratory variables, the association remained (HR 2.80 (95% CI: 1.72, 4.53), p < 0.0001). C-statistics, Net reclassification improvement, and −2 log likelihood were better after adding BCAAs to the traditional risk model (p = 0.01 to <0.001). In conclusions, high concentrations of BCAAs associate with insulin resistance and with increased risk of type 2 diabetes. This association is independent of multiple risk factors, HOMA-IR and β cell function.
Collapse
Affiliation(s)
- Jose L Flores-Guerrero
- Department of Internal Medicine, UMCG, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Maryse C J Osté
- Department of Internal Medicine, UMCG, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Lyanne M Kieneker
- Department of Internal Medicine, UMCG, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Eke G Gruppen
- Department of Endocrinology, UMCG, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | | | - James D Otvos
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC 27560, USA.
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC 27560, USA.
| | - Stephan J L Bakker
- Department of Internal Medicine, UMCG, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Robin P F Dullaart
- Department of Endocrinology, UMCG, University of Groningen, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
134
|
Lee S, Kwak JH, Kim SH, Yun J, Cho JY, Kim K, Hwang D, Jung YS. A comparison of metabolomic changes in type-1 diabetic C57BL/6N mice originating from different sources. Lab Anim Res 2018; 34:232-238. [PMID: 30671110 PMCID: PMC6333615 DOI: 10.5625/lar.2018.34.4.232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 11/21/2022] Open
Abstract
Animal models have been used to elucidate the pathophysiology of varying diseases and to provide insight into potential targets for therapeutic intervention. Although alternatives to animal testing have been proposed to help overcome potential drawbacks related to animal experiments and avoid ethical issues, their use remains vital for the testing of new drug candidates and to identify the most effective strategies for therapeutic intervention. Particularly, the study of metabolic diseases requires the use of animal models to monitor whole-body physiology. In line with this, the National Institute of Food and Drug Safety Evaluation (NIFDS) in Korea has established their own animal strains to help evaluate both efficacy and safety during new drug development. The objective of this study was to characterize the response of C57BL/6NKorl mice from the NIFDS compared with that of other mice originating from the USA and Japan in a chemical-induced diabetic condition. Multiple low-dose treatments with streptozotocin were used to generate a type-1 diabetic animal model which is closely linked to the known clinical pathology of this disease. There were no significantly different responses observed between the varying streptozotocin-induced type-1 diabetic models tested in this study. When comparing control and diabetic mice, increases in liver weight and disturbances in serum amino acids levels of diabetic mice were most remarkable. Although the relationship between type-1 diabetes and BCAA has not been elucidated in this study, the results, which reveal a characteristic increase in diabetic mice of all origins are considered worthy of further study.
Collapse
Affiliation(s)
- Seunghyun Lee
- College of Pharmacy, Pusan National University, Busan, Korea
| | - Jae-Hwan Kwak
- College of Pharmacy, Brain Busan 21 Plus Program, Kyungsung University, Busan, Korea
| | - Sou Hyun Kim
- College of Pharmacy, Pusan National University, Busan, Korea
| | - Jieun Yun
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju, Korea
| | - Joon-Yong Cho
- Department of Health and Exercise Science, Korea National Sport University, Seoul, Korea
| | - Kilsoo Kim
- College of Veterinary Medicine, Kyungpook National University, Daegu, Korea
| | - Daeyeon Hwang
- College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan, Korea
| |
Collapse
|
135
|
Zhang W, Wu Y, Fan W, Chen H, Du H, Rao J. The pattern of plasma BCAA concentration and liver Bckdha gene expression in GK rats during T2D progression. Animal Model Exp Med 2018; 1:305-313. [PMID: 30891580 PMCID: PMC6388062 DOI: 10.1002/ame2.12038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/04/2018] [Accepted: 09/26/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND This study was conducted to measure the concentration of branched chain amino acid (BCAA) in different species and detect the expression pattern of the liver Bckdha gene in Goto-Kakizaki (GK) rats during type 2 diabetes (T2D) progression. METHODS We measured the concentration of BCAA in GK rats, induced T2D cynomolgus monkeys and T2D humans by liquid chromatography tandem mass spectrometry, and used real-time quantitative PCR to analyze the gene expression of Bckdha and Bckdk, which encode the rate-limiting enzymes in catabolism of, respectively, branched chain amino acids and branched chain α-keto acid dehydrogenase kinase. RESULTS In this study, we showed that GK rat BCAA concentrations were significantly reduced at 4 and 8 weeks (P < 0.05 and P < 0.01, respectively), while the expression of Bckdha in GK rat liver was increased at 4 and 8 weeks (1.62-fold and 1.93-fold, respectively). The BCAA concentrations were significantly reduced in diet-induced T2D cynomolgus monkeys (P < 0.01), but significantly increased in T2D humans (P < 0.001). CONCLUSIONS Our results showed that BCAA concentrations changed at different times and by different amounts in different species and during different periods of T2D progress, and the significant changes of BCAA concentration in the three species indicated that BCAA might participate in the progress of T2D. The results suggested that the increased expression of Bckdha in GK rat liver might partially explain the reduced plasma BCAA concentration at 4 and 8 weeks. Further studies are required to investigate the exact mechanism of BCAA changes in non-obese T2D.
Collapse
Affiliation(s)
- Wenlu Zhang
- School of Biological and Biological EngineeringSouth China University of TechnologyGuangzhouChina
| | - Yu'e Wu
- Guangdong Key Laboratory of Laboratory AnimalsGuangzhouChina
| | - Wei Fan
- School of Biological and Biological EngineeringSouth China University of TechnologyGuangzhouChina
| | | | - Hongli Du
- School of Biological and Biological EngineeringSouth China University of TechnologyGuangzhouChina
| | - Junhua Rao
- Guangdong Key Laboratory of Animal Conservation and Resource UtilizationGuangdong Public Laboratory of Wild Animal Conservation and UtilizationGuangdong Institute of Applied Biological ResourcesGuangzhouChina
| |
Collapse
|
136
|
Calorie restriction and its impact on gut microbial composition and global metabolism. Front Med 2018; 12:634-644. [PMID: 30446879 DOI: 10.1007/s11684-018-0670-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 09/27/2018] [Indexed: 02/08/2023]
Abstract
Calorie restriction (CR) is a dietary regimen that reduces calorie intake without incurring malnutrition or a reduction in essential nutrients. It has long been recognized as a natural strategy for promoting health, extending longevity, and prevents the development of metabolic and age-related diseases. In the present review, we focus on the general effect of CR on gut microbiota composition and global metabolism. We also propose mechanisms for its beneficial effect. Results showed that probiotic and butyrate-producing microbes increased their relative abundance, whereas proinflammatory strains exhibited suppressed relative abundance following CR. Analyses of the gut microbial and host metabolisms revealed that most host microbial co-metabolites were changed due to CR. Examples of dramatic CR-induced changes in host metabolism included a decrease in the rate of lipid biosynthesis and an increase in the rates of fatty acid catabolism, β-oxidation, glycogenolysis, and gluconeogenesis. The observed phenotypes and the further verification of the direct link between gut microbiota and metabolome may benefit patients that are at risk for developing metabolic disease. Thus, improved gut microbiota composition and metabolome are potential biomarkers for determining the effectiveness of dietary interventions for age-related and metabolic diseases.
Collapse
|
137
|
Castro-Barquero S, Lamuela-Raventós RM, Doménech M, Estruch R. Relationship between Mediterranean Dietary Polyphenol Intake and Obesity. Nutrients 2018; 10:nu10101523. [PMID: 30336572 PMCID: PMC6213078 DOI: 10.3390/nu10101523] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/13/2018] [Accepted: 10/14/2018] [Indexed: 12/28/2022] Open
Abstract
Obesity is a multifactorial and complex disease defined by excess of adipose mass and constitutes a serious health problem. Adipose tissue acts as an endocrine organ secreting a wide range of inflammatory adipocytokines, which leads to systemic inflammation, insulin resistance, and metabolic disorders. The traditional Mediterranean diet is characterized by a high phenolic-rich foods intake, including extra-virgin olive oil, nuts, red wine, vegetables, fruits, legumes, and whole-grain cereals. Evidence for polyphenols’ effect on obesity and weight control in humans is inconsistent and the health effects of polyphenols depend on the amount consumed and their bioavailability. The mechanisms involved in weight loss in which polyphenols may have a role are: activating β-oxidation; a prebiotic effect for gut microbiota; inducing satiety; stimulating energy expenditure by inducing thermogenesis in brown adipose tissue; modulating adipose tissue inhibiting adipocyte differentiation; promoting adipocyte apoptosis and increasing lipolysis. Even though the intake of some specific polyphenols has been associated with body weight changes, there is still no evidence for the effects of total polyphenols or some polyphenol subclasses in humans on adiposity.
Collapse
Affiliation(s)
- Sara Castro-Barquero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain.
| | - Rosa M Lamuela-Raventós
- Department of Nutrition, Food Science and Gastronomy, XaRTA, INSA-UB, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- CIBEROBN Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Mónica Doménech
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain.
- CIBEROBN Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Ramon Estruch
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain.
- CIBEROBN Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Internal Medicine Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain.
| |
Collapse
|
138
|
Serum free amino acid levels in rheumatoid arthritis according to therapy and physical disability. Cytokine 2018; 113:332-339. [PMID: 30337216 DOI: 10.1016/j.cyto.2018.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 08/07/2018] [Accepted: 10/01/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND In presented study the amino acid analysis was performed in serum derived from rheumatoid arthritis patients (RA) according to undertaken therapy and classification of physical disability. The results were compared with previously published data. METHODS The levels of 31 free amino acids were determined in 50 serum samples derived from RA subjects and 51 controls. The RA patients were divided into two groups according to the therapy (methotrexate/leflunomide, infliximab/adalimumab/etanercept/tocilizumab, prednisolone/NSAID) and classification of physical disability of the patients. Levels of amino acids were measured by LC-MS/MS. The obtained results were subjected to multivariate statistical tests. RESULTS According to the therapy that was being used, threonine differentiated RA patients treated with methotrexate/leflunomide - infliximab/adalimumab/etanercept/tocilizumab (p = 0.00954) and infliximab/adalimumab/etanercept/tocilizumab - prednisolone/NSAID (p = 0.03109), while tryptophan differentiated RA patients treated with methotrexate/leflunomide - infliximab/adalimumab/etanercept/tocilizumab (p = 0.01723). In the functional classification, arginine differentiated RA samples between class III and IV (p = 0.02332), while glycine differentiated them between class I+II and III of the Steinbrocker functional classification (p = 0.03366). CONCLUSIONS An analysis of the metabolome profile requires the use of validated bioanalytical methods that are strictly dedicated for this purpose. The obtained results are not accidental (p value less than 0.05), and all of the selected amino acids play an important role in inflammation and immune response. It is suggested that studied amino acids can be considered as a markers for diagnosis of RA and monitoring pharmacotherapy of the disease.
Collapse
|
139
|
Haydar S, Paillot T, Fagot C, Cogne Y, Fountas A, Tutuncu Y, Vintila M, Tsatsoulis A, Thanh Chi P, Garandeau P, Chetea D, Badiu C, Gheorghiu M, Ylli D, Lautier C, Jarec M, Monnier L, Normand C, Šarac J, Barakat A, Missoni S, Pugeat M, Poucheret P, Hanzu F, Gomis R, Macias JM, Litvinov S, Khusnutdinova E, Poiana C, Pasquali R, Lauro D, Sesti G, Trischitta V, Abdelhak S, Zenati A, Ylli A, Satman I, Kanninen T, Rinato Y, Grigorescu F. Branched-Chain Amino Acid Database Integrated in MEDIPAD Software as a Tool for Nutritional Investigation of Mediterranean Populations. Nutrients 2018; 10:E1392. [PMID: 30275383 PMCID: PMC6213539 DOI: 10.3390/nu10101392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 12/24/2022] Open
Abstract
Branched-chained amino acids (BCAA) are essential dietary components for humans and can act as potential biomarkers for diabetes development. To efficiently estimate dietary intake, we developed a BCAA database for 1331 food items found in the French Centre d'Information sur la Qualité des Aliments (CIQUAL) food table by compiling BCAA content from international tables, published measurements, or by food similarity as well as by calculating 267 items from Greek, Turkish, Romanian, and Moroccan mixed dishes. The database embedded in MEDIPAD software capable of registering 24 h of dietary recalls (24HDR) with clinical and genetic data was evaluated based on archived 24HDR of the Saint Pierre Institute (France) from 2957 subjects, which indicated a BCAA content up to 4.2 g/100 g of food and differences among normal weight and obese subjects across BCAA quartiles. We also evaluated the database of 119 interviews of Romanians, Turkish and Albanians in Greece (27⁻65 years) during the MEDIGENE program, which indicated mean BCAA intake of 13.84 and 12.91 g/day in males and females, respectively, comparable to other studies. The MEDIPAD is user-friendly, multilingual, and secure software and with the BCAA database is suitable for conducting nutritional assessment in the Mediterranean area with particular facilities for food administration.
Collapse
Affiliation(s)
- Sara Haydar
- Unité Mixte de Recherche (UMR)204 NUTRIPASS (Nutrition et Alimentation des Populations aux Suds, IRD, UM, SupAgro), Molecular Endocrinology, Institut Universitaire de Recherche Clinique (IURC), Faculty of Medicine, University of Montpellier, 34093 Montpellier, France.
| | | | | | - Yannick Cogne
- Unité Mixte de Recherche (UMR)204 NUTRIPASS (Nutrition et Alimentation des Populations aux Suds, IRD, UM, SupAgro), Molecular Endocrinology, Institut Universitaire de Recherche Clinique (IURC), Faculty of Medicine, University of Montpellier, 34093 Montpellier, France.
| | - Athanasios Fountas
- Department of Endocrinology, School of Medicine, University of Ioannina, 45110 Ioannina, Greece.
| | - Yildiz Tutuncu
- Department of Internal Medicine, Istanbul University, 34093 Istanbul, Turkey.
| | - Madalina Vintila
- Department of Endocrinology, Universitatea de Medicina si Farmacie Carol Davila, 011863 Bucharest, Romania.
| | - Agathocles Tsatsoulis
- Department of Endocrinology, School of Medicine, University of Ioannina, 45110 Ioannina, Greece.
| | - Pham Thanh Chi
- Unité Mixte de Recherche (UMR)204 NUTRIPASS (Nutrition et Alimentation des Populations aux Suds, IRD, UM, SupAgro), Molecular Endocrinology, Institut Universitaire de Recherche Clinique (IURC), Faculty of Medicine, University of Montpellier, 34093 Montpellier, France.
| | - Patrick Garandeau
- Unité Mixte de Recherche (UMR)204 NUTRIPASS (Nutrition et Alimentation des Populations aux Suds, IRD, UM, SupAgro), Molecular Endocrinology, Institut Universitaire de Recherche Clinique (IURC), Faculty of Medicine, University of Montpellier, 34093 Montpellier, France.
| | - Dan Chetea
- Nicolae Paulescu National Institute, 020475 Bucharest, Romania.
| | - Corin Badiu
- Department of Endocrinology, Universitatea de Medicina si Farmacie Carol Davila, 011863 Bucharest, Romania.
| | - Monica Gheorghiu
- Department of Endocrinology, Universitatea de Medicina si Farmacie Carol Davila, 011863 Bucharest, Romania.
| | - Dorina Ylli
- Faculty of Medicine, Mjekesise University of Tirana, 1005 Tirana, Albania.
| | - Corinne Lautier
- Unité Mixte de Recherche (UMR)204 NUTRIPASS (Nutrition et Alimentation des Populations aux Suds, IRD, UM, SupAgro), Molecular Endocrinology, Institut Universitaire de Recherche Clinique (IURC), Faculty of Medicine, University of Montpellier, 34093 Montpellier, France.
| | - Morana Jarec
- Institute for Anthropological Research, 10000 Zagreb, Croatia.
| | - Louis Monnier
- Unité Mixte de Recherche (UMR)204 NUTRIPASS (Nutrition et Alimentation des Populations aux Suds, IRD, UM, SupAgro), Molecular Endocrinology, Institut Universitaire de Recherche Clinique (IURC), Faculty of Medicine, University of Montpellier, 34093 Montpellier, France.
| | - Christophe Normand
- Unité Mixte de Recherche (UMR)204 NUTRIPASS (Nutrition et Alimentation des Populations aux Suds, IRD, UM, SupAgro), Molecular Endocrinology, Institut Universitaire de Recherche Clinique (IURC), Faculty of Medicine, University of Montpellier, 34093 Montpellier, France.
| | - Jelena Šarac
- Institute for Anthropological Research, 10000 Zagreb, Croatia.
| | | | - Sasa Missoni
- Institute for Anthropological Research, 10000 Zagreb, Croatia.
| | - Michel Pugeat
- Fédération d'Endocrinologie, Cardio-Neuro Hospital, University Claude Bernard de Lyon 1, 69677 Lyon-Bron, France.
| | - Patrick Poucheret
- Faculty of Pharmacy, UMR 95 Qualisud, University of Montpellier, 34398 Montpellier, France.
| | - Felicia Hanzu
- Institut d'Investigacions Biomediques August Pi i Sunyer, 08036 Barcelona, Spain.
| | - Ramon Gomis
- Institut d'Investigacions Biomediques August Pi i Sunyer, 08036 Barcelona, Spain.
| | | | | | | | - Catalina Poiana
- Department of Endocrinology, Universitatea de Medicina si Farmacie Carol Davila, 011863 Bucharest, Romania.
| | - Renato Pasquali
- Division of Endocrinology, University Alma Mater Studiorum, 40138 Bologna, Italy.
| | - Davide Lauro
- Department of Internal Medicine, Universita degli Studi di Roma Tor Vergata, 00173 Roma, Italy.
| | - Giorgio Sesti
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy.
| | | | - Sonia Abdelhak
- Institut Pasteur de Tunis, Laboratory of Biomedical Genomics and Oncogenetics, 1002 Tunis, Tunisia.
| | - Akila Zenati
- Laboratoire de Biochimie Génétique, CHU Bab-El-Oued, Université d'Alger, Alger 16000, Algeria.
| | - Agron Ylli
- Faculty of Medicine, Mjekesise University of Tirana, 1005 Tirana, Albania.
| | - Ilhan Satman
- Department of Internal Medicine, Istanbul University, 34093 Istanbul, Turkey.
| | | | - Yves Rinato
- Intactile Design SA, 34000 Montpellier, France.
| | - Florin Grigorescu
- Unité Mixte de Recherche (UMR)204 NUTRIPASS (Nutrition et Alimentation des Populations aux Suds, IRD, UM, SupAgro), Molecular Endocrinology, Institut Universitaire de Recherche Clinique (IURC), Faculty of Medicine, University of Montpellier, 34093 Montpellier, France.
| |
Collapse
|
140
|
Preconception Micronutrient Supplementation Reduced Circulating Branched Chain Amino Acids at 12 Weeks Gestation in an Open Trial of Guatemalan Women Who Are Overweight or Obese. Nutrients 2018; 10:nu10091282. [PMID: 30208589 PMCID: PMC6165402 DOI: 10.3390/nu10091282] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/04/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022] Open
Abstract
Elevated branched chain amino acids (BCAAs: valine, leucine, and isoleucine) are well-established biomarkers of obesity-associated insulin resistance (IR). Mounting evidence suggests that low- and middle-income countries are suffering from a "double burden" of both undernutrition (growth stunting) and overnutrition (obesity) as these countries undergo a "nutrition transition". The purpose of this study was to examine if pre-pregnancy body mass index (BMI, kg/m²) and a daily lipid-based micronutrient supplement (LNS, Nutriset) would lead to cross-sectional differences in circulating levels of branched chain amino acids (BCAAs) in Guatemalan women experiencing short stature during early pregnancy. Using data from an ongoing randomized controlled trial, Women First, we studied women who were normal weight (NW, BMI range for this cohort = 20.1⁻24.1 kg/m²) or overweight/obese (OW/OB, BMI range for this cohort = 25.6⁻31.9 kg/m²), and divided into two groups: those who received daily LNS ≥ 3 months prior to conception through 12 weeks gestation (+LNS), or no LNS (-LNS) (n = 9⁻10/group). BCAAs levels were obtained from dried blood spot card samples (DBS) assessed at 12 weeks gestation. DBS cards provide a stable, efficient, and reliable means of collecting, transporting, and storing blood samples in low resource or field settings. Circulating maternal leptin, adiponectin, and insulin were determined by immunoassays from serum samples collected at 12 weeks gestation. We found maternal pre-pregnancy body mass index (ppBMI) was associated with higher circulating BCAAs (r² = 0.433, p = 0.002) and higher leptin/adiponectin ratio (r = 0.466, p = 0.044) in -LNS mothers at 12 weeks gestation. +LNS mothers demonstrated no correlations between BCAAs or leptin/adiponectin ratio across ppBMI suggesting LNS may be effective at improving metabolic status in OW/OB mothers during early pregnancy.
Collapse
|
141
|
Wang CY, Neil DL, Home P. 2020 vision - An overview of prospects for diabetes management and prevention in the next decade. Diabetes Res Clin Pract 2018; 143:101-112. [PMID: 29944968 DOI: 10.1016/j.diabres.2018.06.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/24/2018] [Accepted: 06/13/2018] [Indexed: 02/08/2023]
Abstract
After a century of medical progress, people nowadays live longer with diabetes than ever before. However, current preventative approaches, compounded in part by increased life-expectancy, are failing to reduce the prevalence of diabetes. Cardiovascular sequelae account for many of the four million deaths annually attributable to diabetes. Evidence indicates that certain glucose-lowering medications can improve vascular outcomes in some people with type 2 diabetes, which, together with better understanding of using multiple therapies concurrently, offers opportunities for beneficial personalization of medication regimens. However, further well-designed long-term studies are needed to evaluate cardiovascular benefits and safety of new and older medications, particularly in users typical of everyday diabetes care. Although there are numerous other promising advances in pharmacotherapies and biotechnology, these will probably be unaffordable for most people with diabetes globally. Therefore, effective national public health approaches will be essential to reducing the incidence of diabetes and its associated burdens; these may entail politically controversial measures to change unhealthy lifestyle behaviours. Stakeholders could learn from past failures and emulate successes in other health-care initiatives. Without early action at all levels, we face a future in which approaching one-quarter of humans will have diabetes, with more than half afflicted during their lifetime.
Collapse
Affiliation(s)
- Chih-Yuan Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - David L Neil
- Scientific Development Department, Content Ed Net, Taipei, Taiwan
| | - Philip Home
- Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
142
|
Abstract
PURPOSE OF REVIEW Elevations in circulating branched chain amino acids (BCAAs) have gained attention as potential contributors to the development of insulin resistance and diabetes. RECENT FINDINGS Epidemiological evidence strongly supports this conclusion. Suppression of BCAA catabolism in adipose and hepatic tissues appears to be the primary drivers of plasma BCAA elevations. BCAA catabolism may be shunted to skeletal muscle, where it indirectly leads to FA accumulation and insulin resistance, via a number of proposed mechanisms. BCAAs have an important role in the development of IR, but our understanding of how plasma BCAA elevations occur, and how these elevations lead to insulin resistance, is still limited.
Collapse
Affiliation(s)
- Zoltan Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, TRC 11-106 3400 Civic Blvd, Philadelphia, PA, 19104, USA.
| | - Michael Neinast
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, TRC 11-106 3400 Civic Blvd, Philadelphia, PA, 19104, USA
| |
Collapse
|
143
|
Just S, Mondot S, Ecker J, Wegner K, Rath E, Gau L, Streidl T, Hery-Arnaud G, Schmidt S, Lesker TR, Bieth V, Dunkel A, Strowig T, Hofmann T, Haller D, Liebisch G, Gérard P, Rohn S, Lepage P, Clavel T. The gut microbiota drives the impact of bile acids and fat source in diet on mouse metabolism. MICROBIOME 2018; 6:134. [PMID: 30071904 PMCID: PMC6091023 DOI: 10.1186/s40168-018-0510-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/02/2018] [Indexed: 05/06/2023]
Abstract
BACKGROUND As the gut microbiota contributes to metabolic health, it is important to determine specific diet-microbiota interactions that influence host metabolism. Bile acids and dietary fat source can alter phenotypes of diet-induced obesity, but the interplay with intestinal microorganisms is unclear. Here, we investigated metabolic consequences of diets enriched in primary bile acids with or without addition of lard or palm oil, and studied gut microbiota structure and functions in mice. RESULTS In combination with bile acids, dietary lard fed to male C57BL/6N mice for a period of 8 weeks enhanced fat mass accumulation in colonized, but not in germ-free mice when compared to palm oil. This was associated with impaired glucose tolerance, lower fasting insulin levels, lower counts of enteroendocrine cells, fatty liver, and elevated amounts of hepatic triglycerides, cholesteryl esters, and monounsaturated fatty acids. Lard- and bile acid-fed mice were characterized by shifts in dominant gut bacterial communities, including decreased relative abundances of Lachnospiraceae and increased occurrence of Desulfovibrionaceae and the species Clostridium lactatifermentans and Flintibacter butyricus. Metatranscriptomic analysis revealed shifts in microbial functions, including lipid and amino acid metabolism. CONCLUSIONS Caution is required when interpreting data from diet-induced obesity models due to varying effects of dietary fat source. Detrimental metabolic consequences of a diet enriched with lard and primary bile acids were dependent on microbial colonization of the host and were linked to hepatic lipid rearrangements and to alterations of dominant bacterial communities in the cecum.
Collapse
Affiliation(s)
- Sarah Just
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Stanislas Mondot
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Josef Ecker
- Nutritional Physiology, Technical University of Munich, Freising, Germany
| | - Katrin Wegner
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Hamburg, Germany
| | - Eva Rath
- Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Laura Gau
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Hamburg, Germany
| | - Theresa Streidl
- Institute of Medical Microbiology, Functional Microbiome Research Group, University Hospital of RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Genevieve Hery-Arnaud
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Sinah Schmidt
- Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising, Germany
| | - Till Robin Lesker
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Valentin Bieth
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Andreas Dunkel
- Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising, Germany
| | - Till Strowig
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Hofmann
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
- Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising, Germany
| | - Dirk Haller
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
- Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Philippe Gérard
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Sascha Rohn
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Hamburg, Germany
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en-Josas, France
| | - Thomas Clavel
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany.
- Institute of Medical Microbiology, Functional Microbiome Research Group, University Hospital of RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
144
|
Wang J, Xu S, Gao J, Zhang L, Zhang Z, Yang W, Li Y, Liao S, Zhou H, Liu P, Liang B. SILAC-based quantitative proteomic analysis of the livers of spontaneous obese and diabetic rhesus monkeys. Am J Physiol Endocrinol Metab 2018; 315:E294-E306. [PMID: 29664677 DOI: 10.1152/ajpendo.00016.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a severe metabolic disorder that affects more than 10% of the population worldwide. Obesity is a major cause of insulin resistance and contributes to the development of T2DM. Liver is an essential metabolic organ that plays crucial roles in the pathogenesis of obesity and diabetes. However, the underlying mechanisms of liver in the transition of obesity to diabetes are not fully understood. The nonhuman primate rhesus monkey is an appropriate animal for research of human diseases. Here, we first screened and selected three individuals of spontaneously diabetic rhesus monkeys. Interestingly, the diabetic monkeys were obese with a high body mass index at the beginning, but gradually lost their body weight during one year of observation. Furthermore, we performed stable isotope labeling with amino acids in cell culture-based quantitative proteomics to identify proteins and signaling pathways with altered expression in the liver of obese and diabetic monkeys. In total, 3,509 proteins were identified and quantified, of which 185 proteins displayed an altered expression level. Gene ontology analysis revealed that the expression of proteins involved in fatty acids β-oxidation and galactose metabolism was increased in obese monkeys; while proteins involved in oxidative phosphorylation and branched chain amino acid (BCAA) degradation were upregulated in diabetic monkeys. In addition, we observed mild apoptosis in the liver of diabetic monkeys, suggesting liver injury at the late onset of diabetes. Taken together, our liver proteomics may reveal a distinct metabolic transition from fatty acids β-oxidation in obese monkey to BCAA degradation in diabetic monkeys.
Collapse
Affiliation(s)
- Junlong Wang
- College of Pharmaceutical Sciences, Soochow University , Suzhou , China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing , China
| | - Jing Gao
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai , China
| | - Linqiang Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Zhiguo Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Wenhui Yang
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Department of Geriatrics, Yan'an Affiliated Hospital of Kunming Medical University , Kunming , China
| | - Yunhai Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Shasha Liao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai , China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing , China
| | - Bin Liang
- College of Pharmaceutical Sciences, Soochow University , Suzhou , China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| |
Collapse
|
145
|
Lin W, Liu Z, Zheng X, Chen M, Gao D, Tian Z. High-salt diet affects amino acid metabolism in plasma and muscle of Dahl salt-sensitive rats. Amino Acids 2018; 50:1407-1414. [DOI: 10.1007/s00726-018-2615-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/03/2018] [Indexed: 10/28/2022]
|
146
|
Defect of branched-chain amino acid metabolism promotes the development of Alzheimer's disease by targeting the mTOR signaling. Biosci Rep 2018; 38:BSR20180127. [PMID: 29802157 PMCID: PMC6028749 DOI: 10.1042/bsr20180127] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 01/10/2023] Open
Abstract
Diabetes is a risk factor for Alzheimer’s disease (AD) in humans. Branched-chain amino acids (BCAAs, namely valine, leucine, and isoleucine) metabolic defect is observed in human diabetes, which is associated with insulin resistance. But whether BCAAs connect diabetes and AD remains unknown. Here, we show that BCAA metabolic defect may be one of the drivers of AD. BCAA levels were increased in the blood in human patients and mice with diabetes or AD. BCAA-enriched diet promoted the development of AD in mice as evidenced by the behavior and pathological analysis. Branched-chain amino acid transaminase 1 and 2 (BCAT1 and BCAT2) are the two enzymes for the first step metabolism of BCAAs by catalyzing BCAAs to generate branched-chain ketoacids. The expression of Bcat1 but not Bcat2 was significantly down-regulated in the brain tissues of diabetic, aged, and AD mice. Leucine up-regulated the phosphorylation of Tau but not affected the accumulation of amyloid β in the brain tissues or isolated neurons. In addition, knockdown of the expression of Bcat1, which would result in the accumulation of BCAAs, led to the same phenotype as BCAAs supplement in neurons. Interestingly, leucine supplement or Bcat1 knockdown promoted the activation of the mTOR signaling in the brains of AD mice or neurons. Subsequently, mTOR was critically involved in leucine and Bcat1 knockdown-mediated phosphorylation of Tau. Taken together, our findings demonstrated that diabetes-related BCAA accumulation in the brain tissues led to the phosphorylation of Tau and, subsequently, the development of diabetes-related AD.
Collapse
|
147
|
Welsh P, Rankin N, Li Q, Mark PB, Würtz P, Ala-Korpela M, Marre M, Poulter N, Hamet P, Chalmers J, Woodward M, Sattar N. Circulating amino acids and the risk of macrovascular, microvascular and mortality outcomes in individuals with type 2 diabetes: results from the ADVANCE trial. Diabetologia 2018; 61:1581-1591. [PMID: 29728717 PMCID: PMC6445481 DOI: 10.1007/s00125-018-4619-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/21/2018] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESES We aimed to quantify the association of individual circulating amino acids with macrovascular disease, microvascular disease and all-cause mortality in individuals with type 2 diabetes. METHODS We performed a case-cohort study (N = 3587), including 655 macrovascular events, 342 microvascular events (new or worsening nephropathy or retinopathy) and 632 all-cause mortality events during follow-up, in a secondary analysis of the Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE) study. For this study, phenylalanine, isoleucine, glutamine, leucine, alanine, tyrosine, histidine and valine were measured in stored plasma samples by proton NMR metabolomics. Hazard ratios were modelled per SD increase in each amino acid. RESULTS In models investigating associations and potential mechanisms, after adjusting for age, sex and randomised treatment, phenylalanine was positively, and histidine inversely, associated with macrovascular disease risk. These associations were attenuated to the null on further adjustment for extended classical risk factors (including eGFR and urinary albumin/creatinine ratio). After adjustment for extended classical risk factors, higher tyrosine and alanine levels were associated with decreased risk of microvascular disease (HR 0.78; 95% CI 0.67, 0.91 and HR 0.86; 95% CI 0.76, 0.98, respectively). Higher leucine (HR 0.79; 95% CI 0.69, 0.90), histidine (HR 0.89; 95% CI 0.81, 0.99) and valine (HR 0.79; 95% CI 0.70, 0.88) levels were associated with lower risk of mortality. Investigating the predictive ability of amino acids, addition of all amino acids to a risk score modestly improved classification of participants for macrovascular (continuous net reclassification index [NRI] +35.5%, p < 0.001) and microvascular events (continuous NRI +14.4%, p = 0.012). CONCLUSIONS/INTERPRETATION We report distinct associations between circulating amino acids and risk of different major complications of diabetes. Low tyrosine appears to be a marker of microvascular risk in individuals with type 2 diabetes independently of fundamental markers of kidney function.
Collapse
Affiliation(s)
- Paul Welsh
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK.
| | - Naomi Rankin
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Qiang Li
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | - Patrick B Mark
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Peter Würtz
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Nightingale Health Ltd, Helsinki, Finland
| | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Population Health Science, Bristol Medical School, University of Bristol and Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Michel Marre
- Inserm, UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Assistance Publique Hôpitaux de Paris, Bichat Hospital, DHU FIRE, Department of Diabetology, Endocrinology and Nutrition, Paris, France
- University Paris Diderot, Sorbonne Paris Cité, UFR de Médecine, Paris, France
| | - Neil Poulter
- International Centre for Circulatory Health, Imperial College, London, UK
| | - Pavel Hamet
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada
- Department of Medicine, CRCHUM, Université de Montréal, Montreal, QC, Canada
- Department of Medicine, Gene Medicine Services, CRCHUM, Université de Montréal, Montreal, QC, Canada
| | - John Chalmers
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | - Mark Woodward
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
- The George Institute for Global Health, University of Oxford, Oxford, UK
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| |
Collapse
|
148
|
Bhupathiraju SN, Guasch-Ferré M, Gadgil MD, Newgard CB, Bain JR, Muehlbauer MJ, Ilkayeva OR, Scholtens DM, Hu FB, Kanaya AM, Kandula NR. Dietary Patterns among Asian Indians Living in the United States Have Distinct Metabolomic Profiles That Are Associated with Cardiometabolic Risk. J Nutr 2018; 148:1150-1159. [PMID: 29893901 PMCID: PMC6251517 DOI: 10.1093/jn/nxy074] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/08/2018] [Accepted: 03/23/2018] [Indexed: 12/14/2022] Open
Abstract
Background Recent studies, primarily in non-Hispanic whites, suggest that dietary patterns have distinct metabolomic signatures that may influence disease risk. However, evidence in South Asians, a group with unique dietary patterns and a high prevalence of cardiometabolic risk, is lacking. Objective We investigated the metabolomic profiles associated with 2 distinct dietary patterns among a sample of Asian Indians living in the United States. We also examined the cross-sectional associations between metabolomic profiles and cardiometabolic risk markers. Methods We used cross-sectional data from 145 Asian Indians, aged 45-79 y, in the Metabolic Syndrome and Atherosclerosis in South Asians Living in America (MASALA) pilot study. Metabolomic profiles were measured from fasting serum samples. Usual diet was assessed by using a validated food-frequency questionnaire. We used principal components analysis to derive dietary and metabolomic patterns. We used adjusted general linear regression models to examine associations between dietary patterns, individual food groups, metabolite patterns, and cardiometabolic risk markers. Results We observed 2 major principal components or metabolite clusters, the first comprised primarily of medium- to long-chain acylcarnitines (metabolite pattern 1) and the second characterized by branched-chain amino acids, aromatic amino acids, and short-chain acylcarnitines (metabolite pattern 2). A "Western/nonvegetarian" pattern was significantly and positively associated with metabolite pattern 2 (all participants: β ± SE = 0.180 ± 0.090, P = 0.05; participants without type 2 diabetes: β ± SE = 0.323 ± 0.090, P = 0.0005). In all participants, higher scores on metabolite pattern 2 were adversely associated with measures of glycemia (fasting insulin: β ± SE = 2.91 ± 1.29, P = 0.03; 2-h insulin: β ± SE = 22.1 ± 10.3, P = 0.03; homeostasis model assessment of insulin resistance: β ± SE = 0.94 ± 0.42, P = 0.03), total adiponectin (β ± SE = -1.46 ± 0.47, P = 0.002), lipids (total cholesterol: β ± SE = 7.51 ± 3.45, P = 0.03; triglycerides: β ± SE = 14.4 ± 6.67, P = 0.03), and a radiographic measure of hepatic fat (liver-to-spleen attenuation ratio: β ± SE = -0.83 ± 0.42, P = 0.05). Conclusions Our findings suggest that a "Western/nonvegetarian" dietary pattern is associated with a metabolomic profile that is related to an adverse cardiometabolic profile in Asian Indians. Public health efforts to reduce cardiometabolic disease burden in this high-risk group should focus on consuming a healthy plant-based diet.
Collapse
Affiliation(s)
- Shilpa N Bhupathiraju
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston,
MA
- Channing Division of Network Medicine, Harvard Medical School, Boston, MA
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston,
MA
- Channing Division of Network Medicine, Harvard Medical School, Boston, MA
| | - Meghana D Gadgil
- Division of General Internal Medicine, Department of Medicine, University of
California, San Francisco, San Francisco, CA
| | - Christopher B Newgard
- Sarah W Stedman Nutrition and Metabolism Center, Duke Molecular Physiology
Institute, Duke University Medical Center, Durham, NC
| | - James R Bain
- Sarah W Stedman Nutrition and Metabolism Center, Duke Molecular Physiology
Institute, Duke University Medical Center, Durham, NC
| | - Michael J Muehlbauer
- Sarah W Stedman Nutrition and Metabolism Center, Duke Molecular Physiology
Institute, Duke University Medical Center, Durham, NC
| | - Olga R Ilkayeva
- Sarah W Stedman Nutrition and Metabolism Center, Duke Molecular Physiology
Institute, Duke University Medical Center, Durham, NC
| | | | - Frank B Hu
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston,
MA
- Channing Division of Network Medicine, Harvard Medical School, Boston, MA
| | - Alka M Kanaya
- Division of General Internal Medicine, Department of Medicine, University of
California, San Francisco, San Francisco, CA
| | - Namratha R Kandula
- Division of General Internal Medicine and Geriatrics, Department of Medicine,
Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
149
|
Palau-Rodriguez M, Tulipani S, Marco-Ramell A, Miñarro A, Jauregui O, Gonzalez-Dominguez R, Sanchez-Pla A, Ramos-Molina B, Tinahones FJ, Andres-Lacueva C. Characterization of Metabolomic Profile Associated with Metabolic Improvement after Bariatric Surgery in Subjects with Morbid Obesity. J Proteome Res 2018; 17:2704-2714. [DOI: 10.1021/acs.jproteome.8b00144] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Magali Palau-Rodriguez
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable [CIBERfes], Instituto de Salud Carlos III [ISCIII], 28029 Madrid, Spain
| | - Sara Tulipani
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- Biomedical Research Institute [IBIMA], Service of Endocrinology and Nutrition, Malaga Hospital Complex [Virgen de la Victoria], Campus de Teatinos s/n, 29010 Malaga, Spain
| | - Anna Marco-Ramell
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable [CIBERfes], Instituto de Salud Carlos III [ISCIII], 28029 Madrid, Spain
| | - Antonio Miñarro
- Genetics, Microbiology and Statistics Department, Biology Faculty, University of Barcelona, 08028 Barcelona, Spain
| | - Olga Jauregui
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- Scientific and Technological Centres of the University of Barcelona (CCIT-UB), 08028 Barcelona, Spain
| | - Raul Gonzalez-Dominguez
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable [CIBERfes], Instituto de Salud Carlos III [ISCIII], 28029 Madrid, Spain
| | - Alex Sanchez-Pla
- Genetics, Microbiology and Statistics Department, Biology Faculty, University of Barcelona, 08028 Barcelona, Spain
- Statistics and Bioinformatics Unit, Vall d’Hebron Institut de Recerca [VHIR], 08035 Barcelona, Spain
| | - Bruno Ramos-Molina
- Biomedical Research Institute [IBIMA], Service of Endocrinology and Nutrition, Malaga Hospital Complex [Virgen de la Victoria], Campus de Teatinos s/n, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición [CIBERobn], Instituto de Salud Carlos III [ISCIII], 28029 Barcelona, Spain
| | - Francisco J. Tinahones
- Biomedical Research Institute [IBIMA], Service of Endocrinology and Nutrition, Malaga Hospital Complex [Virgen de la Victoria], Campus de Teatinos s/n, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición [CIBERobn], Instituto de Salud Carlos III [ISCIII], 28029 Barcelona, Spain
| | - Cristina Andres-Lacueva
- Biomarkers & Nutrimetabolomic Lab, Nutrition, Food Science and Gastronomy Department, XaRTA, INSA-UB, Campus Torribera, Pharmacy and Food Science Faculty, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable [CIBERfes], Instituto de Salud Carlos III [ISCIII], 28029 Madrid, Spain
| |
Collapse
|
150
|
White PJ, McGarrah RW, Grimsrud PA, Tso SC, Yang WH, Haldeman JM, Grenier-Larouche T, An J, Lapworth AL, Astapova I, Hannou SA, George T, Arlotto M, Olson LB, Lai M, Zhang GF, Ilkayeva O, Herman MA, Wynn RM, Chuang DT, Newgard CB. The BCKDH Kinase and Phosphatase Integrate BCAA and Lipid Metabolism via Regulation of ATP-Citrate Lyase. Cell Metab 2018; 27:1281-1293.e7. [PMID: 29779826 PMCID: PMC5990471 DOI: 10.1016/j.cmet.2018.04.015] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/27/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022]
Abstract
Branched-chain amino acids (BCAA) are strongly associated with dysregulated glucose and lipid metabolism, but the underlying mechanisms are poorly understood. We report that inhibition of the kinase (BDK) or overexpression of the phosphatase (PPM1K) that regulates branched-chain ketoacid dehydrogenase (BCKDH), the committed step of BCAA catabolism, lowers circulating BCAA, reduces hepatic steatosis, and improves glucose tolerance in the absence of weight loss in Zucker fatty rats. Phosphoproteomics analysis identified ATP-citrate lyase (ACL) as an alternate substrate of BDK and PPM1K. Hepatic overexpression of BDK increased ACL phosphorylation and activated de novo lipogenesis. BDK and PPM1K transcript levels were increased and repressed, respectively, in response to fructose feeding or expression of the ChREBP-β transcription factor. These studies identify BDK and PPM1K as a ChREBP-regulated node that integrates BCAA and lipid metabolism. Moreover, manipulation of the BDK:PPM1K ratio relieves key metabolic disease phenotypes in a genetic model of severe obesity.
Collapse
Affiliation(s)
- Phillip J White
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - Robert W McGarrah
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - Paul A Grimsrud
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Shih-Chia Tso
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wen-Hsuan Yang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Jonathan M Haldeman
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Thomas Grenier-Larouche
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Jie An
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Amanda L Lapworth
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Inna Astapova
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - Sarah A Hannou
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Tabitha George
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Michelle Arlotto
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Lyra B Olson
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Michelle Lai
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Mark A Herman
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA
| | - R Max Wynn
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David T Chuang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Departments of Medicine and Pharmacology & Cancer Biology, Durham, NC 27701, USA.
| |
Collapse
|