51
|
Dai S, Wang Z, Yang Y, Du P, Li X. PM 2.5 induced weight loss of mice through altering the intestinal microenvironment: Mucus barrier, gut microbiota, and metabolic profiling. JOURNAL OF HAZARDOUS MATERIALS 2022; 431:128653. [PMID: 35359108 DOI: 10.1016/j.jhazmat.2022.128653] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 06/14/2023]
Abstract
The experimental evidences linking PM2.5 exposure to weight status disorder and the associated mechanisms were lacked. Here, we demonstrated exposure of 198.52 μg/m3 PM2.5 (Baoji city, China) for 40 days induced body weight loss of male Balb/C mice, and then increased after 14-day recovery. Correspondingly, gut microbiota dysbiosis, ileum metabolism alterations, and histopathological changes of liver and ileum elucidated the underlying mechanism. The richness and function modules of flora in feces significantly reduced after exposure, and the ratios of Bacteroidetes/Firmicutes reduced from 1.58 to 0.79. At genus level, Lactobacillus and Clostridium increased markedly, while Bacteroides and Parabacteroides decreased at day 40. After recovery, Oscillospira became the dominant genus. Additionally, the key metabolites in the ileum mediated by PM2.5 identified by metabolomics included arachidonic acid, prostaglandin H2, prostaglandin F2α, 5(S)-HPETE, AMP, and deoxyadenosine. Accordingly, conjoint analysis between the gut micorbiota and metabolic profiling revealed suppression of Arachidonic acid metabolism, linoleic acid metabolism, and PPAR signaling pathway and stimulation of ABC transporters might contribute to the liver injury, ileum inflammation, and then weight loss of mice. Our findings suggested PM2.5 affected weight status of mice by meditating intestinal microenvironment, and provided new insight for further diagnosis of the air pollution dependent disease.
Collapse
Affiliation(s)
- Shuiping Dai
- National Center for Geriatrics Clinical Medicine Research, Department of Geriatrics and Gerontology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhenglu Wang
- College of Oceanography, Hohai University, Nanjing, Jiangsu 210098, PR China.
| | - Ying Yang
- Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Peng Du
- Beijing Key Laboratory of Urban Hydrological Cycle and Sponge City Technology, College of Water Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Xiqing Li
- Laboratory of Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, PR China
| |
Collapse
|
52
|
Lerman JB, Giamberardino SN, Hernandez AF, Felker GM, Shah SH, McGarrah RW. Plasma metabolites associated with functional and clinical outcomes in heart failure with reduced ejection fraction with and without type 2 diabetes. Sci Rep 2022; 12:9183. [PMID: 35654972 PMCID: PMC9163122 DOI: 10.1038/s41598-022-12973-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/19/2022] [Indexed: 12/18/2022] Open
Abstract
Heart failure with reduced ejection fraction (HFrEF) is increasingly treated with medications for type 2 diabetes mellitus (T2DM). Whether metabolic derangements in HFrEF and T2DM are associated with differential outcomes remains unclear. Therefore, understanding molecular pathways in HFrEF and T2DM and their effects on clinical endpoints is important. The FIGHT trial randomized 300 individuals with HFrEF and a recent HF hospitalization to liraglutide (a GLP-1 receptor agonist) versus placebo to assess effects on mortality, HF rehospitalization, and 6-month change in NT-ProBNP. Although the trial showed no clinical benefit of liraglutide, the trial population was highly enriched for individuals with T2DM. Sixty metabolites were quantified via mass spectrometry in plasma from 254 FIGHT participants (N = 147 (57.9%) with T2DM). Principal components analysis reduced the high number of correlated metabolites into uncorrelated factors. The association of factor levels with 90-day changes in 6-min walk distance (6MWD) and NT-proBNP, and with time to mortality or HF hospitalization were evaluated. There were no changes in metabolite factors according to treatment assignment. However, in analyses stratified by T2DM status, changes in five plasma metabolite factors correlated with changes in functional outcomes beyond adjustment: factor 2 (branched-chain amino acids [BCAA]) correlated with changes in NT-proBNP (ρ = − 0.291, p = 4 × 10–4) and 6MWD (ρ= 0.265, p = 0.011); factor 1 (medium-chain acylcarnitines; ρ = 0.220, p = 0.008), factor 4 (long-chain dicarboxylacylcarnitines; ρ = 0.191, p = 0.019), factor 5 (long-chain acylcarnitines; ρ = 0.198, p = 0.017), and factor 8 (urea cycle metabolites; ρ = − 0.239, p = 4 × 10–3), correlated with change in NT-proBNP. Factor 4 was associated with time-to-event (HR = 1.513 [95% CI 1.208–1.896], p = 3 × 10–4) with a trend towards stronger prognostic effect in T2DM (T2DM: p = 1 × 10–3, non-T2DM: p = 0.1). We identified metabolites of BCAA, urea cycle and fatty acid metabolism as biomarkers of HFrEF outcomes, with observed differences in HFrEF patients with T2DM. Such biomarkers might enable future diagnostic or therapeutic interventions in individuals with HFrEF and T2DM. Trial Registration: Clinicaltrials.gov. Identifier: NCT01800968. First posted: February 28, 2013.
Collapse
Affiliation(s)
- Joseph B Lerman
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie N Giamberardino
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 N. Duke St, Durham, NC, 27701, USA
| | - Adrian F Hernandez
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | - G Michael Felker
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | - Svati H Shah
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.,Duke Molecular Physiology Institute, Duke University School of Medicine, 300 N. Duke St, Durham, NC, 27701, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | - Robert W McGarrah
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA. .,Duke Molecular Physiology Institute, Duke University School of Medicine, 300 N. Duke St, Durham, NC, 27701, USA.
| |
Collapse
|
53
|
Fang S, Wade KH, Hughes DA, Fitzgibbon S, Yip V, Timpson NJ, Corbin LJ. A multivariant recall-by-genotype study of the metabolomic signature of BMI. Obesity (Silver Spring) 2022; 30:1298-1310. [PMID: 35598895 PMCID: PMC9324973 DOI: 10.1002/oby.23441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study estimated the effect of BMI on circulating metabolites in young adults using a recall-by-genotype study design. METHODS A recall-by-genotype study was implemented in the Avon Longitudinal Study of Parents and Children. Samples from 756 participants were selected for untargeted metabolomics analysis based on low versus high genetic liability for higher BMI defined by a genetic risk score (GRS). Regression analyses were performed to investigate associations between BMI GRS group and relative abundance of 973 metabolites. RESULTS After correction for multiple testing, 29 metabolites were associated with BMI GRS group. Bilirubin was among the most strongly associated metabolites, with reduced levels measured in individuals in the high-BMI GRS group (β = -0.32, 95% CI: -0.46 to -0.18, Benjamini-Hochberg adjusted p = 0.005). This study observed associations between BMI GRS group and the levels of several potentially diet-related metabolites, including hippurate, which had lower mean abundance in individuals in the high-BMI GRS group (β = -0.29, 95% CI: -0.44 to -0.15, Benjamini-Hochberg adjusted p = 0.008). CONCLUSIONS Together with existing literature, these results suggest that a genetic predisposition to higher BMI captures differences in metabolism leading to adiposity gain. In the absence of prospective data, separating these effects from the downstream consequences of weight gain is challenging.
Collapse
Affiliation(s)
- Si Fang
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Kaitlin H. Wade
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - David A. Hughes
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Sophie Fitzgibbon
- Bristol Bioresource LaboratoriesPopulation Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Vikki Yip
- Bristol Bioresource LaboratoriesPopulation Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Laura J. Corbin
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| |
Collapse
|
54
|
Zhao H, Zheng Y, Zhu L, Xiang L, Xu S, Cai Z. Trimester-specific urinary metabolome alterations associated with gestational diabetes mellitus: A study in different pregnancy stages. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
55
|
Trautman ME, Richardson NE, Lamming DW. Protein restriction and branched-chain amino acid restriction promote geroprotective shifts in metabolism. Aging Cell 2022; 21:e13626. [PMID: 35526271 PMCID: PMC9197406 DOI: 10.1111/acel.13626] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 01/20/2023] Open
Abstract
The proportion of humans suffering from age‐related diseases is increasing around the world, and creative solutions are needed to promote healthy longevity. Recent work has clearly shown that a calorie is not just a calorie—and that low protein diets are associated with reduced mortality in humans and promote metabolic health and extended lifespan in rodents. Many of the benefits of protein restriction on metabolism and aging are the result of decreased consumption of the three branched‐chain amino acids (BCAAs), leucine, isoleucine, and valine. Here, we discuss the emerging evidence that BCAAs are critical modulators of healthy metabolism and longevity in rodents and humans, as well as the physiological and molecular mechanisms that may drive the benefits of BCAA restriction. Our results illustrate that protein quality—the specific composition of dietary protein—may be a previously unappreciated driver of metabolic dysfunction and that reducing dietary BCAAs may be a promising new approach to delay and prevent diseases of aging.
Collapse
Affiliation(s)
- Michaela E. Trautman
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Interdepartmental Graduate Program in Nutritional Sciences University of Wisconsin‐Madison Madison Wisconsin USA
| | - Nicole E. Richardson
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Endocrinology and Reproductive Physiology Graduate Training Program University of Wisconsin‐Madison Madison Wisconsin USA
| | - Dudley W. Lamming
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Endocrinology and Reproductive Physiology Graduate Training Program University of Wisconsin‐Madison Madison Wisconsin USA
| |
Collapse
|
56
|
Sankanagoudar S, Shukla R, Shukla KK, Sharma P. Positive association of branched-chain amino acids with triglyceride and glycated haemoglobin in Indian patients with type 2 diabetes mellitus. Diabetes Metab Syndr 2022; 16:102481. [PMID: 35427914 DOI: 10.1016/j.dsx.2022.102481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/27/2022] [Accepted: 04/05/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Over the past few years, branched-chain amino acids (BCAAs) are increasingly being linked to insulin resistance and type 2 diabetes mellitus (T2DM), but their relevance for metabolic dyslipidaemia in T2DM is unclear. This study aims to determine the plasma and urinary BCAAs and their association with insulin resistance, lipid profile and glycated haemoglobin in patients with T2DM among Indian adults. METHODS In this analytical cross-sectional study, a total of eighty subjects were recruited, 40 T2DM cases and 40 healthy controls. Blood samples collected were subjected to fasting blood sugar (FBS), lipid profile, HbA1c, insulin and BCAAs analysis and urine samples were assessed for BCAAs. All associations were assessed using Spearman Rank Correlation. RESULTS The plasma levels of BCAAs were significantly higher (p < 0.05) in subjects with T2DM than in control subjects. Spearman Rank Correlation analyses revealed a non-significant (p = 0.21) but positive association between BCAAs and homeostasis model assessment of insulin resistance (HOMA-IR) in patients with T2DM (Rho: 0.27). Among lipid profile parameters, only triglycerides had a significant positive correlation to plasma BCAAs in cases (Rho: 0.5971) but not in control subjects. Findings also revealed a significant positive (p < 0.05) association between plasma BCAAs and HbA1c in patients with T2DM (Rho: 0.5325). Urinary BCAAs levels had a non-significant increase in T2DM subjects and did not show any significant correlation with other parameters assessed. CONCLUSION Elevated levels of plasma BCAAs are positively associated with triglyceride and HbA1c. They could serve as an effective marker for the assessment of metabolic dyslipidaemia in subjects with T2DM. Further, large scale studies are needed for confirmation of the same.
Collapse
Affiliation(s)
| | - Ravindra Shukla
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Kamla Kant Shukla
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
57
|
Amino Acid-Related Metabolic Signature in Obese Children and Adolescents. Nutrients 2022; 14:nu14071454. [PMID: 35406066 PMCID: PMC9003189 DOI: 10.3390/nu14071454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
The growing interest in metabolomics has spread to the search for suitable predictive biomarkers for complications related to the emerging issue of pediatric obesity and its related cardiovascular risk and metabolic alteration. Indeed, several studies have investigated the association between metabolic disorders and amino acids, in particular branched-chain amino acids (BCAAs). We have performed a revision of the literature to assess the role of BCAAs in children and adolescents' metabolism, focusing on the molecular pathways involved. We searched on Pubmed/Medline, including articles published until February 2022. The results have shown that plasmatic levels of BCAAs are impaired already in obese children and adolescents. The relationship between BCAAs, obesity and the related metabolic disorders is explained on one side by the activation of the mTORC1 complex-that may promote insulin resistance-and on the other, by the accumulation of toxic metabolites, which may lead to mitochondrial dysfunction, stress kinase activation and damage of pancreatic cells. These compounds may help in the precocious identification of many complications of pediatric obesity. However, further studies are still needed to better assess if BCAAs may be used to screen these conditions and if any other metabolomic compound may be useful to achieve this goal.
Collapse
|
58
|
Lee-Ødegård S, Olsen T, Norheim F, Drevon CA, Birkeland KI. Potential Mechanisms for How Long-Term Physical Activity May Reduce Insulin Resistance. Metabolites 2022; 12:metabo12030208. [PMID: 35323652 PMCID: PMC8950317 DOI: 10.3390/metabo12030208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Insulin became available for the treatment of patients with diabetes 100 years ago, and soon thereafter it became evident that the biological response to its actions differed markedly between individuals. This prompted extensive research into insulin action and resistance (IR), resulting in the universally agreed fact that IR is a core finding in patients with type 2 diabetes mellitus (T2DM). T2DM is the most prevalent form of diabetes, reaching epidemic proportions worldwide. Physical activity (PA) has the potential of improving IR and is, therefore, a cornerstone in the prevention and treatment of T2DM. Whereas most research has focused on the acute effects of PA, less is known about the effects of long-term PA on IR. Here, we describe a model of potential mechanisms behind reduced IR after long-term PA to guide further mechanistic investigations and to tailor PA interventions in the therapy of T2DM. The development of such interventions requires knowledge of normal glucose metabolism, and we briefly summarize an integrated physiological perspective on IR. We then describe the effects of long-term PA on signaling molecules involved in cellular responses to insulin, tissue-specific functions, and whole-body IR.
Collapse
Affiliation(s)
- Sindre Lee-Ødegård
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
| | - Christian Andre Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
- Vitas Ltd. Analytical Services, Oslo Science Park, 0349 Oslo, Norway
| | - Kåre Inge Birkeland
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
- Correspondence:
| |
Collapse
|
59
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 3170] [Impact Index Per Article: 1056.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
60
|
Gonzalez-Covarrubias V, Martínez-Martínez E, del Bosque-Plata L. The Potential of Metabolomics in Biomedical Applications. Metabolites 2022; 12:metabo12020194. [PMID: 35208267 PMCID: PMC8880031 DOI: 10.3390/metabo12020194] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 12/12/2022] Open
Abstract
The metabolome offers a dynamic, comprehensive, and precise picture of the phenotype. Current high-throughput technologies have allowed the discovery of relevant metabolites that characterize a wide variety of human phenotypes with respect to health, disease, drug monitoring, and even aging. Metabolomics, parallel to genomics, has led to the discovery of biomarkers and has aided in the understanding of a diversity of molecular mechanisms, highlighting its application in precision medicine. This review focuses on the metabolomics that can be applied to improve human health, as well as its trends and impacts in metabolic and neurodegenerative diseases, cancer, longevity, the exposome, liquid biopsy development, and pharmacometabolomics. The identification of distinct metabolomic profiles will help in the discovery and improvement of clinical strategies to treat human disease. In the years to come, metabolomics will become a tool routinely applied to diagnose and monitor health and disease, aging, or drug development. Biomedical applications of metabolomics can already be foreseen to monitor the progression of metabolic diseases, such as obesity and diabetes, using branched-chain amino acids, acylcarnitines, certain phospholipids, and genomics; these can assess disease severity and predict a potential treatment. Future endeavors should focus on determining the applicability and clinical utility of metabolomic-derived markers and their appropriate implementation in large-scale clinical settings.
Collapse
Affiliation(s)
| | - Eduardo Martínez-Martínez
- Laboratory of Cell Communication and Extracellular Vesicles, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Laura del Bosque-Plata
- Laboratory of Nutrigenetics and Nutrigenomics, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
- Correspondence: ; Tel.: +52-55-53-50-1974
| |
Collapse
|
61
|
Izundegui DG, Nayor M. Metabolomics of Type 1 and Type 2 Diabetes: Insights into Risk Prediction and Mechanisms. Curr Diab Rep 2022; 22:65-76. [PMID: 35113332 PMCID: PMC8934149 DOI: 10.1007/s11892-022-01449-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Metabolomics enables rapid interrogation of widespread metabolic processes making it well suited for studying diabetes. Here, we review the current status of metabolomic investigation in diabetes, highlighting its applications for improving risk prediction and mechanistic understanding. RECENT FINDINGS Findings of metabolite associations with type 2 diabetes risk have confirmed experimental observations (e.g., branched-chain amino acids) and also pinpointed novel pathways of diabetes risk (e.g., dimethylguanidino valeric acid). In type 1 diabetes, abnormal metabolite patterns are observed prior to the development of autoantibodies and hyperglycemia. Diabetes complications display specific metabolite signatures that are distinct from the metabolic derangements of diabetes and differ across vascular beds. Lastly, metabolites respond acutely to pharmacologic treatment, providing opportunities to understand inter-individual treatment responses. Metabolomic studies have elucidated biological mechanisms underlying diabetes development, complications, and therapeutic response. While not yet ready for clinical translation, metabolomics is a powerful and promising precision medicine tool.
Collapse
Affiliation(s)
| | - Matthew Nayor
- Sections of Cardiology and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, 72 E Concord Street, Suite L-516, Boston, MA, 02118, USA.
| |
Collapse
|
62
|
Huffman KM, Parker DC, Bhapkar M, Racette SB, Martin CK, Redman LM, Das SK, Connelly MA, Pieper CF, Orenduff M, Ross LM, Ramaker ME, Dorling JL, Rosen CJ, Shalaurova I, Otvos JD, Kraus VB, Kraus WE, CALERIE™ Investigators. Calorie restriction improves lipid-related emerging cardiometabolic risk factors in healthy adults without obesity: Distinct influences of BMI and sex from CALERIE™ a multicentre, phase 2, randomised controlled trial. EClinicalMedicine 2022; 43:101261. [PMID: 35028547 PMCID: PMC8741476 DOI: 10.1016/j.eclinm.2021.101261] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND For many cardiovascular risk factors there is no lower limit to which further reduction will result in decreased disease risk; this includes values within ranges considered normal for healthy adults. This seems to be true for new emerging metabolic risk factors identified by innovative technological advances. Further, there seems to be ever evolving evidence of differential responses to lifestyle interventions by sex and body compositions in the normal range. In this secondary analysis, we had the opportunity to test these principles for newly identified molecular biomarkers of cardiometabolic risk in a young (21-50 years), normal weight healthy population undergoing calorie restriction for two years. METHODS The Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy (CALERIE™) was a 24-month, multicenter, randomized controlled trial (May 2007-November 2012) in healthy, adults without obesity to evaluate the potential for calorie restriction (CR) to promote anti-aging adaptations, including those associated with disease risk. 218 participants (age 37.9 ± 7.2 years and body mass index (BMI) 25.1 ± 1.7 kg/m2, mean±SD) were randomized 2:1 to 24 months of CR (prescribed as 25% reduction from baseline calorie intake) versus ad libitum (AL). Fasting plasma from baseline, 12, and 24 months was used for assessments of lipoproteins, metabolites, and inflammatory markers using nuclear magnetic resonance spectroscopy. FINDINGS Averaging 11.9% CR, the CR group had reductions at 12 and 24 months in the cardiovascular disease risk markers, apolipoprotein B and GlycA, and risks for insulin resistance and type 2 diabetes-Lipoprotein Insulin Resistance Index and Diabetes Risk Index (all PCRvsAL ≤0.0009). Insulin resistance and diabetes risk improvements resulted from CR-induced alterations in lipoproteins, specifically reductions in triglyceride-rich lipoprotein particles and low-density lipoprotein particles, a shift to larger high-density lipoprotein particles (more effective cholesterol transporters), and reductions in branched chain amino acids (BCAAs) (all PCRvsAL ≤0.004). These CR responses were more pronounced in overweight than normal weight participants and greater in men than women. INTERPRETATION In normal to slightly overweight adults without overt risk factors or disease, 12 months of ∼12% CR improved newly identified risk markers for atherosclerotic cardiovascular disease, insulin resistance and type 2 diabetes. These markers suggest that CR improves risks by reducing inflammation and BCAAs and shifting lipoproteins from atherogenic to cholesterol transporting. Additionally, these improvements are greater for men and for those with greater BMIs indicating sex and BMI-influences merit attention in future investigations of lifestyle-mediated improvements in disease risk factors. FUNDING The CALERIE™ trial design and implementation were supported by a National Institutes of Health (NIH) U-grant provided to four institutions, the three intervention sites and a coordinating center (U01 AG022132, U01 AG020478, U01 AG020487 U01 AG020480). For this secondary analysis including sample acquisition and processing, data analysis and interpretation, additional funding was provided by the NIH to authors as follows: R01 AG054840 (MO, VBK); R33 AG070455 (KMH, DCP, MB, SBR, CKM, LMR, SKD, CFP, CJR, WEK); P30 DK072476 (CKM, LMR); and U54 GM104940 (CKM, LMR).
Collapse
Affiliation(s)
- Kim M. Huffman
- Divisions of Rheumatology and Immunology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
- Corresponding author.
| | - Daniel C. Parker
- Geriatrics, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Manjushri Bhapkar
- Duke Clinical Research Institute, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Susan B. Racette
- Program in Physical Therapy and Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | - Sai Krupa Das
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | | | - Carl F. Pieper
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Melissa Orenduff
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Leanna M. Ross
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Megan E. Ramaker
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - James L. Dorling
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Clifford J Rosen
- Maine Medical Center Research Institute 81 Research Drive Scarborough, Maine 04074 USA
| | - Irina Shalaurova
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, NC, USA
| | - James D. Otvos
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, NC, USA
| | - Virginia B. Kraus
- Divisions of Rheumatology and Immunology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - William E. Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
- Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
| | | |
Collapse
|
63
|
Xing WL, Liu HX, Niu Q, Wang YT, Zhu Y. Danhong injection improves elective percutaneous coronary intervention in ua patients with blood stasis syndrome revealed by perioperative metabolomics. WORLD JOURNAL OF TRADITIONAL CHINESE MEDICINE 2022. [DOI: 10.4103/wjtcm.wjtcm_63_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
64
|
Hu W, Yang P, Fu Z, Wang Y, Zhou Y, Ye Z, Gong Y, Huang A, Sun L, Zhao Y, Yang T, Li Z, Jiang XC, Yu W, Zhou H. High L-Valine Concentrations Associate with Increased Oxidative Stress and Newly-Diagnosed Type 2 Diabetes Mellitus: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2022; 15:499-509. [PMID: 35221701 PMCID: PMC8865866 DOI: 10.2147/dmso.s336736] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/14/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Branched-chain amino acids (BCAAs) are essential AAs which are widely used as antioxidants in patients with liver and kidney dysfunction. However, BCAAs are strongly correlated with insulin resistance (IR) and diabetes. This study aimed to evaluate the relationship among BCAAs, oxidative stress, and type 2 diabetes mellitus (T2DM) in a Chinese population. METHODS Anthropometric and biochemical examinations were performed in 816 individuals who participated in the Huai'an Diabetes Prevention Program. Serum BCAAs concentrations were measured by hydrophilic interaction chromatography-tandem mass spectrometric method. Oxidative stress was evaluated by malondialdehyde (MDA) as an index of lipid peroxidation and the superoxide dismutase (SOD) activity. RESULTS A total of 816 participants were divided into three groups: normal glucose metabolism (NGM), prediabetes, and newly-diagnosed diabetes mellitus (NDM). Subjects in NDM group show higher MDA and lower SOD levels than subjects in other groups. L-Val levels positively correlated with MDA levels and negatively with SOD in NDM groups. After adjusting for T2DM risk factors, high L-Val levels were significantly associated with higher BMI, WC, FPG, increased LnTG and decreased HDL-C. L-Val was also independently associated with NDM (OR 1.06, 95% CI 1.02-1.10; P = 0.005). Furthermore, the odds ratios for NDM among participants with high L-Val (≥35.25μg/mL) levels showed a 2.25-fold (95% CI 1.11-4.57; P = 0.024) increase compared to participants with low L-Val (<27.26 μg/mL) levels after adjusting for MDA and confounding factors. CONCLUSION High serum L-Val levels are independently associated with oxidative stress, thus promoting IR and NDM. Further study should be done to clarify the mechanism.
Collapse
Affiliation(s)
- Wen Hu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
- Department of Endocrinology and Metabolism, The Affiliated Huai’an Hospital of Xuzhou Medical College, Huai’an, Jiangsu, People’s Republic of China
| | - Panpan Yang
- Department of Respiratory Diseases, The Affiliated Huai’an Hospital of Xuzhou Medical College, Huai’an, Jiangsu, People’s Republic of China
| | - Zhenzhen Fu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Yongqing Wang
- Research Division of Clinical Pharmacology, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Ying Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Zhengqin Ye
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Yingyun Gong
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Aijie Huang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Luning Sun
- Research Division of Clinical Pharmacology, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Yang Zhao
- School of Public Health Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Zhong Li
- Key Laboratory of Rare Metabolic Diseases, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
- Jiangsu Province Key Laboratory of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Xian-Cheng Jiang
- Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Weinan Yu
- Department of Endocrinology and Metabolism, The Affiliated Huai’an Hospital of Xuzhou Medical College, Huai’an, Jiangsu, People’s Republic of China
- Weinan Yu, Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, 223001, People’s Republic of China, Email
| | - Hongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
- Key Laboratory of Rare Metabolic Diseases, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
- Correspondence: Hongwen Zhou, Department of endocrinology, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People’s Republic of China, Tel +862583718836-6893, Fax +862583781781, Email
| |
Collapse
|
65
|
Lanzon B, Martin-Taboada M, Castro-Alves V, Vila-Bedmar R, González de Pablos I, Duberg D, Gomez P, Rodriguez E, Orešič M, Hyötyläinen T, Morales E, Ruperez FJ, Medina-Gomez G. Lipidomic and Metabolomic Signature of Progression of Chronic Kidney Disease in Patients with Severe Obesity. Metabolites 2021; 11:836. [PMID: 34940593 PMCID: PMC8707539 DOI: 10.3390/metabo11120836] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Severe obesity is a major risk for chronic kidney disease (CKD). Early detection and careful monitoring of renal function are critical for the prevention of CKD during obesity, since biopsies are not performed in patients with CKD and diagnosis is dependent on the assessment of clinical parameters. To explore whether distinct lipid and metabolic signatures in obesity may signify early stages of pathogenesis toward CKD, liquid chromatography-mass spectrometry (LC-MS) and gas chromatography-high resolution accurate mass-mass spectrometry (GC-HRAM-MS) analyses were performed in the serum and the urine of severely obese patients with and without CKD. Moreover, the impact of bariatric surgery (BS) in lipid and metabolic signature was also studied, through LC-MS and GC-HRAM-MS analyses in the serum and urine of patients with severe obesity and CKD before and after undergoing BS. Regarding patients with severe obesity and CKD compared to severely obese patients without CKD, serum lipidome analysis revealed significant differences in lipid signature. Furthermore, serum metabolomics profile revealed significant changes in specific amino acids, with isoleucine and tyrosine, increased in CKD patients compared with patients without CKD. LC-MS and GC-HRAM-MS analysis in serum of patients with severe obesity and CKD after BS showed downregulation of levels of triglycerides (TGs) and diglycerides (DGs) as well as a decrease in branched-chain amino acid (BCAA), lysine, threonine, proline, and serine. In addition, BS removed most of the correlations in CKD patients against biochemical parameters related to kidney dysfunction. Concerning urine analysis, hippuric acid, valine and glutamine were significantly decreased in urine from CKD patients after surgery. Interestingly, bariatric surgery did not restore all the lipid species, some of them decreased, hence drawing attention to them as potential targets for early diagnosis or therapeutic intervention. Results obtained in this study would justify the use of comprehensive mass spectrometry-based lipidomics to measure other lipids aside from conventional lipid profiles and to validate possible early markers of risk of CKD in patients with severe obesity.
Collapse
Affiliation(s)
- Borja Lanzon
- LIPOBETA Group, Department Basic Sciences of Health, Faculty of Sciences of Health, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (B.L.); (M.M.-T.); (R.V.-B.)
| | - Marina Martin-Taboada
- LIPOBETA Group, Department Basic Sciences of Health, Faculty of Sciences of Health, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (B.L.); (M.M.-T.); (R.V.-B.)
| | - Victor Castro-Alves
- School of Science and Technology, Örebro University, 702 81 Örebro, Sweden; (V.C.-A.); (D.D.); (T.H.)
| | - Rocio Vila-Bedmar
- LIPOBETA Group, Department Basic Sciences of Health, Faculty of Sciences of Health, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (B.L.); (M.M.-T.); (R.V.-B.)
| | | | - Daniel Duberg
- School of Science and Technology, Örebro University, 702 81 Örebro, Sweden; (V.C.-A.); (D.D.); (T.H.)
| | - Pilar Gomez
- Department of Surgery, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (P.G.); (E.R.)
| | - Elias Rodriguez
- Department of Surgery, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; (P.G.); (E.R.)
| | - Matej Orešič
- School of Medical Sciences, Örebro University, 702 81 Örebro, Sweden;
- Turku Bioscience, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Tuulia Hyötyläinen
- School of Science and Technology, Örebro University, 702 81 Örebro, Sweden; (V.C.-A.); (D.D.); (T.H.)
| | - Enrique Morales
- Department of Nephrology, University Hospital 12 de Octubre, 28041 Madrid, Spain; (I.G.d.P.); (E.M.)
- Research Institute of University Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Medicine, Complutense University of Madrid, 28041 Madrid, Spain
| | - Francisco J. Ruperez
- Centro de Metabolómica y Bioanálisis (CEMBIO), Universidad San Pablo-CEU, CEU Universities, 28668 Boadilla del Monte, Spain;
| | - Gema Medina-Gomez
- LIPOBETA Group, Department Basic Sciences of Health, Faculty of Sciences of Health, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (B.L.); (M.M.-T.); (R.V.-B.)
- LAFEMEX Laboratory, Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
| |
Collapse
|
66
|
Williams MB, Palmer JW, Chehade SB, Hall AJ, Barry RJ, Powell ML, Harris ML, Sun LY, Watts SA. Effect of Long-Term Consumption of Poultry Egg Products on Growth, Body Composition, and Liver Gene Expression in Zebrafish, Danio rerio. Curr Dev Nutr 2021; 5:nzab134. [PMID: 34993387 PMCID: PMC8718328 DOI: 10.1093/cdn/nzab134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/07/2021] [Accepted: 10/29/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Poultry eggs are a low-cost, high-protein nutrient package that can be consumed as part of quality diets. However, consumption of poultry egg products is historically contentious, which highlights the importance of investigating impacts of long-term egg consumption on metabolic health. OBJECTIVE Our study utilized the zebrafish, Danio rerio, a newly defined model of human metabolic health, to understand the metabolic consequence of consuming egg products in lieu of other well-described protein sources. METHODS Reference diets were formulated to contain multisource protein with casein and fish protein hydrolysate (CON; control protein sources), the protein sources that have been historically utilized in numerous reference diets. These proteins were then partially replaced with either whole egg (WE; protein and lipid source), egg white (EW; protein source), wheat gluten (WG; cereal protein source), or a high-lipid-content diet containing a multisource protein with casein and fish protein hydrolysate (HFCON; isonitrogenous and isolipidic with the WE diet) in a 34-wk trial (n = 8 tanks, 10 fish per tank). Daily feeding was initiated at the early juvenile life stage and terminated at the late reproductive adult stage. RESULTS The amino acid composition of control versus egg product diets did not vary substantially, although methionine and lysine were apparently limiting in fish fed WG. At termination, fish fed EW as the protein source had weight gain and body composition similar to those fed the CON diet. Fasting and postprandial blood glucose did not differ between any dietary treatment. Assessment of the liver transcriptome using RNAseq revealed no differential gene expression between zebrafish fed CON or WE diets. Zebrafish fed WG had lower weight gain in males. CONCLUSIONS Long-term consumption of egg products promoted metabolic health equal to that of historically relevant proteins. These data support the value of egg products for maintaining long-term metabolic health in animal diets.
Collapse
Affiliation(s)
- Michael B Williams
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joseph W Palmer
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sophie B Chehade
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alex J Hall
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert J Barry
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mickie L Powell
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Melissa L Harris
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Liou Y Sun
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephen A Watts
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
67
|
Flores-Guerrero JL, Connelly MA, Shalaurova I, Garcia E, Bakker SJL, Dullaart RPF. A metabolomic index based on lipoprotein subfractions and branched chain amino acids is associated with incident hypertension. Eur J Intern Med 2021; 94:56-63. [PMID: 34321184 DOI: 10.1016/j.ejim.2021.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/25/2021] [Accepted: 07/10/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE The present study aims to evaluate the performance of the Diabetes Risk Index (DRI), a metabolomic index based on lipoprotein particles and branched chain amino acids, on the incidence of newly developed hypertension in a large community dwelling cohort. METHODS The DRI was calculated by combining 6 lipoprotein parameters [sizes of very-low-density lipoprotein (VLDL), low-density lipoprotein (LDL) and high-density lipoprotein (HDL), concentrations of large VLDL, small LDL, and large HDL particles], and the concentrations of valine and leucine. DRI scores were estimated in 4169 participants from the PREVEND prospective cohort. Cox proportional hazards regression was used to evaluate the association of DRI scores with incident hypertension. RESULTS During a median follow-up of 8.6 years, 924 new hypertension cases were ascertained. In analyses adjusted for age and sex, there was a significant association between DRI and incident hypertension with a hazard ratio (HR) per 1 SD increase of 1.45 (95% CI 1.36,1.54; p < 0.001). After additional adjustment for traditional risk factors, the HR remained significant (HRadj 1.21, 95% CI 1.10, 1.33, p <0.001). Likewise, subjects in the top quartile of DRI presented with a higher risk of hypertension (HRadj 1.64, 95% CI 1.28, 2.10, p <0.001). Furthermore, the net reclassification improvement assessment improved after the addition of DRI to a traditional risk model (p <0.001), allowing proper reclassification of 34% of the participants. CONCLUSION Higher DRI scores were associated with an increased risk of incident hypertension. Such association was independent of traditional clinical risk factors for hypertension.
Collapse
Affiliation(s)
- Jose L Flores-Guerrero
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands..
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, North Carolina, USA
| | - Irina Shalaurova
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, North Carolina, USA
| | - Erwin Garcia
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, North Carolina, USA
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Robin P F Dullaart
- Department of Internal Medicine, Division of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
68
|
Mahbub MH, Yamaguchi N, Nakagami Y, Hase R, Takahashi H, Ishimaru Y, Watanabe R, Saito H, Shimokawa J, Yamamoto H, Kikuchi S, Tanabe T. Association of Plasma Branched-Chain and Aromatic Amino Acids with Reduction in Kidney Function Evaluated in Apparently Healthy Adults. J Clin Med 2021; 10:jcm10225234. [PMID: 34830517 PMCID: PMC8625145 DOI: 10.3390/jcm10225234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 01/09/2023] Open
Abstract
The published literature on the association of circulatory branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs) with reduced kidney function is inconsistent or conflicting. Clarification of it might help to better understand the underlying pathophysiology and to determine potential biomarkers for early detection and evaluation of kidney function decline. Our main purpose was to explore and clarify the potential relationships of individual BCAAs and AAAs with estimated glomerular filtration rate (eGFR) decline. We included the data from 2804 healthy subjects and categorized them into three groups based on eGFR tertiles. The associations between individual amino acids and eGFR were explored by covariate-adjusted logistic regression models. There was a progressive increase in the concentrations of BCAAs and AAAs from the upper to the lower tertiles. We revealed significant positive associations of isoleucine, leucine, and phenylalanine with lower tertiles of eGFR in the adjusted models (p < 0.01-0.001). The findings hold a promising potential of using plasma isoleucine, leucine, and phenylalanine levels for evaluation of kidney function decline. Future longitudinal studies should investigate the causal association between altered levels of these amino acids and impaired kidney function and also the utility of the former as potential biomarkers for evaluating the risk and early detection of the latter.
Collapse
Affiliation(s)
- MH Mahbub
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
- Correspondence: ; Tel.: +81-836-22-2231
| | - Natsu Yamaguchi
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Yuki Nakagami
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Ryosuke Hase
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hidekazu Takahashi
- Department of Public Health, Faculty of Veterinary Medicine, Okayama University of Science, Imabari 794-8555, Japan;
| | - Yasutaka Ishimaru
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Rie Watanabe
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hiroyuki Saito
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Junki Shimokawa
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hiroshi Yamamoto
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki 210-8681, Japan; (H.Y.); (S.K.)
| | - Shinya Kikuchi
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki 210-8681, Japan; (H.Y.); (S.K.)
| | - Tsuyoshi Tanabe
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (N.Y.); (Y.N.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| |
Collapse
|
69
|
The Association between Branched-Chain Amino Acids (BCAAs) and Cardiometabolic Risk Factors in Middle-Aged Caucasian Women Stratified According to Glycemic Status. Nutrients 2021; 13:nu13103307. [PMID: 34684308 PMCID: PMC8538048 DOI: 10.3390/nu13103307] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 02/07/2023] Open
Abstract
We examined the glycemic status-stratified relationships between total serum branched-chain amino acid (BCAA) concentrations and cardiometabolic risk factors in middle-aged Caucasian women. The study included 349 women divided into 2 subgroups: a normoglycemic group (NG, n = 184) and a dysglycemic group (DG, n = 165). Blood samples, anthropometric parameters, and blood pressure were measured. HOMA-IR, albumin-corrected calcium (CCa), and fatty liver index (FLI) were calculated. BCAA concentrations were higher in the women with dysglycemia. BCAAs moderately correlated with BMI and FLI in the NG group and with BMI, FLI, total calcium (TCa), CCa, HbA1c, TG/HDL-C, and HDL-C in the DG group. After adjusting for age and BMI, correlations for TCa, CCa, HbA1c, HDL-C, and TG/HDL-C remained significant. The coexistence of increased BCAAs with dysglycemic status was associated with markedly higher concentrations of TCa, CCa, HbA1c, and TG, which were not observed in the DG women with low level of BCAAs. Multiple regression showed that TCa or CCa, age and BCAAs were significantly associated with HbA1c independently of BMI only in the DG group. We conclude that dysglycemia in particular predisposes women to a significant relationship between total BCAAs and circulating calcium and HbA1c, and that these relationships are independent of BMI and may reflect the pathophysiological calcium-dependent mechanisms connecting BCAAs with metabolic disturbances.
Collapse
|
70
|
Bozadjieva Kramer N, Evers SS, Shin JH, Silverwood S, Wang Y, Burant CF, Sandoval DA, Seeley RJ. The Role of Elevated Branched-Chain Amino Acids in the Effects of Vertical Sleeve Gastrectomy to Reduce Weight and Improve Glucose Regulation. Cell Rep 2021; 33:108239. [PMID: 33053352 PMCID: PMC7605275 DOI: 10.1016/j.celrep.2020.108239] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/24/2020] [Accepted: 09/15/2020] [Indexed: 12/16/2022] Open
Abstract
Elevated levels of branched-chain amino acids (BCAAs) and their metabolites are strongly positively associated with obesity, insulin resistance, and type 2 diabetes. Bariatric surgery is among the best treatments for weight loss and associated morbidities. Clinical studies have reported that bariatric surgery decreases the circulating levels of BCAAs. The objective of this study was to test the hypothesis that reduced BCAA levels contribute to the metabolic improvements of sustained weight loss and improved glucose tolerance after vertical sleeve gastrectomy (VSG). We find that, as in humans, circulating BCAAs are significantly lower in VSG rats and mice. To increase circulating BCAAs, we tested mice with either increased dietary intake of BCAAs or impaired BCAA catabolism by total body deletion of mitochondrial phosphatase 2C (Pp2cm). Our results show that a decrease in circulating BCAAs is not necessary for sustained body weight loss and improved glucose tolerance after VSG. Increased branched-chain amino acid (BCAA) levels are biomarkers of metabolic disease, and bariatric surgeries reduce BCAA levels. Bozadjieva Kramer et al. show that both dietary and genetic manipulations can block the surgical effect on BCAAs but do not alter potent, beneficial effects on weight loss and glucose tolerance.
Collapse
Affiliation(s)
| | - Simon S Evers
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jae Hoon Shin
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sierra Silverwood
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yibin Wang
- Departments of Anesthesiology, Medicine, and Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
71
|
Sjögren RJO, Rizo-Roca D, Chibalin AV, Chorell E, Furrer R, Katayama S, Harada J, Karlsson HKR, Handschin C, Moritz T, Krook A, Näslund E, Zierath JR. Branched-chain amino acid metabolism is regulated by ERRα in primary human myotubes and is further impaired by glucose loading in type 2 diabetes. Diabetologia 2021; 64:2077-2091. [PMID: 34131782 PMCID: PMC8382616 DOI: 10.1007/s00125-021-05481-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/12/2021] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Increased levels of branched-chain amino acids (BCAAs) are associated with type 2 diabetes pathogenesis. However, most metabolomic studies are limited to an analysis of plasma metabolites under fasting conditions, rather than the dynamic shift in response to a metabolic challenge. Moreover, metabolomic profiles of peripheral tissues involved in glucose homeostasis are scarce and the transcriptomic regulation of genes involved in BCAA catabolism is partially unknown. This study aimed to identify differences in circulating and skeletal muscle BCAA levels in response to an OGTT in individuals with normal glucose tolerance (NGT) or type 2 diabetes. Additionally, transcription factors involved in the regulation of the BCAA gene set were identified. METHODS Plasma and vastus lateralis muscle biopsies were obtained from individuals with NGT or type 2 diabetes before and after an OGTT. Plasma and quadriceps muscles were harvested from skeletal muscle-specific Ppargc1a knockout and transgenic mice. BCAA-related metabolites and genes were assessed by LC-MS/MS and quantitative RT-PCR, respectively. Small interfering RNA and adenovirus-mediated overexpression techniques were used in primary human skeletal muscle cells to study the role of PPARGC1A and ESRRA in the expression of the BCAA gene set. Radiolabelled leucine was used to analyse the impact of oestrogen-related receptor α (ERRα) knockdown on leucine oxidation. RESULTS Impairments in BCAA catabolism in people with type 2 diabetes under fasting conditions were exacerbated after a glucose load. Branched-chain keto acids were reduced 37-56% after an OGTT in the NGT group, whereas no changes were detected in individuals with type 2 diabetes. These changes were concomitant with a stronger correlation with glucose homeostasis biomarkers and downregulated expression of branched-chain amino acid transaminase 2, branched-chain keto acid dehydrogenase complex subunits and 69% of downstream BCAA-related genes in skeletal muscle. In primary human myotubes overexpressing peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α, encoded by PPARGC1A), 61% of the analysed BCAA genes were upregulated, while 67% were downregulated in the quadriceps of skeletal muscle-specific Ppargc1a knockout mice. ESRRA (encoding ERRα) silencing completely abrogated the PGC-1α-induced upregulation of BCAA-related genes in primary human myotubes. CONCLUSIONS/INTERPRETATION Metabolic inflexibility in type 2 diabetes impacts BCAA homeostasis and attenuates the decrease in circulating and skeletal muscle BCAA-related metabolites after a glucose challenge. Transcriptional regulation of BCAA genes in primary human myotubes via PGC-1α is ERRα-dependent.
Collapse
Affiliation(s)
- Rasmus J O Sjögren
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - David Rizo-Roca
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | - Shintaro Katayama
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Jun Harada
- Cardiovascular-Metabolics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Håkan K R Karlsson
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | | | - Thomas Moritz
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Erik Näslund
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
72
|
Wang QY, You LH, Xiang LL, Zhu YT, Zeng Y. Current progress in metabolomics of gestational diabetes mellitus. World J Diabetes 2021; 12:1164-1186. [PMID: 34512885 PMCID: PMC8394228 DOI: 10.4239/wjd.v12.i8.1164] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/20/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common metabolic disorders of pregnancy and can cause short- and long-term adverse effects in both pregnant women and their offspring. However, the etiology and pathogenesis of GDM are still unclear. As a metabolic disease, GDM is well suited to metabolomics study, which can monitor the changes in small molecular metabolites induced by maternal stimuli or perturbations in real time. The application of metabolomics in GDM can be used to discover diagnostic biomarkers, evaluate the prognosis of the disease, guide the application of diet or drugs, evaluate the curative effect, and explore the mechanism. This review provides comprehensive documentation of metabolomics research methods and techniques as well as the current progress in GDM research. We anticipate that the review will contribute to identifying gaps in the current knowledge or metabolomics technology, provide evidence-based information, and inform future research directions in GDM.
Collapse
Affiliation(s)
- Qian-Yi Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 21000, Jiangsu Province, China
| | - Liang-Hui You
- Nanjing Maternity and Child Health Care Institute, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Lan-Lan Xiang
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Yi-Tian Zhu
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Yu Zeng
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| |
Collapse
|
73
|
Nayor M, Shah SH, Murthy V, Shah RV. Molecular Aspects of Lifestyle and Environmental Effects in Patients With Diabetes: JACC Focus Seminar. J Am Coll Cardiol 2021; 78:481-495. [PMID: 34325838 DOI: 10.1016/j.jacc.2021.02.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/07/2021] [Accepted: 02/01/2021] [Indexed: 01/04/2023]
Abstract
Diabetes is characterized as an integrated condition of dysregulated metabolism across multiple tissues, with well-established consequences on the cardiovascular system. Recent advances in precision phenotyping in biofluids and tissues in large human observational and interventional studies have afforded a unique opportunity to translate seminal findings in models and cellular systems to patients at risk for diabetes and its complications. Specifically, techniques to assay metabolites, proteins, and transcripts, alongside more recent assessment of the gut microbiome, underscore the complexity of diabetes in patients, suggesting avenues for precision phenotyping of risk, response to intervention, and potentially novel therapies. In addition, the influence of external factors and inputs (eg, activity, diet, medical therapies) on each domain of molecular characterization has gained prominence toward better understanding their role in prevention. Here, the authors provide a broad overview of the role of several of these molecular domains in human translational investigation in diabetes.
Collapse
Affiliation(s)
- Matthew Nayor
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/MattNayor
| | - Svati H Shah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA. https://twitter.com/SvatiShah
| | - Venkatesh Murthy
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA. https://twitter.com/venkmurthy
| | - Ravi V Shah
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
74
|
Kelsey M, Pagidipati N. Towards a personalised approach for obesity treatment: one size does not fit all. Heart 2021; 107:1526-1527. [PMID: 34326134 DOI: 10.1136/heartjnl-2021-319726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Michelle Kelsey
- Duke Clinical Research Institute, Durham, North Carolina, USA .,Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
| | - Neha Pagidipati
- Duke Clinical Research Institute, Durham, North Carolina, USA.,Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
75
|
Metabolomics in Bariatric Surgery: Towards Identification of Mechanisms and Biomarkers of Metabolic Outcomes. Obes Surg 2021; 31:4564-4574. [PMID: 34318371 DOI: 10.1007/s11695-021-05566-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022]
Abstract
Bariatric surgery has been widely performed for the treatment of obesity and type 2 diabetes. Efforts have been made to investigate the mechanisms underlying the metabolic effects achieved by bariatric surgery and to identify candidates who will benefit from this surgery. Metabolomics, which includes comprehensive profiling of metabolites in biological samples, has been utilized for various disease entities to discover pathophysiological metabolic pathways and biomarkers predicting disease progression or prognosis. Over the last decade, metabolomic studies on patients undergoing bariatric surgery have identified significant biomarkers related to metabolic effects. This review describes the significance, progress, and challenges for the future of metabolomics in the area of bariatric surgery.
Collapse
|
76
|
Integrated metabolomics analysis of the effect of PPARδ agonist GW501516 on catabolism of BCAAs and carboxylic acids in diabetic mice. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2020.12.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
77
|
Lee J, Vijayakumar A, White PJ, Xu Y, Ilkayeva O, Lynch CJ, Newgard CB, Kahn BB. BCAA Supplementation in Mice with Diet-induced Obesity Alters the Metabolome Without Impairing Glucose Homeostasis. Endocrinology 2021; 162:6188397. [PMID: 33765118 PMCID: PMC8183497 DOI: 10.1210/endocr/bqab062] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Indexed: 12/26/2022]
Abstract
Circulating branched chain amino acid (BCAA) levels are elevated in obese humans and genetically obese rodents. However, the relationship of BCAAs to insulin resistance in diet-induced obese mice, a commonly used model to study glucose homeostasis, is still ill-defined. Here we examined how high-fat high-sucrose (HFHS) or high-fat diet (HFD) feeding, with or without BCAA supplementation in water, alters the metabolome in serum/plasma and tissues in mice and whether raising circulating BCAA levels worsens insulin resistance and glucose intolerance. Neither HFHS nor HFD feeding raised circulating BCAA levels in insulin-resistant diet-induced obese mice. BCAA supplementation raised circulating BCAA and branched-chain α-keto acid levels and C5-OH/C3-DC acylcarnitines (AC) in muscle from mice fed an HFHS diet or HFD, but did not worsen insulin resistance. A set of short- and long-chain acyl CoAs were elevated by diet alone in muscle, liver, and white adipose tissue (WAT), but not increased further by BCAA supplementation. HFD feeding reduced valine and leucine oxidation in WAT but not in muscle. BCAA supplementation markedly increased valine oxidation in muscle from HFD-fed mice, while leucine oxidation was unaffected by diet or BCAA treatment. Here we establish an extensive metabolome database showing tissue-specific changes in mice on 2 different HFDs, with or without BCAA supplementation. We conclude that mildly elevating circulating BCAAs and a subset of ACs by BCAA supplementation does not worsen insulin resistance or glucose tolerance in mice. This work highlights major differences in the effects of BCAAs on glucose homeostasis in diet-induced obese mice versus data reported in obese rats and in humans.
Collapse
Affiliation(s)
- Jennifer Lee
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Archana Vijayakumar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
- Current Affiliation: Archana Vijayakumar, Gilead Sciences, Inc, Foster City, CA 94404, USA; Christopher J. Lynch, Office of Nutrition Research, National Institutes of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Phillip J White
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA
- 3Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrine Division, Duke University Medical Center, Durham, NC 27701, USA
| | - Yuping Xu
- Cellular and Molecular Physiology Department, Pennsylvania State University, Hershey, PA 17033, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA
- 3Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrine Division, Duke University Medical Center, Durham, NC 27701, USA
| | - Christopher J Lynch
- Cellular and Molecular Physiology Department, Pennsylvania State University, Hershey, PA 17033, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA
- 3Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrine Division, Duke University Medical Center, Durham, NC 27701, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
- Correspondence: Barbara B Kahn, MD, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA. E-mail:
| |
Collapse
|
78
|
Bihlmeyer NA, Kwee LC, Clish CB, Deik AA, Gerszten RE, Pagidipati NJ, Laferrère B, Svetkey LP, Newgard CB, Kraus WE, Shah SH. Metabolomic profiling identifies complex lipid species and amino acid analogues associated with response to weight loss interventions. PLoS One 2021; 16:e0240764. [PMID: 34043632 PMCID: PMC8158886 DOI: 10.1371/journal.pone.0240764] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is an epidemic internationally. While weight loss interventions are efficacious, they are compounded by heterogeneity with regards to clinically relevant metabolic responses. Thus, we sought to identify metabolic biomarkers that are associated with beneficial metabolic changes to weight loss and which distinguish individuals with obesity who would most benefit from a given type of intervention. Liquid chromatography mass spectrometry-based profiling was used to measure 765 metabolites in baseline plasma from three different weight loss studies: WLM (behavioral intervention, N = 443), STRRIDE-PD (exercise intervention, N = 163), and CBD (surgical cohort, N = 125). The primary outcome was percent change in insulin resistance (as measured by the Homeostatic Model Assessment of Insulin Resistance [%ΔHOMA-IR]) over the intervention. Overall, 92 individual metabolites were associated with %ΔHOMA-IR after adjustment for multiple comparisons. Concordantly, the most significant metabolites were triacylglycerols (TAGs; p = 2.3e-5) and diacylglycerols (DAGs; p = 1.6e-4), with higher baseline TAG and DAG levels associated with a greater improvement in insulin resistance with weight loss. In tests of heterogeneity, 50 metabolites changed differently between weight loss interventions; we found amino acids, peptides, and their analogues to be most significant (4.7e-3) in this category. Our results highlight novel metabolic pathways associated with heterogeneity in response to weight loss interventions, and related biomarkers which could be used in future studies of personalized approaches to weight loss interventions.
Collapse
Affiliation(s)
- Nathan A. Bihlmeyer
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Lydia Coulter Kwee
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Clary B. Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Amy Anderson Deik
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Neha J. Pagidipati
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Blandine Laferrère
- Columbia University Irving Medical Center, New York, New York, United States of America
| | - Laura P. Svetkey
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Christopher B. Newgard
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - William E. Kraus
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
| | - Svati H. Shah
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
79
|
White PJ, McGarrah RW, Herman MA, Bain JR, Shah SH, Newgard CB. Insulin action, type 2 diabetes, and branched-chain amino acids: A two-way street. Mol Metab 2021; 52:101261. [PMID: 34044180 PMCID: PMC8513145 DOI: 10.1016/j.molmet.2021.101261] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A strong association of obesity and insulin resistance with increased circulating levels of branched-chain and aromatic amino acids and decreased glycine levels has been recognized in human subjects for decades. SCOPE OF REVIEW More recently, human metabolomics and genetic studies have confirmed and expanded upon these observations, accompanied by a surge in preclinical studies that have identified mechanisms involved in the perturbation of amino acid homeostasis- how these events are connected to dysregulated glucose and lipid metabolism, and how elevations in branched-chain amino acids (BCAA) may participate in the development of insulin resistance, type 2 diabetes (T2D), and other cardiometabolic diseases and conditions. MAJOR CONCLUSIONS In human cohorts, BCAA and related metabolites are now well established as among the strongest biomarkers of obesity, insulin resistance, T2D, and cardiovascular diseases. Lowering of BCAA and branched-chain ketoacid (BCKA) levels by feeding BCAA-restricted diet or by the activation of the rate-limiting enzyme in BCAA catabolism, branched-chain ketoacid dehydrogenase (BCKDH), in rodent models of obesity have clear salutary effects on glucose and lipid homeostasis, but BCAA restriction has more modest effects in short-term studies in human T2D subjects. Feeding of rats with diets enriched in sucrose or fructose result in the induction of the ChREBP transcription factor in the liver to increase expression of the BCKDH kinase (BDK) and suppress the expression of its phosphatase (PPM1K) resulting in the inactivation of BCKDH and activation of the key lipogenic enzyme ATP-citrate lyase (ACLY). These and other emergent links between BCAA, glucose, and lipid metabolism motivate ongoing studies of possible causal actions of BCAA and related metabolites in the development of cardiometabolic diseases.
Collapse
Affiliation(s)
- Phillip J White
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, 27701, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA; Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, 27701, USA; Department of Medicine, Divisions of Endocrinology & Metabolism and Cardiology, Duke University Medical Center, Durham, NC, 27701, USA
| | - Robert W McGarrah
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, 27701, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA; Department of Medicine, Divisions of Endocrinology & Metabolism and Cardiology, Duke University Medical Center, Durham, NC, 27701, USA
| | - Mark A Herman
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, 27701, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA; Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, 27701, USA; Department of Medicine, Divisions of Endocrinology & Metabolism and Cardiology, Duke University Medical Center, Durham, NC, 27701, USA
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, 27701, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA; Department of Medicine, Divisions of Endocrinology & Metabolism and Cardiology, Duke University Medical Center, Durham, NC, 27701, USA
| | - Svati H Shah
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, 27701, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA; Department of Medicine, Divisions of Endocrinology & Metabolism and Cardiology, Duke University Medical Center, Durham, NC, 27701, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, 27701, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA; Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, 27701, USA; Department of Medicine, Divisions of Endocrinology & Metabolism and Cardiology, Duke University Medical Center, Durham, NC, 27701, USA.
| |
Collapse
|
80
|
Holeček M. The role of skeletal muscle in the pathogenesis of altered concentrations of branched-chain amino acids (valine, leucine, and isoleucine) in liver cirrhosis, diabetes, and other diseases. Physiol Res 2021; 70:293-305. [PMID: 33982576 DOI: 10.33549/physiolres.934648] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The article shows that skeletal muscle plays a dominant role in the catabolism of branched-chain amino acids (BCAAs; valine, leucine, and isoleucine) and the pathogenesis of their decreased concentrations in liver cirrhosis, increased concentrations in diabetes, and nonspecific alterations in disorders with signs of systemic inflammatory response syndrome (SIRS), such as burn injury and sepsis. The main role of skeletal muscle in BCAA catabolism is due to its mass and high activity of BCAA aminotransferase, which is absent in the liver. Decreased BCAA levels in liver cirrhosis are due to increased use of the BCAA as a donor of amino group to alpha-ketoglutarate for synthesis of glutamate, which in muscles acts as a substrate for ammonia detoxification to glutamine. Increased BCAA levels in diabetes are due to alterations in glycolysis, citric acid cycle, and fatty acid oxidation. Decreased glycolysis and citric cycle activity impair BCAA transamination to branched-chain keto acids (BCKAs) due to decreased supply of amino group acceptors (alpha-ketoglutarate, pyruvate, and oxaloacetate); increased fatty acid oxidation inhibits flux of BCKA through BCKA dehydrogenase due to increased supply of NADH and acyl-CoAs. Alterations in BCAA levels in disorders with SIRS are inconsistent due to contradictory effects of SIRS on muscles. Specifically, increased proteolysis and insulin resistance tend to increase BCAA levels, whereas activation of BCKA dehydrogenase and glutamine synthesis tend to decrease BCAA levels. The studies are needed to elucidate the role of alterations in BCAA metabolism and the effects of BCAA supplementation on the outcomes of specific diseases.
Collapse
Affiliation(s)
- M Holeček
- Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic.
| |
Collapse
|
81
|
The effect of physical activity level and exercise training on the association between plasma branched-chain amino acids and intrahepatic lipid content in participants with obesity. Int J Obes (Lond) 2021; 45:1510-1520. [PMID: 33935282 PMCID: PMC8236400 DOI: 10.1038/s41366-021-00815-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/01/2021] [Accepted: 04/09/2021] [Indexed: 01/05/2023]
Abstract
Aims To evaluate whether the association between plasma branched-chain amino acids (BCAA) and intrahepatic lipid (IHL) was affected by physical activity level. Furthermore, to investigate if a conventional exercise training program, a subcategory of physical activity, could lower plasma BCAA along with alterations in IHL content in patients with type 2 diabetes (T2DM) and people with nonalcoholic fatty liver (NAFL). Methods To investigate the effect of physical activity on the association between plasma BCAA and IHL content, linear regression analyses were performed in 1983 individuals from the Netherlands Epidemiology of Obesity (NEO) stratified by physical activity frequency. Furthermore, the effect of a 12-week supervised combined aerobic resistance-exercise program on plasma BCAA, insulin sensitivity (hyperinsulinemic–euglycemic clamp), and IHL (proton-magnetic resonance spectroscopy (1H-MRS)) was investigated in seven patients with T2DM, seven individuals with NAFL and seven BMI-matched control participants (CON). Results We observed positive associations between plasma valine, isoleucine and leucine level, and IHL content (1.29 (95% CI: 1.21, 1.38), 1.52 (95% CI: 1.43, 1.61), and 1.54 (95% CI: 1.44, 1.64) times IHL, respectively, per standard deviation of plasma amino acid level). Similar associations were observed in less active versus more active individuals. Exercise training did not change plasma BCAA levels among groups, but reduced IHL content in NAFL (from 11.6 ± 3.0% pre-exercise to 8.1 ± 2.0% post exercise, p < 0.05) and CON (from 2.4 ± 0.6% pre-exercise to 1.6 ± 1.4% post exercise, p < 0.05), and improved peripheral insulin sensitivity in NAFL as well by ~23% (p < 0.05). Conclusions The association between plasma BCAA levels and IHL is not affected by physical activity level. Exercise training reduced IHL without affecting plasma BCAA levels in individuals with NAFL and CON. We conclude that exercise training-induced reduction in IHL content is not related to changes in plasma BCAA levels. Trial registration Trial registry number: NCT01317576.
Collapse
|
82
|
Kliemann N, Viallon V, Murphy N, Beeken RJ, Rothwell JA, Rinaldi S, Assi N, van Roekel EH, Schmidt JA, Borch KB, Agnoli C, Rosendahl AH, Sartor H, Huerta JM, Tjønneland A, Halkjær J, Bueno-de-Mesquita B, Gicquiau A, Achaintre D, Aleksandrova K, Schulze MB, Heath AK, Tsilidis KK, Masala G, Panico S, Kaaks R, Fortner RT, Van Guelpen B, Dossus L, Scalbert A, Keun HC, Travis RC, Jenab M, Johansson M, Ferrari P, Gunter MJ. Metabolic signatures of greater body size and their associations with risk of colorectal and endometrial cancers in the European Prospective Investigation into Cancer and Nutrition. BMC Med 2021; 19:101. [PMID: 33926456 PMCID: PMC8086283 DOI: 10.1186/s12916-021-01970-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/22/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The mechanisms underlying the obesity-cancer relationship are incompletely understood. This study aimed to characterise metabolic signatures of greater body size and to investigate their association with two obesity-related malignancies, endometrial and colorectal cancers, and with weight loss within the context of an intervention study. METHODS Targeted mass spectrometry metabolomics data from 4326 participants enrolled in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort and 17 individuals from a single-arm pilot weight loss intervention (Intercept) were used in this analysis. Metabolic signatures of body size were first determined in discovery (N = 3029) and replication (N = 1297) sets among EPIC participants by testing the associations between 129 metabolites and body mass index (BMI), waist circumference (WC), and waist-to-hip ratio (WHR) using linear regression models followed by partial least squares analyses. Conditional logistic regression models assessed the associations between the metabolic signatures with endometrial (N = 635 cases and 648 controls) and colorectal (N = 423 cases and 423 controls) cancer risk using nested case-control studies in EPIC. Pearson correlation between changes in the metabolic signatures and weight loss was tested among Intercept participants. RESULTS After adjustment for multiple comparisons, greater BMI, WC, and WHR were associated with higher levels of valine, isoleucine, glutamate, PC aa C38:3, and PC aa C38:4 and with lower levels of asparagine, glutamine, glycine, serine, lysoPC C17:0, lysoPC C18:1, lysoPC C18:2, PC aa C42:0, PC ae C34:3, PC ae C40:5, and PC ae C42:5. The metabolic signature of BMI (OR1-sd 1.50, 95% CI 1.30-1.74), WC (OR1-sd 1.46, 95% CI 1.27-1.69), and WHR (OR1-sd 1.54, 95% CI 1.33-1.79) were each associated with endometrial cancer risk. Risk of colorectal cancer was positively associated with the metabolic signature of WHR (OR1-sd: 1.26, 95% CI 1.07-1.49). In the Intercept study, a positive correlation was observed between weight loss and changes in the metabolic signatures of BMI (r = 0.5, 95% CI 0.06-0.94, p = 0.03), WC (r = 0.5, 95% CI 0.05-0.94, p = 0.03), and WHR (r = 0.6, 95% CI 0.32-0.87, p = 0.01). CONCLUSIONS Obesity is associated with a distinct metabolic signature comprising changes in levels of specific amino acids and lipids which is positively associated with both colorectal and endometrial cancer and is potentially reversible following weight loss.
Collapse
Affiliation(s)
- Nathalie Kliemann
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Vivian Viallon
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Neil Murphy
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Rebecca J Beeken
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
- Department of Behavioural Science and Health, University College London, London, UK
| | - Joseph A Rothwell
- Health Across Generations team, Centre for Research in Epidemiology and Population Health (CESP), INSERM U1018, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Sabina Rinaldi
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Nada Assi
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Eline H van Roekel
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Julie A Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Kristin Benjaminsen Borch
- Department of Community Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Claudia Agnoli
- Epidemiology and Prevention Unit. Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ann H Rosendahl
- Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Hanna Sartor
- Diagnostic Radiology, Lund University, Lund, Sweden
| | - José María Huerta
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | | | - Jytte Halkjær
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Audrey Gicquiau
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - David Achaintre
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Krasimira Aleksandrova
- Nutrition, Immunity and Metabolism Senior Scientist Group, Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Matthias B Schulze
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Giovanna Masala
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network - ISPRO, Florence, Italy
| | - Salvatore Panico
- Dipartimento di Medicin Clinica e Chirurgia, Frederico II Univeristy, Naples, Italy
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Renée T Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology, Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Laure Dossus
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Augustin Scalbert
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Hector C Keun
- Cancer Metabolism and Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College, London, UK
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Mazda Jenab
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Mattias Johansson
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Pietro Ferrari
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Marc J Gunter
- International Agency for Research on Cancer, World Health Organization, Lyon, France.
| |
Collapse
|
83
|
Ooi DSQ, Ling JQR, Sadananthan SA, Velan SS, Ong FY, Khoo CM, Tai ES, Henry CJ, Leow MKS, Khoo EYH, Tan CS, Lee YS, Chong MFF. Branched-Chain Amino Acid Supplementation Does Not Preserve Lean Mass or Affect Metabolic Profile in Adults with Overweight or Obesity in a Randomized Controlled Weight Loss Intervention. J Nutr 2021; 151:911-920. [PMID: 33537760 DOI: 10.1093/jn/nxaa414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/18/2020] [Accepted: 12/01/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Branched-chain amino acid (BCAA) supplementation has been shown to increase muscle mass or prevent muscle loss during weight loss. OBJECTIVE We aimed to investigate the effects of a BCAA-supplemented hypocaloric diet on lean mass preservation and insulin sensitivity. METHODS A total of 132 Chinese adults (63 men and 69 women aged 21-45 y, BMI 25-36 kg/m2) were block randomly assigned by gender and BMI into 3 hypocaloric diet (deficit of 500 kcal/d) groups: standard-protein (14%) with placebo (control, CT) or BCAA supplements at 0.1 g · kg-1 body weight · d-1 (BCAA) or high-protein (27%) with placebo (HP). The subjects underwent 16 wk of dietary intervention with provision of meals and supplements, followed by 8 wk of weight maintenance with provision of supplements only. One-way ANOVA analysis was conducted to analyze the primary (lean mass and insulin sensitivity) and secondary outcomes (anthropometric and metabolic parameters) among the 3 groups. Paired t-test was used to analyze the change in each group. RESULTS The 3 groups demonstrated similar significant reductions in body weight (7.97%), fat mass (13.8%), and waist circumference (7.27%) after 16 wk of energy deficit. Lean mass loss in BCAA (4.39%) tended to be lower than in CT (5.39%) and higher compared with HP (3.67%) (P = 0.06). Calf muscle volume increased 3.4% in BCAA and intramyocellular lipids (IMCLs) decreased in BCAA (17%) and HP (18%) (P < 0.05) over 16 wk. During the 8 wk weight maintenance period, lean mass gain in BCAA (1.03%) tended to be lower compared with CT (1.58%) and higher than in HP (-0.002%) (P = 0.04). Lean mass gain differed significantly between CT and HP (P = 0.03). Insulin sensitivity and metabolic profiles did not differ among the groups throughout the study period. CONCLUSIONS BCAA supplementation does not preserve lean mass or affect insulin sensitivity in overweight and obese adults during weight loss. A higher protein diet may be more advantageous for lean mass preservation.
Collapse
Affiliation(s)
- Delicia S Q Ooi
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore
| | - Jennifer Q R Ling
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore
| | - Suresh Anand Sadananthan
- Clinical Nutrition Research Center, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - S Sendhil Velan
- Clinical Nutrition Research Center, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore.,Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore
| | - Fang Yi Ong
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - E Shyong Tai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christiani Jeyakumar Henry
- Clinical Nutrition Research Center, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Melvin K S Leow
- Clinical Nutrition Research Center, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore.,Department of Endocrinology, Division of Medicine, Tan Tock Seng Hospital, Singapore
| | - Eric Y H Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chuen Seng Tan
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Yung Seng Lee
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Mary F F Chong
- Clinical Nutrition Research Center, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| |
Collapse
|
84
|
Jachthuber Trub C, Balikcioglu M, Freemark M, Bain J, Muehlbauer M, Ilkayeva O, White PJ, Armstrong S, Østbye T, Grambow S, Gumus Balikcioglu P. Impact of lifestyle Intervention on branched-chain amino acid catabolism and insulin sensitivity in adolescents with obesity. ENDOCRINOLOGY DIABETES & METABOLISM 2021; 4:e00250. [PMID: 34277974 PMCID: PMC8279626 DOI: 10.1002/edm2.250] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Revised: 02/16/2021] [Accepted: 03/13/2021] [Indexed: 12/30/2022]
Abstract
Insulin resistance in adolescents with obesity associates with a sex‐dependent metabolic ‘signature’ comprising branched‐chain amino acids (BCAAs), glutamate and C3/C5 acylcarnitines (C3/C5), implicating altered flux through BCAA catabolic pathways. Here, we investigated the effects of lifestyle intervention on BCAA catabolism and insulin sensitivity. We hypothesized (1) weight reduction and improved insulin sensitivity associate with enhanced BCAA catabolism; (2) baseline BCAAs and their metabolic by‐products predict changes in weight and insulin sensitivity during lifestyle intervention.
Collapse
Affiliation(s)
| | | | - Michael Freemark
- Division of Pediatric Endocrinology and Diabetes and the Duke Molecular Physiology Institute Duke University Medical Center Durham NC USA
| | - James Bain
- Duke Molecular Physiology Institute Duke Molecular Physiology Institute Duke University Medical Center Durham NC USA
| | - Michael Muehlbauer
- Duke Molecular Physiology Institute Duke Molecular Physiology Institute Duke University Medical Center Durham NC USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute Duke Molecular Physiology Institute Duke University Medical Center Durham NC USA
| | - Phillip J White
- Duke Molecular Physiology Institute Duke Molecular Physiology Institute Duke University Medical Center Durham NC USA
| | - Sarah Armstrong
- Division of General Pediatrics Duke University Medical Center Durham NC USA.,Department of Family Medicine and Community Health Duke University Medical Center Durham NC USA.,Department of Population Health Sciences Duke University Medical Center Durham NC USA.,Duke Clinical Research Institute Duke University Medical Center Durham NC USA
| | - Truls Østbye
- Department of Family Medicine and Community Health Duke University Medical Center Durham NC USA
| | - Steven Grambow
- Department of Biostatistics and Bioinformatics Duke University Medical Center Durham NC USA
| | - Pinar Gumus Balikcioglu
- Division of Pediatric Endocrinology and Diabetes and the Duke Molecular Physiology Institute Duke University Medical Center Durham NC USA
| |
Collapse
|
85
|
Vanweert F, de Ligt M, Hoeks J, Hesselink MKC, Schrauwen P, Phielix E. Elevated Plasma Branched-Chain Amino Acid Levels Correlate With Type 2 Diabetes-Related Metabolic Disturbances. J Clin Endocrinol Metab 2021; 106:e1827-e1836. [PMID: 33079174 DOI: 10.1210/clinem/dgaa751] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/14/2020] [Indexed: 01/14/2023]
Abstract
CONTEXT Patients with type 2 diabetes mellitus (T2DM) have elevated plasma branched-chain amino acid (BCAA) levels. The underlying cause, however, is not known. Low mitochondrial oxidation of BCAA levels could contribute to higher plasma BCAA levels. OBJECTIVE We aimed to investigate ex vivo muscle mitochondrial oxidative capacity and in vivo BCAA oxidation measured by whole-body leucine oxidation rates in patients with T2DM, first-degree relatives (FDRs), and control participants (CONs) with overweight or obesity. DESIGN AND SETTING An observational, community-based study was conducted. PARTICIPANTS Fifteen patients with T2DM, 13 FDR, and 17 CONs were included (age, 40-70 years; body mass index, 27-35 kg/m2). MAIN OUTCOME MEASURES High-resolution respirometry was used to examine ex vivo mitochondrial oxidative capacity in permeabilized muscle fibers. A subgroup of 5 T2DM patients and 5 CONs underwent hyperinsulinemic-euglycemic clamps combined with 1-13C leucine-infusion to determine whole-body leucine oxidation. RESULTS Total BCAA levels were higher in patients with T2DM compared to CONs, but not in FDRs, and correlated negatively with muscle mitochondrial oxidative capacity (r = -0.44, P < .001). Consistently, whole-body leucine oxidation rate was lower in patients with T2DM vs CON under basal conditions (0.202 ± 0.049 vs 0.275 ± 0.043 μmol kg-1 min-1, P < .05) and tended to be lower during high insulin infusion (0.326 ± 0.024 vs 0.382 ± 0.013 μmol kg-1 min-1, P = .075). CONCLUSIONS In patients with T2DM, a compromised whole-body leucine oxidation rate supports our hypothesis that higher plasma BCAA levels may originate at least partly from a low mitochondrial oxidative capacity.
Collapse
Affiliation(s)
- Froukje Vanweert
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Marlies de Ligt
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Matthijs K C Hesselink
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, Maastricht University and Medical Center, Maastricht, the Netherlands
- NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University and Medical Center, Maastricht, the Netherlands
| |
Collapse
|
86
|
Díaz-Rizzolo DA, Serra A, Colungo C, Sala-Vila A, Sisó-Almirall A, Gomis R. Type 2 diabetes preventive effects with a 12-months sardine-enriched diet in elderly population with prediabetes: An interventional, randomized and controlled trial. Clin Nutr 2021; 40:2587-2598. [PMID: 33932804 DOI: 10.1016/j.clnu.2021.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Fish could play a role in preventing type 2 diabetes (T2D) but there has been little specification about the type of fish and the preventive mechanism involved in its health claim. The sardine is a source of omega-3 and taurine that, in isolation or in synergy, would produce T2D-delaying through different molecular mechanism. HYPOTHESIS The consumption of twice a week of sardine, during one year would reduce T2D-developing risk in a population with prediabetes (preDM) and old age. DESIGN 152 subjects with fasting glucose between 100-124 mg/dL aged ≥65 yo were recruited from three primary care centers in Barcelona and were randomly distributed among two interventional groups: control group (CG) and sardine group (SG). Both groups received same T2D-prevention nutritional during a year but only SG had to add 200 g of sardine per week. All variables were collected before to start and at the end of the diet. (ClinicalTrials.gov: NCT03557541). RESULTS 152 people were randomized into CG (n=77) and SG (n=75) with 18 and 12 drop outs respectively. Subjects in SG, significantly compared to CG, decreased percentage classified-individuals in a very high risk group to develop T2D according to FINDRISC (p=0.035). In addition to increasing HDL-cholesterol and adiponectin and decreasing triglycerides (p<0.05) and blood pressure (<0.05), SG showed a lower HOMA-IR (p=0.032). The consumption of sardine characteristics nutrients as omega-3, EPA and DHA, vitamin D, fluorine and taurine were higher for SG (p<0.05). These results agreed with the increased of taurine, fatty acid (FA) omega-3 and bile acids circulating metabolites (p<0.05). Changes erythrocyte membrane FA were detected only in SG with a decrease of 5 omega-6 FA (p<0.001) and an increase of 3 omega-3 FA types (p<0.001). CONCLUSION We conclude that a year T2D-prevention diet with sardine supplementation has a greater protective effect against developing T2D and CV events.
Collapse
Affiliation(s)
- D A Díaz-Rizzolo
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) - Hospital Clinic of Barcelona, Barcelona, Spain; Universitat Oberta de Catalunya, Barcelona, Spain; Primary Healthcare Transversal Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Spain.
| | - A Serra
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) - Hospital Clinic of Barcelona, Barcelona, Spain
| | - C Colungo
- Primary Healthcare Transversal Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Spain; Primary Care Centre, CAPSBE, Barcelona, Spain
| | - A Sala-Vila
- IMIM - Hospital del Mar Medical Research Institute, Barcelona, Spain; Fatty Acid Research Institute, Sioux Falls, SD, USA
| | - A Sisó-Almirall
- Primary Healthcare Transversal Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Spain; Primary Care Centre, CAPSBE, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - R Gomis
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) - Hospital Clinic of Barcelona, Barcelona, Spain; Universitat Oberta de Catalunya, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain; Department of Endocrinology and Nutrition, Hospital Clinic of Barcelona, Barcelona, Spain
| |
Collapse
|
87
|
Brennan L, de Roos B. Nutrigenomics: lessons learned and future perspectives. Am J Clin Nutr 2021; 113:503-516. [PMID: 33515029 DOI: 10.1093/ajcn/nqaa366] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
The omics technologies of metabolomics, transcriptomics, proteomics, and metagenomics are playing an increasingly important role in nutrition science. With the emergence of the concept of precision nutrition and the need to understand individual responses to dietary interventions, it is an opportune time to examine the impact of these tools to date in human nutrition studies. Advances in our mechanistic understanding of dietary interventions were realized through incorporation of metabolomics, proteomics, and, more recently, metagenomics. A common observation across the studies was the low intra-individual variability of the omics measurements and the high inter-individual variation. Harnessing this data for use in the development of precision nutrition will be important. Metabolomics in particular has played a key role in the development of biomarkers of food intake in an effort to enhance the accuracy of dietary assessments. Further work is needed to realize the full potential of such biomarkers and to demonstrate integration with current strategies, with the goal of overcoming the well-established limitations of self-reported approaches. Although many of the nutrigenomic studies performed to date were labelled as proof-of-concept or pilot studies, there is ample evidence to support the use of these technologies in nutrition science. Incorporating omic technologies from the start of study designs will ensure that studies are sufficiently powered for such data. Furthermore, multi-disciplinary collaborations are likely to become even more important to aid analyses and interpretation of the data.
Collapse
Affiliation(s)
- Lorraine Brennan
- Institute of Food and Health and Conway Institute, University College Dublin (UCD) School of Agriculture and Food Science, UCD, Belfield, Dublin, Ireland
| | - Baukje de Roos
- The Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| |
Collapse
|
88
|
McCann JR, Bihlmeyer NA, Roche K, Catherine C, Jawahar J, Kwee LC, Younge NE, Silverman J, Ilkayeva O, Sarria C, Zizzi A, Wootton J, Poppe L, Anderson P, Arlotto M, Wei Z, Granek JA, Valdivia RH, David LA, Dressman HK, Newgard CB, Shah SH, Seed PC, Rawls JF, Armstrong SC. The Pediatric Obesity Microbiome and Metabolism Study (POMMS): Methods, Baseline Data, and Early Insights. Obesity (Silver Spring) 2021; 29:569-578. [PMID: 33624438 PMCID: PMC7927749 DOI: 10.1002/oby.23081] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The purpose of this study was to establish a biorepository of clinical, metabolomic, and microbiome samples from adolescents with obesity as they undergo lifestyle modification. METHODS A total of 223 adolescents aged 10 to 18 years with BMI ≥95th percentile were enrolled, along with 71 healthy weight participants. Clinical data, fasting serum, and fecal samples were collected at repeated intervals over 6 months. Herein, the study design, data collection methods, and interim analysis-including targeted serum metabolite measurements and fecal 16S ribosomal RNA gene amplicon sequencing among adolescents with obesity (n = 27) and healthy weight controls (n = 27)-are presented. RESULTS Adolescents with obesity have higher serum alanine aminotransferase, C-reactive protein, and glycated hemoglobin, and they have lower high-density lipoprotein cholesterol when compared with healthy weight controls. Metabolomics revealed differences in branched-chain amino acid-related metabolites. Also observed was a differential abundance of specific microbial taxa and lower species diversity among adolescents with obesity when compared with the healthy weight group. CONCLUSIONS The Pediatric Metabolism and Microbiome Study (POMMS) biorepository is available as a shared resource. Early findings suggest evidence of a metabolic signature of obesity unique to adolescents, along with confirmation of previously reported findings that describe metabolic and microbiome markers of obesity.
Collapse
Affiliation(s)
- Jessica R. McCann
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
| | - Nathan A. Bihlmeyer
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC, USA 27710
| | - Kimberly Roche
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
| | | | - Jayanth Jawahar
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
| | - Lydia Coulter Kwee
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC, USA 27710
| | - Noelle E. Younge
- Department of Pediatrics, Duke University, Durham, NC, USA 27710
| | | | - Olga Ilkayeva
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC, USA 27710
| | - Charles Sarria
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
| | - Alexandra Zizzi
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
| | - Janet Wootton
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
| | - Lisa Poppe
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC, USA 27710
| | - Paul Anderson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC, USA 27710
| | - Michelle Arlotto
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC, USA 27710
| | - Zhengzheng Wei
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
| | - Joshua A. Granek
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
- Duke Microbiome Center, Duke University Durham, NC, USA 27710
| | - Raphael H. Valdivia
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
- Duke Microbiome Center, Duke University Durham, NC, USA 27710
| | - Lawrence A. David
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
- Duke Microbiome Center, Duke University Durham, NC, USA 27710
| | - Holly K. Dressman
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
- Duke Microbiome Center, Duke University Durham, NC, USA 27710
| | - Christopher B. Newgard
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC, USA 27710
| | - Svati H. Shah
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, NC, USA 27710
- Duke Clinical Research Institute, Duke University, Durham, NC, USA 27710
| | - Patrick C. Seed
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Stanley Manne Children’s Research Institute, Northwestern University Medical Center, Chicago, IL, USA 60611
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708
- Duke Microbiome Center, Duke University Durham, NC, USA 27710
| | - Sarah C. Armstrong
- Department of Pediatrics, Duke University, Durham, NC, USA 27710
- Duke Clinical Research Institute, Duke University, Durham, NC, USA 27710
| |
Collapse
|
89
|
Kwee LC, Ilkayeva O, Muehlbauer MJ, Bihlmeyer N, Wolfe B, Purnell JQ, Xavier Pi-Sunyer F, Chen H, Bahnson J, Newgard CB, Shah SH, Laferrère B. Metabolites and diabetes remission after weight loss. Nutr Diabetes 2021; 11:10. [PMID: 33627633 PMCID: PMC7904757 DOI: 10.1038/s41387-021-00151-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
There is marked heterogeneity in the response to weight loss interventions with regards to weight loss amount and metabolic improvement. We sought to identify biomarkers predictive of type 2 diabetes remission and amount of weight loss in individuals with severe obesity enrolled in the Longitudinal Assessment of Bariatric Surgery (LABS) and the Look AHEAD (Action for Health in Diabetes) studies. Targeted mass spectrometry-based profiling of 135 metabolites was performed in pre-intervention blood samples using a nested design for diabetes remission over five years (n = 93 LABS, n = 80 Look AHEAD; n = 87 remitters), and for extremes of weight loss at five years (n = 151 LABS; n = 75 with high weight loss). Principal components analysis (PCA) was used for dimensionality reduction, with PCA-derived metabolite factors tested for association with both diabetes remission and weight loss. Metabolic markers were tested for incremental improvement to clinical models, including the DiaRem score. Two metabolite factors were associated with diabetes remission: one primarily composed of branched chain amino acids (BCAA) and tyrosine (odds ratio (95% confidence interval) [OR (95% CI)] = 1.4 [1.0–1.9], p = 0.045), and one with betaine and choline (OR [95% CI] = 0.7 [0.5–0.9], p = 0.02).These results were not significant after adjustment for multiple tests. Inclusion of these two factors in clinical models yielded modest improvements in model fit and performance: in a constructed clinical model, the C-statistic improved from 0.87 to 0.90 (p = 0.02), while the net reclassification index showed improvement in prediction compared to the DiaRem score (NRI = 0.26, p = 0.0013). No metabolite factors associated with weight loss at five years. Baseline levels of metabolites in the BCAA and trimethylamine-N-oxide (TMAO)-microbiome-related pathways are independently and incrementally associated with sustained diabetes remission after weight loss interventions in individuals with severe obesity. These metabolites could serve as clinically useful biomarkers to identify individuals who will benefit the most from weight loss interventions.
Collapse
Affiliation(s)
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Durham, NC, USA.,Sarah W. Stedman Nutrition and Metabolism Center, Durham, NC, USA
| | - Michael J Muehlbauer
- Duke Molecular Physiology Institute, Durham, NC, USA.,Sarah W. Stedman Nutrition and Metabolism Center, Durham, NC, USA
| | | | - Bruce Wolfe
- Departments of Surgery and Medicine, Oregon Health & Science University,, Portland, OR, USA
| | - Jonathan Q Purnell
- Departments of Surgery and Medicine, Oregon Health & Science University,, Portland, OR, USA
| | - F Xavier Pi-Sunyer
- New York Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Haiying Chen
- Department of Biostatistics and Data Science, Wake Forest School of Medicine Medical Center, Winston-Salem, NC, USA
| | - Judy Bahnson
- Department of Biostatistics and Data Science, Wake Forest School of Medicine Medical Center, Winston-Salem, NC, USA
| | - Christopher B Newgard
- Duke Molecular Physiology Institute, Durham, NC, USA.,Sarah W. Stedman Nutrition and Metabolism Center, Durham, NC, USA.,Department of Pharmacology & Cancer Biology and Division of Endocrinology, Department of Medicine, Duke University, Durham, DC, USA
| | - Svati H Shah
- Duke Molecular Physiology Institute, Durham, NC, USA.,Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, DC, USA
| | - Blandine Laferrère
- New York Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
90
|
Al-Mrabeh A. β-Cell Dysfunction, Hepatic Lipid Metabolism, and Cardiovascular Health in Type 2 Diabetes: New Directions of Research and Novel Therapeutic Strategies. Biomedicines 2021; 9:226. [PMID: 33672162 PMCID: PMC7927138 DOI: 10.3390/biomedicines9020226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/09/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) remains a major problem for people with type 2 diabetes mellitus (T2DM), and dyslipidemia is one of the main drivers for both metabolic diseases. In this review, the major pathophysiological and molecular mechanisms of β-cell dysfunction and recovery in T2DM are discussed in the context of abnormal hepatic lipid metabolism and cardiovascular health. (i) In normal health, continuous exposure of the pancreas to nutrient stimulus increases the demand on β-cells. In the long term, this will not only stress β-cells and decrease their insulin secretory capacity, but also will blunt the cellular response to insulin. (ii) At the pre-diabetes stage, β-cells compensate for insulin resistance through hypersecretion of insulin. This increases the metabolic burden on the stressed β-cells and changes hepatic lipoprotein metabolism and adipose tissue function. (iii) If this lipotoxic hyperinsulinemic environment is not removed, β-cells start to lose function, and CVD risk rises due to lower lipoprotein clearance. (iv) Once developed, T2DM can be reversed by weight loss, a process described recently as remission. However, the precise mechanism(s) by which calorie restriction causes normalization of lipoprotein metabolism and restores β-cell function are not fully established. Understanding the pathophysiological and molecular basis of β-cell failure and recovery during remission is critical to reduce β-cell burden and loss of function. The aim of this review is to highlight the link between lipoprotein export and lipid-driven β-cell dysfunction in T2DM and how this is related to cardiovascular health. A second aim is to understand the mechanisms of β-cell recovery after weight loss, and to explore new areas of research for developing more targeted future therapies to prevent T2DM and the associated CVD events.
Collapse
Affiliation(s)
- Ahmad Al-Mrabeh
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Magnetic Resonance Centre, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
91
|
Cifarelli V, Beeman SC, Smith GI, Yoshino J, Morozov D, Beals JW, Kayser BD, Watrous JD, Jain M, Patterson BW, Klein S. Decreased adipose tissue oxygenation associates with insulin resistance in individuals with obesity. J Clin Invest 2021; 130:6688-6699. [PMID: 33164985 DOI: 10.1172/jci141828] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUNDData from studies conducted in rodent models have shown that decreased adipose tissue (AT) oxygenation is involved in the pathogenesis of obesity-induced insulin resistance. Here, we evaluated the potential influence of AT oxygenation on AT biology and insulin sensitivity in people.METHODSWe evaluated subcutaneous AT oxygen partial pressure (pO2); liver and whole-body insulin sensitivity; AT expression of genes and pathways involved in inflammation, fibrosis, and branched-chain amino acid (BCAA) catabolism; systemic markers of inflammation; and plasma BCAA concentrations, in 3 groups of participants that were rigorously stratified by adiposity and insulin sensitivity: metabolically healthy lean (MHL; n = 11), metabolically healthy obese (MHO; n = 15), and metabolically unhealthy obese (MUO; n = 20).RESULTSAT pO2 progressively declined from the MHL to the MHO to the MUO group, and was positively associated with hepatic and whole-body insulin sensitivity. AT pO2 was positively associated with the expression of genes involved in BCAA catabolism, in conjunction with an inverse relationship between AT pO2 and plasma BCAA concentrations. AT pO2 was negatively associated with AT gene expression of markers of inflammation and fibrosis. Plasma PAI-1 increased from the MHL to the MHO to the MUO group and was negatively correlated with AT pO2, whereas the plasma concentrations of other cytokines and chemokines were not different among the MHL and MUO groups.CONCLUSIONThese results support the notion that reduced AT oxygenation in individuals with obesity contributes to insulin resistance by increasing plasma PAI-1 concentrations and decreasing AT BCAA catabolism and thereby increasing plasma BCAA concentrations.TRIAL REGISTRATIONClinicalTrials.gov NCT02706262.FUNDINGThis study was supported by NIH grants K01DK109119, T32HL130357, K01DK116917, R01ES027595, P42ES010337, DK56341 (Nutrition Obesity Research Center), DK20579 (Diabetes Research Center), DK052574 (Digestive Disease Research Center), and UL1TR002345 (Clinical and Translational Science Award); NIH Shared Instrumentation Grants S10RR0227552, S10OD020025, and S10OD026929; and the Foundation for Barnes-Jewish Hospital.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, and
| | - Scott C Beeman
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, and.,Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gordon I Smith
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, and
| | - Jun Yoshino
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, and
| | - Darya Morozov
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joseph W Beals
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, and
| | - Brandon D Kayser
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, and
| | - Jeramie D Watrous
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Mohit Jain
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Bruce W Patterson
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, and
| | - Samuel Klein
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, and
| |
Collapse
|
92
|
Mikkola TM, Salonen MK, Kajantie E, Kautiainen H, Eriksson JG. Associations of Fat and Lean Body Mass with Circulating Amino Acids in Older Men and Women. J Gerontol A Biol Sci Med Sci 2021; 75:885-891. [PMID: 31095700 DOI: 10.1093/gerona/glz126] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Indexed: 12/16/2022] Open
Abstract
Circulating amino acids are potential markers of body composition. Previous studies are mainly limited to middle age and focus on either fat or lean mass, thereby ignoring overall body composition. We investigated the associations of fat and lean body mass with circulating amino acids in older men and women. We studied 594 women and 476 men from the Helsinki Birth Cohort Study (age 62-74 years). Bioelectrical impedance analysis was used to indicate two main body compartments by fat (fat mass/height2) and lean mass indices (lean mass/height2), dichotomized based on sex-specific medians. Eight serum amino acids were quantified using nuclear magnetic resonance spectroscopy. General linear models were adjusted for age, smoking, and fasting glucose. Higher lean mass index (LMI) was associated with higher concentrations of branched-chain amino acids in both sexes (p ≤ .001). In men, LMI was also positively associated with tyrosine (p = .006) and inversely with glycine (p < .001). Higher fat mass index was associated with higher concentrations of all branched-chain amino acids, aromatic amino acids (phenylalanine and tyrosine), and alanine in both sexes (p ≤ .008). Associations between body composition and amino acids are largely similar in older men and women. The associations are largely similar to those previously observed in younger adults.
Collapse
Affiliation(s)
| | - Minna K Salonen
- Folkhälsan Research Center, Helsinki, Finland.,Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Eero Kajantie
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland.,PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Finland.,Department of Clinical and Molecular Medicine, Norwegian University for Science and Technology, Trondheim, Norway.,Children's Hospital, Helsinki University Hospital and University of Helsinki, Finland
| | - Hannu Kautiainen
- Folkhälsan Research Center, Helsinki, Finland.,Primary Health Care Unit, Kuopio University Hospital, Finland
| | - Johan G Eriksson
- Folkhälsan Research Center, Helsinki, Finland.,Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Finland.,Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research.,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore
| |
Collapse
|
93
|
Lee S, Gulseth HL, Langleite TM, Norheim F, Olsen T, Refsum H, Jensen J, Birkeland KI, Drevon CA. Branched-chain amino acid metabolism, insulin sensitivity and liver fat response to exercise training in sedentary dysglycaemic and normoglycaemic men. Diabetologia 2021; 64:410-423. [PMID: 33123769 PMCID: PMC7801320 DOI: 10.1007/s00125-020-05296-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022]
Abstract
AIMS/HYPOTHESIS Obesity and insulin resistance may be associated with elevated plasma concentration of branched-chain amino acids (BCAAs) and impaired BCAA metabolism. However, it is unknown whether the insulin-sensitising effect of long-term exercise can be explained by concomitant change in BCAAs and their metabolism. METHODS We included 26 sedentary overweight and normal-weight middle-aged men from the MyoGlu clinical trial, with or without dysglycaemia, for 12 weeks of supervised intensive exercise intervention, including two endurance and two resistance sessions weekly. Insulin sensitivity was measured as the glucose infusion rate (GIR) from a hyperinsulinaemic-euglycaemic clamp. In addition, maximum oxygen uptake, upper and lower body strength and adipose tissue depots (using MRI and spectroscopy) were measured, and subcutaneous white adipose tissue (ScWAT) and skeletal muscle (SkM) biopsies were harvested both before and after the 12 week intervention. In the present study we have measured plasma BCAAs and related metabolites using CG-MS/MS and HPLC-MS/MS, and performed global mRNA-sequencing pathway analysis on ScWAT and SkM. RESULTS In MyoGlu, men with dysglycaemia displayed lower GIR, more fat mass and higher liver fat content than normoglycaemic men at baseline, and 12 weeks of exercise increased GIR, improved body composition and reduced liver fat content similarly for both groups. In our current study we observed higher plasma concentrations of BCAAs (14.4%, p = 0.01) and related metabolites, such as 3-hydroxyisobutyrate (19.4%, p = 0.034) in dysglycaemic vs normoglycaemic men at baseline. Baseline plasma BCAA levels correlated negatively to the change in GIR (ρ = -0.41, p = 0.037) and [Formula: see text] (ρ = -0.47, p = 0.015) after 12 weeks of exercise and positively to amounts of intraperitoneal fat (ρ = 0.40, p = 0.044) and liver fat (ρ = 0.58, p = 0.01). However, circulating BCAAs and related metabolites did not respond to 12 weeks of exercise, with the exception of isoleucine, which increased in normoglycaemic men (10 μmol/l, p = 0.01). Pathway analyses of mRNA-sequencing data implied reduced BCAA catabolism in both SkM and ScWAT in men with dysglycaemia compared with men with normoglycaemia at baseline. Gene expression levels related to BCAA metabolism correlated positively with GIR and markers of mitochondrial content in both SkM and ScWAT, and negatively with fat mass generally, and particularly with intraperitoneal fat mass. mRNA-sequencing pathway analysis also implied increased BCAA metabolism after 12 weeks of exercise in both groups and in both tissues, including enhanced expression of the gene encoding branched-chain α-ketoacid dehydrogenase (BCKDH) and reduced expression of the BCKDH phosphatase in both groups and tissues. Gene expression of SLC25A44, which encodes a mitochondrial BCAA transporter, was increased in SkM in both groups, and gene expression of BCKDK, which encodes BCKDH kinase, was reduced in ScWAT in dysglycaemic men. Mediation analyses indicated a pronounced effect of enhanced SkM (~53%, p = 0.022), and a moderate effect of enhanced ScWAT (~18%, p = 0.018) BCAA metabolism on improved insulin sensitivity after 12 weeks of exercise, based on mRNA sequencing. In comparison, plasma concentration of BCAAs did not mediate any effect in this regard. CONCLUSION/INTERPRETATION Plasma BCAA concentration was largely unresponsive to long-term exercise and unrelated to exercise-induced insulin sensitivity. On the other hand, the insulin-sensitising effect of long-term exercise in men may be explained by enhanced SkM and, to a lesser degree, also by enhanced ScWAT BCAA catabolism. Graphical abstract.
Collapse
Affiliation(s)
- Sindre Lee
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway.
| | - Hanne L Gulseth
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, Oslo, Norway
| | - Torgrim M Langleite
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Kåre I Birkeland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
94
|
Abstract
Sensing and responding to changes in nutrient levels, including those of glucose, lipids, and amino acids, by the body is necessary for survival. Accordingly, perturbations in nutrient sensing are tightly linked with human pathologies, particularly metabolic diseases such as obesity, type 2 diabetes mellitus, and other complications of metabolic syndromes. The conventional view is that amino acids are fundamental elements for protein and peptide synthesis, while recent studies have revealed that amino acids are also important bioactive molecules that play key roles in signaling pathways and metabolic regulation. Different pathways that sense intracellular and extracellular levels of amino acids are integrated and coordinated at the organismal level, and, together, these pathways maintain whole metabolic homeostasis. In this review, we discuss the studies describing how important sensing signals respond to amino acid availability and how these sensing mechanisms modulate metabolic processes, including energy, glucose, and lipid metabolism. We further discuss whether dysregulation of amino acid sensing signals can be targeted to promote metabolic disorders, and discuss how to translate these mechanisms to treat human diseases. This review will help to enhance our overall understanding of the correlation between amino acid sensing and metabolic homeostasis, which have important implications for human health.
Collapse
Affiliation(s)
- Xiaoming Hu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
95
|
Abstract
Atherosclerotic cardiovascular disease (ASCVD) proceeds through a series of stages: initiation, progression (or regression), and complications. By integrating known biology regarding molecular signatures of each stage with recent advances in high-dimensional molecular data acquisition platforms (to assay the genome, epigenome, transcriptome, proteome, metabolome, and gut microbiome), snapshots of each phase of atherosclerotic cardiovascular disease development can be captured. In this review, we will summarize emerging approaches for assessment of atherosclerotic cardiovascular disease risk in humans using peripheral blood molecular signatures and molecular imaging approaches. We will then discuss the potential (and challenges) for these snapshots to be integrated into a personalized movie providing dynamic readouts of an individual's atherosclerotic cardiovascular disease risk status throughout the life course.
Collapse
Affiliation(s)
- Matthew Nayor
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Kemar J. Brown
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ramachandran S. Vasan
- Sections of Preventive Medicine & Epidemiology, and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA; Department of Epidemiology, Boston University School of Public Health; Boston University Center for Computing and Data Sciences
| |
Collapse
|
96
|
Medehouenou TCM, Roy C, Tremblay PY, St-Jean A, Meziou S, Muckle G, Ayotte P, Lucas M. Metabolic features of adiposity and glucose homoeostasis among school-aged inuit children from Nunavik (Northern Quebec, Canada). Int J Circumpolar Health 2021; 80:1858605. [PMID: 33395372 PMCID: PMC7801047 DOI: 10.1080/22423982.2020.1858605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
In contrast to most Indigenous people in Canada, Inuit appeared until recently to have been protected from type 2 diabetes (T2D) related to obesity. We assessed the associations of metabolites (amino acids, acylcarnitines) with adiposity and biomarkers of T2D in school-aged Inuit children of Nunavik (Canada). Concentrations of metabolite were measured in plasma samples from a cross-sectional analysis of 248 children (mean age = 10.8 years). We assessed associations of plasma metabolites with adiposity measures (BMI, skinfold thicknesses) and T2D markers (insulin, glucose, adiponectin). Plasma concentrations of valine and tyrosine were higher in obese and overweight children compared to those of normal weight children (P < 0.05). An increment of 1-SD in BMI (SD = 3.3 kg/m2) was statistically associated with an increment of 0.21 (95% CI: 0.08, 0.33) for valine, 0.15 (95% CI: 0.02, 0.27) for isoleucine and 0.17 (95% CI: 0.04, 0.29) for tyrosine. Insulin concentration increased with concentrations of all amino acids (P < 0.05) except methionine. None of the acylcarnitines measured were statistically significantly associated with adiposity or T2D biomarkers A signature of metabolites, particularly higher levels of branched-chain amino acids, might allow for early detection of T2D among school-aged Inuit children.
Collapse
Affiliation(s)
- Thierry Comlan Marc Medehouenou
- Population Health and Optimal Health Practices Research Unit, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval , Québec, Quebec, Canada
| | - Cynthia Roy
- Population Health and Optimal Health Practices Research Unit, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval , Québec, Quebec, Canada.,Centre de Toxicologie du Québec, INSPQ , Québec, Quebec, Canada
| | - Pierre-Yves Tremblay
- Population Health and Optimal Health Practices Research Unit, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval , Québec, Quebec, Canada.,Centre de Toxicologie du Québec, INSPQ , Québec, Quebec, Canada
| | - Audray St-Jean
- Population Health and Optimal Health Practices Research Unit, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval , Québec, Quebec, Canada
| | - Salma Meziou
- Population Health and Optimal Health Practices Research Unit, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval , Québec, Quebec, Canada
| | - Gina Muckle
- Population Health and Optimal Health Practices Research Unit, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval , Québec, Quebec, Canada.,School of Psychology, Université Laval , Québec, Quebec, Canada
| | - Pierre Ayotte
- Population Health and Optimal Health Practices Research Unit, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval , Québec, Quebec, Canada.,Centre de Toxicologie du Québec, INSPQ , Québec, Quebec, Canada.,Department of Social and Preventive Medicine, Université Laval , Québec, Quebec, Canada
| | - Michel Lucas
- Population Health and Optimal Health Practices Research Unit, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval , Québec, Quebec, Canada.,Department of Social and Preventive Medicine, Université Laval , Québec, Quebec, Canada
| |
Collapse
|
97
|
Ramzan I, Taylor M, Phillips B, Wilkinson D, Smith K, Hession K, Idris I, Atherton P. A Novel Dietary Intervention Reduces Circulatory Branched-Chain Amino Acids by 50%: A Pilot Study of Relevance for Obesity and Diabetes. Nutrients 2020; 13:nu13010095. [PMID: 33396718 PMCID: PMC7824725 DOI: 10.3390/nu13010095] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 11/16/2022] Open
Abstract
Elevated circulating branched-chain amino acids (BCAAs; isoleucine, leucine, and valine) are associated with obesity and type 2 diabetes (T2D). Reducing circulatory BCAAs by dietary restriction was suggested to mitigate these risks in rodent models, but this is a challenging paradigm to deliver in humans. We aimed to design and assess the feasibility of a diet aimed at reducing circulating BCAA concentrations in humans, while maintaining energy balance and overall energy/protein intake. Twelve healthy individuals were assigned to either a 7-day BCAA-restricted diet or a 7-day control diet. Diets were iso-nitrogenous and iso-caloric, with only BCAA levels differing between the two. The BCAA-restricted diet significantly reduced circulating BCAA concentrations by ~50% i.e., baseline 437 ± 60 to 217 ± 40 µmol/L (p < 0.005). Individually, both valine (245 ± 33 to 105 ± 23 µmol/L; p < 0.0001), and leucine (130 ± 20 to 75 ± 13 µmol/L; p < 0.05), decreased significantly in response to the BCAA-restricted diet. The BCAA-restricted diet marginally lowered Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) levels: baseline 1.5 ± 0.2 to 1.0 ± 0.1; (p = 0.096). We successfully lowered circulating BCAAs by 50% while maintaining iso-nitrogenous, iso-caloric dietary intakes, and while meeting the recommended daily allowances (RDA) for protein requirements. The present pilot study represents a novel dietary means by which to reduce BCAA, and as such, provides a blueprint for a potential dietary therapeutic in obesity/diabetes.
Collapse
|
98
|
Mahbub MH, Yamaguchi N, Hase R, Takahashi H, Ishimaru Y, Watanabe R, Saito H, Shimokawa J, Yamamoto H, Kikuchi S, Tanabe T. Plasma Branched-Chain and Aromatic Amino Acids in Relation to Hypertension. Nutrients 2020; 12:nu12123791. [PMID: 33322015 PMCID: PMC7764357 DOI: 10.3390/nu12123791] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Findings of the available studies regarding the roles of branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs) in hypertension are inconsistent, conflicting and inconclusive. The purpose of this study was to explore and clarify the existence of any relationships of individual BCAAs and AAAs with hypertension with adjustments for potential relevant confounders. A total of 2805 healthy controls and 2736 hypertensive patients were included in the current analysis. The associations between individual amino acids and hypertension were explored by logistic regression analyses adjusted for potential confounding variables. Among the investigated amino acids, only the BCAAs showed consistently significant positive associations with hypertension in the adjusted models (p-trend < 0.05 to 0.001). However, compared with the corresponding lowest quartile of individual BCAAs, the positive association with hypertension remained significant only in the highest quartile (p < 0.01 to 0.001). We confirmed in a relatively large cohort of subjects that BCAAs, not AAAs, demonstrated consistent positive associations with hypertension. The results display the promising potential for the use of BCAAs as relevant and accessible biomarkers, and provide perspectives on interventions directed towards the reduction in plasma BCAA levels in the prevention and management of hypertension.
Collapse
Affiliation(s)
- M. H. Mahbub
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
- Correspondence: ; Tel.: +81-836-22-2231
| | - Natsu Yamaguchi
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Ryosuke Hase
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hidekazu Takahashi
- Department of Public Health, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan;
| | - Yasutaka Ishimaru
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Rie Watanabe
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hiroyuki Saito
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Junki Shimokawa
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| | - Hiroshi Yamamoto
- Institute for Innovation, Ajinomoto Co. Inc., Kawasaki, Kanagawa 210-8681, Japan; (H.Y.); (S.K.)
| | - Shinya Kikuchi
- Institute for Innovation, Ajinomoto Co. Inc., Kawasaki, Kanagawa 210-8681, Japan; (H.Y.); (S.K.)
| | - Tsuyoshi Tanabe
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan; (N.Y.); (R.H.); (Y.I.); (R.W.); (H.S.); (J.S.); (T.T.)
| |
Collapse
|
99
|
Tan HC, Hsu JW, Kovalik JP, Eng A, Chan WH, Khoo CM, Tai ES, Chacko S, Jahoor F. Branched-Chain Amino Acid Oxidation Is Elevated in Adults with Morbid Obesity and Decreases Significantly after Sleeve Gastrectomy. J Nutr 2020; 150:3180-3189. [PMID: 33097955 DOI: 10.1093/jn/nxaa298] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/07/2020] [Accepted: 09/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Plasma concentrations of branched-chain amino acids (BCAAs) are elevated in obese individuals with insulin resistance (IR) and decrease after bariatric surgery. However, the metabolic mechanisms are unclear. OBJECTIVES Our objectives are to compare leucine kinetics between morbidly obese and healthy-weight individuals cross-sectionally, and to prospectively evaluate changes in the morbidly obese after sleeve gastrectomy. We hypothesized that leucine oxidation is slower in obese individuals and increases after surgery. METHODS Ten morbidly obese [BMI (in kg/m2) ≥32.5, age 21-50 y] and 10 healthy-weight participants (BMI <25), matched for age (median ∼30 y) but not gender, were infused with [U-13C6] leucine and [2H5] glycerol to quantify leucine and glycerol kinetics. Morbidly obese participants were studied again 6 mo postsurgery. Primary outcomes were kinetic parameters related to BCAA metabolism. Data were analyzed by nonparametric methods and presented as median (IQR). RESULTS Participants with obesity had IR with an HOMA-IR (4.89; 4.36-8.76) greater than that of healthy-weight participants (1.32; 0.99-1.49; P < 0.001) and had significantly faster leucine flux [218; 196-259 compared with 145; 138-149 μmol · kg fat-free mass (FFM)-1 · h-1], oxidation (24.0; 17.9-29.8 compared with 16.1; 14.3-18.5 μmol · kg FFM-1 · h-1), and nonoxidative disposal (204; 190-247 compared with 138; 129-140 μmol · kg FFM-1 · h-1) (P < 0.017 for all). After surgery, the morbidly obese had a marked improvement in IR (3.54; 3.06-6.08; P = 0.008) and significant reductions in BCAA concentrations (113; 95-157 μmol/L) and leucine oxidation (9.37; 6.85-15.2 μmol · kg FFM-1 · h-1) (P = 0.017 for both). Further, leucine flux in this group correlated significantly with IR (r = 0.78, P < 0.001). CONCLUSIONS BCAA oxidation is not impaired but elevated in individuals with morbid obesity. Plasma BCAA concentrations are lowered after surgery owing to slower breakdown of body proteins as insulin's ability to suppress proteolysis is restored. These findings suggest that IR is the underlying cause and not the consequence of elevated BCAAs in obesity.
Collapse
Affiliation(s)
- Hong Chang Tan
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Jean W Hsu
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jean-Paul Kovalik
- Cardiovascular and Metabolic Disease Program, Duke-NUS Medical School, Singapore
| | - Alvin Eng
- Department of Upper GI and Bariatric Surgery, Singapore General Hospital, Singapore
| | - Weng Hoong Chan
- Department of Upper GI and Bariatric Surgery, Singapore General Hospital, Singapore
| | - Chin Meng Khoo
- Department of Medicine, National University Health System, Singapore
| | - E Shyong Tai
- Department of Medicine, National University Health System, Singapore
| | - Shaji Chacko
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Farook Jahoor
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
100
|
Smith K, Bowden Davies KA, Stevenson EJ, West DJ. The Clinical Application of Mealtime Whey Protein for the Treatment of Postprandial Hyperglycaemia for People With Type 2 Diabetes: A Long Whey to Go. Front Nutr 2020; 7:587843. [PMID: 33195375 PMCID: PMC7607659 DOI: 10.3389/fnut.2020.587843] [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: 07/27/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Mitigating postprandial hyperglycaemic excursions may be effective in not only enhancing glycaemic control for people with type 2 diabetes but also reducing the onset of diabetes-related complications. However, there are growing concerns over the long-term efficacy of anti-hyperglycaemic pharmacotherapies, which coupled with their rising financial costs, underlines the need for further non-pharmaceutical treatments to regulate postprandial glycaemic excursions. One promising strategy that acutely improves postprandial glycaemia for people with type 2 diabetes is through the provision of mealtime whey protein, owing to the slowing of gastric emptying and increased secretion of insulin and the incretin peptides. The magnitude of this effect appears greater when whey protein is consumed before, rather than with, a meal. Herein, this dietary tool may offer a simple and inexpensive strategy in the management of postprandial hyperglycaemia for people with type 2 diabetes. However, there are insufficient long-term studies that have investigated the use of mealtime whey protein as a treatment option for individuals with type 2 diabetes. The methodological approaches applied in acute studies and outcomes reported may also not portray what is achievable long-term in practice. Therefore, studies are needed to refine the application of this mealtime strategy to maximize its clinical potential to treat hyperglycaemia and to apply these long-term to address key components of successful diabetes care. This review discusses evidence surrounding the provision of mealtime whey protein to treat postprandial hyperglycaemia in individuals with type 2 diabetes and highlights areas to help facilitate its clinical application.
Collapse
Affiliation(s)
- Kieran Smith
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kelly A Bowden Davies
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Emma J Stevenson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Daniel J West
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|