151
|
Modulation of Free Amino Acid Profile in Healthy Humans Administered with Mastiha Terpenes. An Open-Label Trial. Nutrients 2018; 10:nu10060715. [PMID: 29865269 PMCID: PMC6024789 DOI: 10.3390/nu10060715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/29/2018] [Accepted: 05/29/2018] [Indexed: 12/22/2022] Open
Abstract
We aimed to explore whether plasma-free amino acids are modified in response to terpenes administration in healthy humans. In this open-label, single-arm acute trial, seventeen healthy male volunteers were administered with a naturally occurring product of known terpenes-namely mastiha-after overnight fasting. Blood samples were collected at different time points before and after ingestion. We aimed at identifying and quantifying 60 free amino acids in plasma applying Gas Chromatography-Mass Spectrometry. A total of 24 free amino acids were quantified. Branched-chain valine significantly decreased 4 h post-ingestion, whereas proline decreased at 6 h and ornithine at 2 h, compared to 0 h. These novel findings demonstrate that free amino acids levels are modulated in response to terpenes intake in healthy subjects.
Collapse
|
152
|
A. R, Agrawal N, Kumar H, Kumar V. Emerging role of branched chain amino acids in metabolic disorders: A mechanistic review. PHARMANUTRITION 2018. [DOI: 10.1016/j.phanu.2018.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
153
|
Merz B, Frommherz L, Rist MJ, Kulling SE, Bub A, Watzl B. Dietary Pattern and Plasma BCAA-Variations in Healthy Men and Women-Results from the KarMeN Study. Nutrients 2018; 10:E623. [PMID: 29762522 PMCID: PMC5985475 DOI: 10.3390/nu10050623] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/08/2018] [Accepted: 05/11/2018] [Indexed: 01/04/2023] Open
Abstract
Branched-chain amino acids (BCAA) in plasma are discussed as risk factors for the onset of several diseases. Information about the contribution of the overall diet to plasma BCAA concentrations is controversial. Our objective was to investigate which dietary pattern is associated with plasma BCAA concentrations and whether other additional nutrients besides BCAA further characterize this dietary pattern. Based on the cross-sectional KarMeN study, fasting plasma amino acid (AA) concentrations, as well as current and habitual dietary intake were assessed in 298 healthy individuals. Using reduced rank regression, we derived a habitual dietary pattern that explained 32.5% of plasma BCAA variation. This pattern was high in meat, sausages, sauces, eggs, and ice cream but low in nuts, cereals, mushrooms, and pulses. The age, sex, and energy intake adjusted dietary pattern score was associated with an increase in animal-based protein together with a decrease in plant-based protein, dietary fiber, and an unfavorable fatty acid composition. Besides BCAA, alanine, lysine and the aromatic AA were positively associated with the dietary pattern score as well. All of these factors were reported to be associated with risk of type 2 diabetes and cardiovascular diseases before. Our data suggest that rather than the dietary intake of BCAA, the overall dietary pattern that contributes to high BCAA plasma concentrations may modulate chronic diseases risk.
Collapse
Affiliation(s)
- Benedikt Merz
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| | - Lara Frommherz
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| | - Manuela J Rist
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| | - Sabine E Kulling
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| | - Achim Bub
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| | - Bernhard Watzl
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| |
Collapse
|
154
|
Abu Bakar MH, Sarmidi MR. Association of cultured myotubes and fasting plasma metabolite profiles with mitochondrial dysfunction in type 2 diabetes subjects. MOLECULAR BIOSYSTEMS 2018; 13:1838-1853. [PMID: 28726959 DOI: 10.1039/c7mb00333a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Accumulating evidence implicates mitochondrial dysfunction-induced insulin resistance in skeletal muscle as the root cause for the greatest hallmarks of type 2 diabetes (T2D). However, the identification of specific metabolite-based markers linked to mitochondrial dysfunction in T2D has not been adequately addressed. Therefore, we sought to identify the markers-based metabolomics for mitochondrial dysfunction associated with T2D. First, a cellular disease model was established using human myotubes treated with antimycin A, an oxidative phosphorylation inhibitor. Non-targeted metabolomic profiling of intracellular-defined metabolites on the cultured myotubes with mitochondrial dysfunction was then determined. Further, a targeted MS-based metabolic profiling of fasting blood plasma from normal (n = 32) and T2D (n = 37) subjects in a cross-sectional study was verified. Multinomial logical regression analyses for defining the top 5% of the metabolites within a 95% group were employed to determine the differentiating metabolites. The myotubes with mitochondrial dysfunction exhibited insulin resistance, oxidative stress and inflammation with impaired insulin signalling activities. Four metabolic pathways were found to be strongly associated with mitochondrial dysfunction in the cultured myotubes. Metabolites derived from these pathways were validated in an independent pilot investigation of the fasting blood plasma of healthy and diseased subjects. Targeted metabolic analysis of the fasting blood plasma with specific baseline adjustment revealed 245 significant features based on orthogonal partial least square discriminant analysis (PLS-DA) with a p-value < 0.05. Among these features, 20 significant metabolites comprised primarily of branched chain and aromatic amino acids, glutamine, aminobutyric acid, hydroxyisobutyric acid, pyroglutamic acid, acylcarnitine species (acetylcarnitine, propionylcarnitine, dodecenoylcarnitine, tetradecenoylcarnitine hexadecadienoylcarnitine and oleylcarnitine), free fatty acids (palmitate, arachidonate, stearate and linoleate) and sphingomyelin (d18:2/16:0) were identified as predictive markers for mitochondrial dysfunction in T2D subjects. The current study illustrates how cellular metabolites provide potential signatures associated with the biochemical changes in the dysregulated body metabolism of diseased subjects. Our finding yields additional insights into the identification of robust biomarkers for T2D associated with mitochondrial dysfunction in cultured myotubes.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia.
| | | |
Collapse
|
155
|
Abstract
Disturbances in cardiac metabolism underlie most cardiovascular diseases. Metabolomics, one of the newer omics technologies, has emerged as a powerful tool for defining changes in both global and cardiac-specific metabolism that occur across a spectrum of cardiovascular disease states. Findings from metabolomics studies have contributed to better understanding of the metabolic changes that occur in heart failure and ischemic heart disease and have identified new cardiovascular disease biomarkers. As technologies advance, the metabolomics field continues to evolve rapidly. In this review, we will discuss the current state of metabolomics technologies, including consideration of various metabolomics platforms and elements of study design; the emerging utility of stable isotopes for metabolic flux studies; and the use of metabolomics to better understand specific cardiovascular diseases, with an emphasis on recent advances in the field.
Collapse
Affiliation(s)
- Robert W McGarrah
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
- Division of Cardiology (R.W.M., S.H.S.)
- Department of Medicine (R.W.M., G.F.Z., S.H.S., C.B.N.)
| | - Scott B Crown
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
| | - Guo-Fang Zhang
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
- Division of Endocrinology (G.F.Z., C.B.N.)
- Department of Medicine (R.W.M., G.F.Z., S.H.S., C.B.N.)
| | - Svati H Shah
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
- Division of Cardiology (R.W.M., S.H.S.)
- Department of Medicine (R.W.M., G.F.Z., S.H.S., C.B.N.)
| | - Christopher B Newgard
- From the Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute (R.W.M., S.B.C., G.F.Z., S.H.S., C.B.N.)
- Division of Endocrinology (G.F.Z., C.B.N.)
- Department of Medicine (R.W.M., G.F.Z., S.H.S., C.B.N.)
- Departments of Pharmacology and Cancer Biology (C.B.N.), Duke University Medical Center, Durham, NC
| |
Collapse
|
156
|
Gevi F, Fanelli G, Zolla L. Metabolic patterns in insulin-resistant male hypogonadism. Cell Death Dis 2018; 9:671. [PMID: 29867095 PMCID: PMC5986816 DOI: 10.1038/s41419-018-0587-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/28/2018] [Accepted: 04/12/2018] [Indexed: 01/21/2023]
Abstract
Male hypogonadism associated with insulin resistance (IR) very often leads to metabolic syndrome, at variance with hypogonadism in its first stadium of insulin sensitivity (IS). A plasma metabolomic investigation of these patients can provide useful information in comparison with the values of IS patients. To this aim plasma from insulin-resistant males with hypogonadism were analysed by using ultra high-performance liquid chromatography (UHPLC) and high-resolution mass spectrometry (HRMS). Thus, metabolites were compared to the controls through multivariate statistical analysis and grouped by metabolic pathways. Metabolite database searches and pathway analyses identified imbalances in 18-20 metabolic pathways. Glucose metabolism (e.g., glycolysis and the Krebs cycle) is fuelled by amino acids degradation, in particular of branched amino acids, in individuals with lean body mass. Gluconeogenesis is strongly activated. Some crucial pathways such as glycerol are skewed. Mitochondrial electron transport is affected with a reduction in ATP production. Beta-oxidation of short and medium chain fatty acids did not represent an energy source in hypogonadism, at variance with long and branched fatty acids, justifying the increase in fat mass. Carnosine and β-alanine are strongly reduced resulting in increased fatigue and mental confusion. A comparison of IR with IS male hypogonadism will contribute to a better understanding of how these two hormones work in synergy or antagonise each other in humans. It could also help to select patients who will respond to hormone treatment, and provide accurate biomarkers to measure the response to treatment eventually leading to better strategies in preventing systemic complications in patients not fit for hormone replacement therapy.
Collapse
Affiliation(s)
- Federica Gevi
- Department of Science and Technology for Agriculture, Forestry, Nature and Energy (DAFNE), University of Tuscia, Viterbo, Italy
| | - Giuseppina Fanelli
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Viterbo, Italy
| | - Lello Zolla
- Department of Science and Technology for Agriculture, Forestry, Nature and Energy (DAFNE), University of Tuscia, Viterbo, Italy.
| |
Collapse
|
157
|
Shi T, Yan X, Qiao L, Li B, Cheng L, Pan Y, Jing J, Cao N, Liu W. MiR-330-5p negatively regulates ovine preadipocyte differentiation by targeting branched-chain aminotransferase 2. Anim Sci J 2018; 89:858-867. [DOI: 10.1111/asj.12995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 01/10/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Tao Shi
- College of Animal Science and Veterinary Medicine; Shanxi Agricultural University; Taigu China
| | - Xiaoru Yan
- College of Animal Science and Veterinary Medicine; Shanxi Agricultural University; Taigu China
| | - Liying Qiao
- College of Animal Science and Veterinary Medicine; Shanxi Agricultural University; Taigu China
| | - Baojun Li
- College of Animal Science and Veterinary Medicine; Shanxi Agricultural University; Taigu China
| | - Lifen Cheng
- Division of Animal and Poultry Breeding; Department of Agriculture of Shanxi Province; Taiyuan China
| | - Yangyang Pan
- College of Animal Science and Veterinary Medicine; Shanxi Agricultural University; Taigu China
| | - Jiongjie Jing
- College of Animal Science and Veterinary Medicine; Shanxi Agricultural University; Taigu China
| | - Ningxian Cao
- Division of Animal and Poultry Breeding; Department of Agriculture of Shanxi Province; Taiyuan China
| | - Wenzhong Liu
- College of Animal Science and Veterinary Medicine; Shanxi Agricultural University; Taigu China
| |
Collapse
|
158
|
Wolak-Dinsmore J, Gruppen EG, Shalaurova I, Matyus SP, Grant RP, Gegen R, Bakker SJL, Otvos JD, Connelly MA, Dullaart RPF. A novel NMR-based assay to measure circulating concentrations of branched-chain amino acids: Elevation in subjects with type 2 diabetes mellitus and association with carotid intima media thickness. Clin Biochem 2018; 54:92-99. [PMID: 29432757 DOI: 10.1016/j.clinbiochem.2018.02.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/22/2017] [Accepted: 02/03/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Plasma branched-chain amino acid (BCAA) levels, measured on nuclear magnetic resonance (NMR) metabolomics research platforms or by mass spectrometry, have been shown to be associated with type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). We developed a new test for quantification of BCAA on a clinical NMR analyzer and used this test to determine the clinical correlates of BCAA in 2 independent cohorts. DESIGN AND METHODS The performance of the NMR-based BCAA assay was evaluated. A method comparison study was performed with mass spectrometry (LC-MS/MS). Plasma BCAA were measured in the Insulin Resistance Atherosclerosis Study (IRAS, n = 1209; 376 T2DM subjects) and in a Groningen cohort (n = 123; 67 T2DM subjects). In addition, carotid intima media thickness (cIMT) was measured successfully in 119 subjects from the Groningen cohort. RESULTS NMR-based BCAA assay results were linear over a range of concentrations. Coefficients of variation for inter- and intra-assay precision ranged from 1.8-6.0, 1.7-5.4, 4.4-9.1, and 8.8-21.3%, for total BCAA, valine, leucine, and isoleucine, respectively. BCAA quantified from the same samples using NMR and LC-MS/MS were highly correlated (R2 = 0.97, 0.95 and 0.90 for valine, leucine and isoleucine). In both cohorts total and individual BCAA were elevated in T2DM (P = 0.01 to ≤0.001). Moreover, cIMT was associated with BCAA independent of age, sex, T2DM and metabolic syndrome (MetS) categorization or alternatively of individual MetS components. CONCLUSIONS BCAA levels, measured by NMR in the clinical laboratory, are elevated in T2DM and may be associated with cIMT, a proxy of subclinical atherosclerosis.
Collapse
Affiliation(s)
| | - Eke G Gruppen
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Irina Shalaurova
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, Burlington, NC, USA.
| | - Steven P Matyus
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, Burlington, NC, USA.
| | - Russell P Grant
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, Burlington, NC, USA.
| | - Ray Gegen
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, Burlington, NC, USA.
| | - Stephan J L Bakker
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - James D Otvos
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, Burlington, NC, USA.
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, Burlington, NC, USA.
| | - Robin P F Dullaart
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
159
|
Li QR, Wang ZM, Wewer Albrechtsen NJ, Wang DD, Su ZD, Gao XF, Wu QQ, Zhang HP, Zhu L, Li RX, Jacobsen S, Jørgensen NB, Dirksen C, Bojsen-Møller KN, Petersen JS, Madsbad S, Clausen TR, Diderichsen B, Chen LN, Holst JJ, Zeng R, Wu JR. Systems Signatures Reveal Unique Remission-path of Type 2 Diabetes Following Roux-en-Y Gastric Bypass Surgery. EBioMedicine 2018; 28:234-240. [PMID: 29422288 PMCID: PMC5835566 DOI: 10.1016/j.ebiom.2018.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/14/2022] Open
Abstract
Roux-en-Y Gastric bypass surgery (RYGB) is emerging as a powerful tool for treatment of obesity and may also cause remission of type 2 diabetes. However, the molecular mechanism of RYGB leading to diabetes remission independent of weight loss remains elusive. In this study, we profiled plasma metabolites and proteins of 10 normal glucose-tolerant obese (NO) and 9 diabetic obese (DO) patients before and 1-week, 3-months, 1-year after RYGB. 146 proteins and 128 metabolites from both NO and DO groups at all four stages were selected for further analysis. By analyzing a set of bi-molecular associations among the corresponding network of the subjects with our newly developed computational method, we defined the represented physiological states (called the edge-states that reflect the interactions among the bio-molecules), and the related molecular networks of NO and DO patients, respectively. The principal component analyses (PCA) revealed that the edge states of the post-RYGB NO subjects were significantly different from those of the post-RYGB DO patients. Particularly, the time-dependent changes of the molecular hub-networks differed between DO and NO groups after RYGB. In conclusion, by developing molecular network-based systems signatures, we for the first time reveal that RYGB generates a unique path for diabetes remission independent of weight loss.
Collapse
Affiliation(s)
- Qing-Run Li
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zi-Ming Wang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, China
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dan-Dan Wang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China; University of Chinese Academy of Sciences, China
| | - Zhi-Duan Su
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xian-Fu Gao
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qing-Qing Wu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hui-Ping Zhang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Li Zhu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Rong-Xia Li
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - SivHesse Jacobsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nils Bruun Jørgensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Dirksen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine N Bojsen-Møller
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Sten Madsbad
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Luo-Nan Chen
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China.
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Rong Zeng
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China.
| | - Jia-Rui Wu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China.
| |
Collapse
|
160
|
Gender-Associated Impact of Early Leucine Supplementation on Adult Predisposition to Obesity in Rats. Nutrients 2018; 10:nu10010076. [PMID: 29329236 PMCID: PMC5793304 DOI: 10.3390/nu10010076] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/15/2017] [Accepted: 01/06/2018] [Indexed: 12/14/2022] Open
Abstract
Early nutrition plays an important role in development and may constitute a relevant contributor to the onset of obesity in adulthood. The aim of this study was to evaluate the long-term impact of maternal leucine (Leu) supplementation during lactation on progeny in rats. A chow diet, supplemented with 2% Leu, was supplied during lactation (21 days) and, from weaning onwards, was replaced by a standard chow diet. Then, at adulthood (6 months of age), this was replaced with hypercaloric diets (either with high-fat (HF) or high-carbohydrate (HC) content), for two months, to induce obesity. Female offspring from Leu-supplemented dams showed higher increases in body weight and in body fat (62%) than their respective controls; whereas males were somehow protected (15% less fat than the corresponding controls). This profile in Leu-females was associated with altered neuronal architecture at the paraventricular nucleus (PVN), involving neuropeptide Y (NPY) fibers and impaired expression of neuropeptides and factors of the mTOR signaling pathway in the hypothalamus. Interestingly, leptin and adiponectin expression in adipose tissue at weaning and at the time before the onset of obesity could be defined as early biomarkers of metabolic disturbance, predisposing towards adult obesity under the appropriate environment.
Collapse
|
161
|
Abstract
Multiple diseases have a strong metabolic component, and metabolomics as a powerful phenotyping technology, in combination with orthogonal biological and clinical approaches, will undoubtedly play a determinant role in accelerating the understanding of mechanisms that underlie these complex diseases determined by a set of genetic, lifestyle, and environmental exposure factors. Here, we provide several examples of valuable findings from metabolomics-led studies in diabetes and obesity metabolism, neurodegenerative disorders, and cancer metabolism and offer a longer term vision toward personalized approach to medicine, from population-based studies to pharmacometabolomics.
Collapse
Affiliation(s)
- Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 19, 1005, Lausanne, Switzerland.
| | - Aurelien Thomas
- Unit of Toxicology, CURML, CHUV Lausanne University Hospital, HUG Geneva University Hospitals, Vulliette 04, 1000, Lausanne, Switzerland.
- Faculty of Biology and Medicine, University of Lausanne, Vulliette 04, 1000, Lausanne, Switzerland.
| |
Collapse
|
162
|
Fazelzadeh P, Hangelbroek RWJ, Joris PJ, Schalkwijk CG, Esser D, Afman L, Hankemeier T, Jacobs DM, Mihaleva VV, Kersten S, van Duynhoven J, Boekschoten MV. Weight loss moderately affects the mixed meal challenge response of the plasma metabolome and transcriptome of peripheral blood mononuclear cells in abdominally obese subjects. Metabolomics 2018; 14:46. [PMID: 29527144 PMCID: PMC5838115 DOI: 10.1007/s11306-018-1328-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The response to dietary challenges has been proposed as a more accurate measure of metabolic health than static measurements performed in the fasted state. This has prompted many groups to explore the potential of dietary challenge tests for assessment of diet and lifestyle induced shifts in metabolic phenotype. OBJECTIVES We examined whether the response to a mixed-meal challenge could provide a readout for a weight loss (WL)-induced phenotype shift in abdominally obese male subjects. The underlying assumption of a mixed meal challenge is that it triggers all aspects of phenotypic flexibility and provokes a more prolonged insulin response, possibly allowing for better differentiation between individuals. METHODS Abdominally obese men (n = 29, BMI = 30.3 ± 2.4 kg/m2) received a mixed-meal challenge prior to and after an 8-week WL or no-WL control intervention. Lean subjects (n = 15, BMI = 23.0 ± 2.0 kg/m2) only received the mixed meal challenge at baseline to have a benchmark for WL-induced phenotype shifts. RESULTS Levels of several plasma metabolites were significantly different between lean and abdominally obese at baseline as well as during postprandial metabolic responses. Genes related to oxidative phosphorylation in peripheral blood mononuclear cells (PBMCs) were expressed at higher levels in abdominally obese subjects as compared to lean subjects at fasting, which was partially reverted after WL. The impact of WL on the postprandial response was modest, both at the metabolic and gene expression level in PBMCs. CONCLUSION We conclude that mixed-meal challenges are not necessarily superior to measurements in the fasted state to assess metabolic health. Furthermore, the mechanisms accounting for the observed differences between lean and abdominally obese in the fasted state are different from those underlying the dissimilarity observed during the postprandial response.
Collapse
Affiliation(s)
- Parastoo Fazelzadeh
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
- Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Roland W J Hangelbroek
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
- Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Peter J Joris
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
- Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Diederik Esser
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Lydia Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | | | - Doris M Jacobs
- Netherlands Metabolomics Centre, Leiden, The Netherlands
- Unilever R&D, Vlaardingen, The Netherlands
| | - Velitchka V Mihaleva
- Netherlands Metabolomics Centre, Leiden, The Netherlands
- Unilever R&D, Vlaardingen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - John van Duynhoven
- Laboratory of Biophysics, Wageningen University, Stippeneng 4, 6708WE, Wageningen, The Netherlands.
- Top Institute Food and Nutrition, Wageningen, The Netherlands.
- Netherlands Metabolomics Centre, Leiden, The Netherlands.
- Unilever R&D, Vlaardingen, The Netherlands.
| | - Mark V Boekschoten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
- Top Institute Food and Nutrition, Wageningen, The Netherlands
| |
Collapse
|
163
|
Draper CF, Vassallo I, Di Cara A, Milone C, Comminetti O, Monnard I, Godin JP, Scherer M, Su M, Jia W, Guiraud SP, Praplan F, Guignard L, Ammon Zufferey C, Shevlyakova M, Emami N, Moco S, Beaumont M, Kaput J, Martin FP. A 48-Hour Vegan Diet Challenge in Healthy Women and Men Induces a BRANCH-Chain Amino Acid Related, Health Associated, Metabolic Signature. Mol Nutr Food Res 2017; 62. [PMID: 29087622 DOI: 10.1002/mnfr.201700703] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/24/2017] [Indexed: 12/14/2022]
Abstract
SCOPE Research is limited on diet challenges to improve health. A short-term, vegan protein diet regimen nutritionally balanced in macronutrient composition compared to an omnivorous diet is hypothesized to improve metabolic measurements of blood sugar regulation, blood lipids, and amino acid metabolism. METHODS AND RESULTS This randomized, cross-over, controlled vegan versus animal diet challenge is conducted on 21 (11 female,10 male) healthy participants. Fasting plasma is measured during a 3 d diet intervention for clinical biochemistry and metabonomics. Intervention diet plans meet individual caloric needs. Meals are provided and supervised. Diet compliance is monitored. CONCLUSIONS The vegan diet lowers triglycerides, insulin and homeostatic model assessment (HOMA-IR), bile acids, elevated magnesium levels, and changed branched-chain amino acids (BCAAs) metabolism (p < 0.05), potentiating insulin and blood sugar control after 48 h. Cholesterol control improves significantly in the vegan versus omnivorous diets. Plasma amino acid and magnesium concentrations positively correlate with dietary amino acids. Polyunsaturated fatty acids and dietary fiber inversely correlate with insulin, HOMA-IR, and triglycerides. Nutritional biochemistries, BCAAs, insulin, and HOMA-IR are impacted by sexual dimorphism. A health-promoting, BCAA-associated metabolic signature is produced from a short-term, healthy, controlled, vegan diet challenge when compared with a healthy, controlled, omnivorous diet.
Collapse
Affiliation(s)
- Colleen Fogarty Draper
- Nestle Institute of Health Sciences (NIHS), Lausanne, Switzerland.,Leiden University, Leiden, The Netherlands
| | | | | | - Cristiana Milone
- Independent Clinical Dietitian.,Canton Hospital of Luzern, Luzern, Switzerland
| | | | | | | | | | - MingMing Su
- University of Hawaii Cancer Center (UHCC), HI, USA
| | - Wei Jia
- University of Hawaii Cancer Center (UHCC), HI, USA
| | - Seu-Ping Guiraud
- Nestle Institute of Health Sciences (NIHS), Lausanne, Switzerland
| | | | | | | | | | - Nashmil Emami
- Clinical Development Unit, NRC, Lausanne, Switzerland
| | - Sofia Moco
- Nestle Institute of Health Sciences (NIHS), Lausanne, Switzerland
| | | | - Jim Kaput
- Nestle Institute of Health Sciences (NIHS), Lausanne, Switzerland
| | | |
Collapse
|
164
|
Cummings NE, Williams EM, Kasza I, Konon EN, Schaid MD, Schmidt BA, Poudel C, Sherman DS, Yu D, Arriola Apelo SI, Cottrell SE, Geiger G, Barnes ME, Wisinski JA, Fenske RJ, Matkowskyj KA, Kimple ME, Alexander CM, Merrins MJ, Lamming DW. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J Physiol 2017; 596:623-645. [PMID: 29266268 DOI: 10.1113/jp275075] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/20/2017] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS We recently found that feeding healthy mice a diet with reduced levels of branched-chain amino acids (BCAAs), which are associated with insulin resistance in both humans and rodents, modestly improves glucose tolerance and slows fat mass gain. In the present study, we show that a reduced BCAA diet promotes rapid fat mass loss without calorie restriction in obese mice. Selective reduction of dietary BCAAs also restores glucose tolerance and insulin sensitivity to obese mice, even as they continue to consume a high-fat, high-sugar diet. A low BCAA diet transiently induces FGF21 (fibroblast growth factor 21) and increases energy expenditure. We suggest that dietary protein quality (i.e. the precise macronutrient composition of dietary protein) may impact the effectiveness of weight loss diets. ABSTRACT Obesity and diabetes are increasing problems around the world, and although even moderate weight loss can improve metabolic health, reduced calorie diets are notoriously difficult to sustain. Branched-chain amino acids (BCAAs; leucine, isoleucine and valine) are elevated in the blood of obese, insulin-resistant humans and rodents. We recently demonstrated that specifically reducing dietary levels of BCAAs has beneficial effects on the metabolic health of young, growing mice, improving glucose tolerance and modestly slowing fat mass gain. In the present study, we examine the hypothesis that reducing dietary BCAAs will promote weight loss, reduce adiposity, and improve blood glucose control in diet-induced obese mice with pre-existing metabolic syndrome. We find that specifically reducing dietary BCAAs rapidly reverses diet-induced obesity and improves glucoregulatory control in diet-induced obese mice. Most dramatically, mice eating an otherwise unhealthy high-calorie, high-sugar Western diet with reduced levels of BCAAs lost weight and fat mass rapidly until regaining a normal weight. Importantly, this normalization of weight was mediated not by caloric restriction or increased activity, but by increased energy expenditure, and was accompanied by a transient induction of the energy balance regulating hormone FGF21 (fibroblast growth factor 21). Consumption of a Western diet reduced in BCAAs was also accompanied by a dramatic improvement in glucose tolerance and insulin resistance. Our results link dietary BCAAs with the regulation of metabolic health and energy balance in obese animals, and suggest that specifically reducing dietary BCAAs may represent a highly translatable option for the treatment of obesity and insulin resistance.
Collapse
Affiliation(s)
- Nicole E Cummings
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth M Williams
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth N Konon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael D Schaid
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian A Schmidt
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chetan Poudel
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Dawn S Sherman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Deyang Yu
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Sebastian I Arriola Apelo
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Sara E Cottrell
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Rural and Urban Scholars in Community Health Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gabriella Geiger
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Macy E Barnes
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Jaclyn A Wisinski
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rachel J Fenske
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Kristina A Matkowskyj
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle E Kimple
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Caroline M Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew J Merrins
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
165
|
Association of plasma free amino acids with hyperuricemia in relation to diabetes mellitus, dyslipidemia, hypertension and metabolic syndrome. Sci Rep 2017; 7:17616. [PMID: 29247200 PMCID: PMC5732272 DOI: 10.1038/s41598-017-17710-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/29/2017] [Indexed: 12/22/2022] Open
Abstract
Previous studies demonstrated independent contributions of plasma free amino acids (PFAAs) and high uric acid (UA) concentrations to increased risks of lifestyle-related diseases (LSRDs), but the important associations between these factors and LSRDs remain unknown. We quantified PFAAs and UA amongst Japanese subjects without LSRDs (no-LSRD, n = 2805), and with diabetes mellitus (DM, n = 415), dyslipidemia (n = 3207), hypertension (n = 2736) and metabolic syndrome (MetS, n = 717). The concentrations of most amino acids differed significantly between the subjects with and without hyperuricemia (HU) and also between the no-LSRD and LSRD groups (p < 0.05 to 0.001). After adjustment, the logistic regression analyses revealed that lysine in DM, alanine, proline and tyrosine in dyslipidemia, histidine, lysine and ornithine in hypertension, and lysine and tyrosine in MetS demonstrated significant positive associations with HU among the patients with LSRDs only (p < 0.05 to 0.005). By contrast, arginine, asparagine and threonine showed significant inverse associations with HU in the no-LSRD group only (p < 0.05 to 0.01). For the first time, we provide evidence for distinct patterns of association between PFAAs and HU in LSRDs, and postulate the possibility of interplay between PFAAs and UA in their pathophysiology.
Collapse
|
166
|
Gar C, Rottenkolber M, Prehn C, Adamski J, Seissler J, Lechner A. Serum and plasma amino acids as markers of prediabetes, insulin resistance, and incident diabetes. Crit Rev Clin Lab Sci 2017; 55:21-32. [PMID: 29239245 DOI: 10.1080/10408363.2017.1414143] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Presently, routine screening misses many cases of prediabetes and early type 2 diabetes (T2D). Therefore, better biomarkers are needed for a simple and early detection of abnormalities of glucose metabolism and prediction of future T2D. Possible candidates for this include plasma or serum amino acids because glucose and amino acid metabolism are closely connected. This review presents the available evidence of this connectivity and discusses its clinical implications. First, we examine the underlying physiological, pre-analytical, and analytical issues. Then, we summarize results of human studies that evaluate amino acid levels as markers for insulin resistance, prediabetes, and future incident T2D. Finally, we illustrate the interconnection of amino acid levels and metabolic syndrome with our own data from a deeply phenotyped human cohort. We also discuss how amino acids may contribute to the pathophysiology of T2D. We conclude that elevated branched-chain amino acids and reduced glycine are currently the most robust and consistent amino acid markers for prediabetes, insulin resistance, and future T2D. Yet, we are cautious regarding the clinical potential even of these parameters because their discriminatory power is insufficient and their levels depend not only on glycemia, but also on other components of the metabolic syndrome. The identification of more precise intermediates of amino acid metabolism or combinations with other biomarkers will, therefore, be necessary to obtain in order to develop laboratory tests that can improve T2D screening.
Collapse
Affiliation(s)
- C Gar
- a Diabetes Research Group , Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich , Germany.,b Clinical Cooperation Group Type 2 Diabetes , Helmholtz Zentrum München , Neuherberg , Germany.,c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany
| | - M Rottenkolber
- a Diabetes Research Group , Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich , Germany.,b Clinical Cooperation Group Type 2 Diabetes , Helmholtz Zentrum München , Neuherberg , Germany.,c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany
| | - C Prehn
- d Institute of Experimental Genetics, Genome Analysis Center , Helmholtz Zentrum München, German Research Center for Environmental Health , Neuherberg , Germany
| | - J Adamski
- c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany.,d Institute of Experimental Genetics, Genome Analysis Center , Helmholtz Zentrum München, German Research Center for Environmental Health , Neuherberg , Germany.,e Lehrstuhl fu¨r Experimentelle Genetik , Technische Universität München , Freising , Germany
| | - J Seissler
- a Diabetes Research Group , Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich , Germany.,b Clinical Cooperation Group Type 2 Diabetes , Helmholtz Zentrum München , Neuherberg , Germany.,c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany
| | - A Lechner
- a Diabetes Research Group , Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich , Germany.,b Clinical Cooperation Group Type 2 Diabetes , Helmholtz Zentrum München , Neuherberg , Germany.,c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany
| |
Collapse
|
167
|
Xiang L, Wei J, Tian XY, Wang B, Chan W, Li S, Tang Z, Zhang H, Cheang WS, Zhao Q, Zhao H, Yang Z, Hong Y, Huang Y, Cai Z. Comprehensive Analysis of Acylcarnitine Species in db/db Mouse Using a Novel Method of High-Resolution Parallel Reaction Monitoring Reveals Widespread Metabolic Dysfunction Induced by Diabetes. Anal Chem 2017; 89:10368-10375. [PMID: 28859482 DOI: 10.1021/acs.analchem.7b02283] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Acylcarnitines are exerting a variety of biological functions depending on the differences in lengths, saturation levels, and conjugation groups, which to a great extent contribute to the challenges of acylcarnitines quantifications due to various kinds of isomers. Here, we describe a novel method by using high-resolution parallel reaction monitoring (PRM) liquid chromatography-tandem mass spectrometry (LC-MS/MS). Both reversed-phase and normal-phase column were used in order to get accurate, reliable, widespread quantification of acylcarnitines, and without tedious sample preparation procedure. The method provided the most comprehensive acylcarnitine profile with high-resolution MS and MS/MS confirmation to date. A total of 117 acylcarnitines were detected from plasma and urine samples. The application of targeted profiling of acylcarnitines in db/m+ control and db/db diabetic mice indicated incomplete amino acid and fatty acid oxidation on diabetic mice. Interestingly, the reduction of medium odd-numbered chain acylcarnitines in urine samples was first observed between db/m+ and db/db mice. The high-resolution PRM method makes it possible to monitor the widespread metabolic changes of the acylcarnitines in response to stimuli. Besides, the accurate MS and MS/MS spectra data of the 117 acylcarnitines could be used as mass spectrometric resources for the identification of acylcarnitines.
Collapse
Affiliation(s)
- Li Xiang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Juntong Wei
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Xiao Yu Tian
- School of Biomedical Sciences, Chinese University of Hong Kong , Hong Kong 999077, P. R. China
| | - Bei Wang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Wan Chan
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Shangfu Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Zhi Tang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Hongsong Zhang
- School of Biomedical Sciences, Chinese University of Hong Kong , Hong Kong 999077, P. R. China
| | - Wai San Cheang
- School of Biomedical Sciences, Chinese University of Hong Kong , Hong Kong 999077, P. R. China
| | - Qian Zhao
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Hongzhi Zhao
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Zhiyi Yang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Yanjun Hong
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| | - Yu Huang
- School of Biomedical Sciences, Chinese University of Hong Kong , Hong Kong 999077, P. R. China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University , Hong Kong 999077, P. R. China
| |
Collapse
|
168
|
Untargeted metabolomics identifies a plasma sphingolipid-related signature associated with lifestyle intervention in prepubertal children with obesity. Int J Obes (Lond) 2017; 42:72-78. [PMID: 28947825 DOI: 10.1038/ijo.2017.201] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/25/2017] [Accepted: 08/02/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Childhood obesity is a strong risk factor for adult obesity and metabolic diseases, including type 2 diabetes and cardiovascular disease. Early lifestyle intervention in children with obesity reduces future disease risk. The objective of this study is to identify metabolic signatures associated with lifestyle intervention in prepubertal children with obesity. METHODS Thirty-five prepubertal children (7-10 years) with obesity (body mass index (BMI)>2 standard deviations) were enrolled in the study and participated in a 6-month-long lifestyle intervention program. Physiological and biochemical data and blood samples were collected both at baseline and after the intervention. A liquid chromatography-mass spectrometry (LC-MS)-based metabolomics approach was applied to obtain a comprehensive profiling of plasma samples, identifying 2581 distinct metabolite. Principal component analysis (PCA) was performed to consolidate all features into 8 principal components. Associations between metabolites and physiological and biochemical variables were investigated. RESULTS The intervention program significantly decreased mean (95% CI) BMI standard deviation score from 3.56 (3.29-3.84) to 3.11 (2.88-3.34) (P<0.001). PCA identified one component (PC1) significantly altered by the intervention (Bonferroni adjusted P=0.008). A sphingolipid metabolism-related signature was identified as the major contributor to PC1. Sphingolipid metabolites were decreased by the intervention, and included multiple sphingomyelin, ceramide, glycosylsphingosine and sulfatide species. Changes in several sphingolipid metabolites were associated with intervention-induced improvements in HbA1c levels. CONCLUSIONS Decreased circulating sphingolipid-related metabolites were associated with lifestyle intervention in prepubertal children with obesity, and correlated to improvements in HbA1c.
Collapse
|
169
|
A Novel Dual Eigen-Analysis of Mouse Multi-Tissues' Expression Profiles Unveils New Perspectives into Type 2 Diabetes. Sci Rep 2017; 7:5044. [PMID: 28698587 PMCID: PMC5506042 DOI: 10.1038/s41598-017-05405-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/26/2017] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex and polygenic disease yet in need of a complete picture of its development mechanisms. To better understand the mechanisms, we examined gene expression profiles of multi-tissues from outbred mice fed with a high-fat diet (HFD) or regular chow at weeks 1, 9, and 18. To analyze such complex data, we proposed a novel dual eigen-analysis, in which the sample- and gene-eigenvectors correspond respectively to the macro- and micro-biology information. The dual eigen-analysis identified the HFD eigenvectors as well as the endogenous eigenvectors for each tissue. The results imply that HFD influences the hepatic function or the pancreatic development as an exogenous factor, while in adipose HFD's impact roughly coincides with the endogenous eigenvector driven by aging. The enrichment analysis of the eigenvectors revealed diverse HFD impact on the three tissues over time. The diversity includes: inflammation, degradation of branched chain amino acids (BCAA), and regulation of peroxisome proliferator activated receptor gamma (PPARγ). We reported that in the pancreas remarkable up-regulation of angiogenesis as downstream of the HIF signaling pathway precedes hyperinsulinemia. The dual eigen-analysis and discoveries provide new evaluations/guidance in T2D prevention and therapy, and will also promote new thinking in biology and medicine.
Collapse
|
170
|
Metabolomics in nutrition research-a powerful window into nutritional metabolism. Essays Biochem 2017; 60:451-458. [PMID: 27980095 DOI: 10.1042/ebc20160029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 10/27/2016] [Accepted: 10/29/2016] [Indexed: 01/22/2023]
Abstract
Metabolomics is the study of small molecules present in biological samples. In recent years it has become evident that such small molecules, called metabolites, play a key role in the development of disease states. Furthermore, metabolomic applications can reveal information about alterations in certain metabolic pathways under different conditions. Data acquisition in metabolomics is usually performed using nuclear magnetic resonance (NMR)-based approaches or mass spectrometry (MS)-based approaches with a more recent trend including the application of multiple platforms in order to maximise the coverage in terms of metabolites measured. The application of metabolomics is rapidly increasing and the present review will highlight applications in nutrition research.
Collapse
|
171
|
Liu R, Li H, Fan W, Jin Q, Chao T, Wu Y, Huang J, Hao L, Yang X. Leucine Supplementation Differently Modulates Branched-Chain Amino Acid Catabolism, Mitochondrial Function and Metabolic Profiles at the Different Stage of Insulin Resistance in Rats on High-Fat Diet. Nutrients 2017; 9:nu9060565. [PMID: 28574481 PMCID: PMC5490544 DOI: 10.3390/nu9060565] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 01/09/2023] Open
Abstract
The available findings concerning the association between branched-chain amino acids (BCAAs)—particularly leucine—and insulin resistance are conflicting. BCAAs have been proposed to elicit different or even opposite effects, depending on the prevalence of catabolic and anabolic states. We tested the hypothesis that leucine supplementation may exert different effects at different stages of insulin resistance, to provide mechanistic insights into the role of leucine in the progression of insulin resistance. Male Sprague-Dawley rats were fed a normal chow diet, high-fat diet (HFD), HFD supplemented with 1.5% leucine, or HFD with a 20% calorie restriction for 24 or 32 weeks. Leucine supplementation led to abnormal catabolism of BCAA and the incompletely oxidized lipid species that contributed to mitochondrial dysfunction in skeletal muscle in HFD-fed rats in the early stage of insulin resistance (24 weeks). However, leucine supplementation induced no remarkable alternations in BCAA catabolism, but did enhance mitochondrial biogenesis with a concomitant improvement in lipid oxidation and mitochondrial function during the hyperglycaemia stage (32 weeks). These findings suggest that leucine trigger different effects on metabolic signatures at different stages of insulin resistance, and the overall metabolic status of the organisms should be carefully considered to potentiate the benefits of leucine.
Collapse
Affiliation(s)
- Rui Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Hui Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Wenjuan Fan
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Qiu Jin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Tingting Chao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Yuanjue Wu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Junmei Huang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Liping Hao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Xuefeng Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
172
|
Geidenstam N, Al-Majdoub M, Ekman M, Spégel P, Ridderstråle M. Metabolite profiling of obese individuals before and after a one year weight loss program. Int J Obes (Lond) 2017; 41:1369-1378. [PMID: 28529327 DOI: 10.1038/ijo.2017.124] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 04/19/2017] [Accepted: 05/03/2017] [Indexed: 11/09/2022]
Abstract
OBJECTIVE We and others have previously characterized changes in circulating metabolite levels following diet-induced weight loss. Our aim was to investigate whether baseline metabolite levels and weight-loss-induced changes in these are predictive of or associated with changes in body mass index (BMI) and metabolic risk traits. METHODS Serum metabolites were analyzed with gas and liquid chromatography/mass spectrometry in 91 obese individuals at baseline and after participating in a 1 year non-surgical weight loss program.ResultsA total of 137 metabolites were identified and semi-quantified at baseline (BMI 42.7±5.8, mean±s.d.) and at follow-up (BMI 36.3±6.6). Weight-loss-induced modification was observed for levels of 57 metabolites in individuals with ⩾10% weight loss. Lower baseline levels of xylitol was predictive of a greater decrease in BMI (β=0.06, P<0.01) and ⩾10% weight loss (odds ratio (OR)=0.2, confidence interval (CI)=0.07-0.7, P=0.01). Decreases in levels of isoleucine, leucine, valine and tyrosine were associated with decrease in BMI (β>0.1, P<0.05) and ⩾10% weight loss (isoleucine: OR=0.08, CI=0.01-0.3, leucine: OR=0.1, CI=0.01-0.6, valine: OR=0.1, CI=0.02-0.5, tyrosine: OR=0.1, CI=0.03-0.6, P<0.02). CONCLUSIONS Diet-induced weight loss leads to mainly reduced levels of metabolites that are elevated in obese insulin resistant individuals. We identified multiple new associations with metabolic risk factors and validated several previous findings related to weight loss-mediated metabolite changes. Levels of specific metabolites, such as xylitol, may be predictive of the response to non-surgical weight loss already at baseline.
Collapse
Affiliation(s)
- N Geidenstam
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - M Al-Majdoub
- Department of Clinical Sciences Malmö, Unit of Molecular Metabolism, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - M Ekman
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - P Spégel
- Department of Clinical Sciences Malmö, Unit of Molecular Metabolism, Lund University Diabetes Center, Lund University, Malmö, Sweden.,Department of Chemistry, Centre for Analysis and Synthesis, Lund University, Lund, Sweden
| | - M Ridderstråle
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden.,Steno Diabetes Center A/S, Gentofte, Denmark.,Novo Nordisk A/S, Søborg, Denmark
| |
Collapse
|
173
|
Barceló A, Morell-Garcia D, Salord N, Esquinas C, Pérez G, Pérez A, Monasterio C, Gasa M, Fortuna AM, Montserrat JM, Mayos M. A randomized controlled trial: branched-chain amino acid levels and glucose metabolism in patients with obesity and sleep apnea. J Sleep Res 2017; 26:773-781. [PMID: 28513068 DOI: 10.1111/jsr.12551] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/28/2017] [Indexed: 12/11/2022]
Abstract
There is evidence that changes in branched-chain amino acid (BCAA) levels may correlate with the efficacy of therapeutic interventions for affecting improvement in metabolic control. The objective of this study was to evaluate whether serum concentrations of BCAAs (leucine, isoleucine, valine) could mediate in insulin sensitivity and glucose tolerance after continuous positive airway pressure (CPAP) treatment in patients with obstructive sleep apnea (OSA). A prospective randomized controlled trial of OSA patients with morbid obesity was conducted. Eighty patients were randomized into two groups: 38 received conservative treatment and 42 received CPAP treatment for 12 weeks. Plasma levels of BCAA, glucose tolerance and insulin resistance were evaluated at baseline and after treatment. After treatment, significant decreases of leucine levels were observed in both groups when compared with baseline levels (P < 0.005). With respect to patients with normal glucose tolerance (NGT), patients with impaired glucose tolerance (IGT) had higher baseline levels of isoleucine (78 ± 16 versus 70 ± 13 μmol L-1 , P = 0.014) and valine (286 ± 36 versus 268 ± 41 μmol L-1 , P = 0.049), respectively. Changes in levels of leucine and isoleucine after treatment were related negatively to changes in fasting plasma glucose and glycosylated haemoglobin values only in the conservative group (P < 0.05). In summary, we found that the treatment with CPAP for 12 weeks caused similar changes in circulating BCAAs concentrations to conservative treatment and a differential metabolic response of CPAP and conservative treatment was observed between the relationship of BCAAs and glucose homeostasis. Additional studies are needed to determine the interplay between branched-chain amino acids and glucose metabolism in patients with sleep apnea.
Collapse
Affiliation(s)
- Antonia Barceló
- Servei Análisis Clíniques, Hospital Universitari Son Espases, Palma de Mallorca, Spain.,CIBER Enfermedades Respiratorias (CibeRes) (CB06/06), Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
| | - Daniel Morell-Garcia
- Servei Análisis Clíniques, Hospital Universitari Son Espases, Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
| | - Neus Salord
- CIBER Enfermedades Respiratorias (CibeRes) (CB06/06), Spain.,Sleep Unit. Department of Respiratory Medicine, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Respiratory Medicine Section, Hospitalet de Llobregat, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Esquinas
- Department of Respiratory Medicine, Hospital Universitari Vall d'Hebrón, Barcelona, Spain
| | - Gerardo Pérez
- Servei Análisis Clíniques, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Antonio Pérez
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), Spain
| | - Carmen Monasterio
- CIBER Enfermedades Respiratorias (CibeRes) (CB06/06), Spain.,Sleep Unit. Department of Respiratory Medicine, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Respiratory Medicine Section, Hospitalet de Llobregat, Spain
| | - Merce Gasa
- Sleep Unit. Department of Respiratory Medicine, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Respiratory Medicine Section, Hospitalet de Llobregat, Spain
| | - Ana Maria Fortuna
- CIBER Enfermedades Respiratorias (CibeRes) (CB06/06), Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Sleep Unit, Department of Respiratory Medicine, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Josep Maria Montserrat
- CIBER Enfermedades Respiratorias (CibeRes) (CB06/06), Spain.,Sleep Unit, Department of Respiratory Medicine, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Mercedes Mayos
- CIBER Enfermedades Respiratorias (CibeRes) (CB06/06), Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Sleep Unit, Department of Respiratory Medicine, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
174
|
Posod A, Müller S, Komazec IO, Dejaco D, Peglow UP, Griesmaier E, Scholl-Bürgi S, Karall D, Kiechl-Kohlendorfer U. Former very preterm infants show alterations in plasma amino acid profiles at a preschool age. Pediatr Res 2017; 81:787-794. [PMID: 28141791 DOI: 10.1038/pr.2017.24] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/30/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Amino acid analysis is a valuable tool for cardiovascular risk assessment. Preterm infants display plasma amino acid changes in the newborn period. Whether these changes persist is unknown to date. The aim of this study was to assess whether former very preterm infants (VPI) show alterations in amino acid patterns indicative of an unfavorable cardiovascular risk profile at a preschool age. METHODS From 5-7 y-old children born at term or <32 wk gestation (VPI) were included in the study. Plasma amino acid concentrations were determined after an overnight fast. RESULTS 29 former term infants and 79 former VPI were included in the study. Former VPI showed changes in various plasma amino acids including glutamine, arginine, citrulline, tryptophan, glutamate, ornithine, and taurine. Branched-chain amino acids were lower, alanine/lysine ratios significantly higher in the preterm population. CONCLUSION Former VPI show altered plasma amino acid profiles indicative of a dualistic cardiovascular risk profile (e.g., potentially beneficial elevations in citrulline, arginine, glutamine, and tryptophan, but also raised alanine/lysine ratios, low ornithine and taurine levels) at a preschool age. Whether this is associated with an adverse cardiovascular outcome has to be addressed by future studies. Long-term cardiometabolic follow-up of VPI might be warranted.
Collapse
Affiliation(s)
- Anna Posod
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Susanne Müller
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Irena Odri Komazec
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria.,Pediatrics III (Pediatric Cardiology), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniel Dejaco
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Ulrike Pupp Peglow
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Elke Griesmaier
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Sabine Scholl-Bürgi
- Pediatrics I (Inherited Metabolic Disorders), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Karall
- Pediatrics I (Inherited Metabolic Disorders), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Ursula Kiechl-Kohlendorfer
- Pediatrics II (Neonatology), Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
175
|
Deng H, Li Z, Liu G, Li X, Chen Y, Zhang Y, Sun Y, Fu J. Elevated serum interferon γ-inducible protein-10 in women with polycystic ovary syndrome. Gynecol Endocrinol 2017; 33:363-367. [PMID: 28051885 DOI: 10.1080/09513590.2016.1269740] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Interferon γ-induced protein 10 kDa (IP10/CXCL10) is a chemokine related to endocrine disorders; however, the serum concentrations of IP10 in women with polycystic ovary syndrome (PCOS) have not yet been reported. Therefore, we investigated whether IP10 is increased in PCOS patients and its potential clinical value in PCOS patients. METHODS For this research, the serum IP10, glucose, insulin, high sensitivity C-reactive protein (hs-CRP), follicle-stimulating hormone (FSH), luteinizing hormone (LH) and total testosterone (TT) concentrations were measured in 60 women with PCOS and healthy controls. RESULTS The median IP10 concentration was 45.60 pg/mL [interquartile range (IQR):29.75, 79.69], which was significantly higher than that of the body mass index (BMI)-matched controls (median: 36.46 pg/mL; IQR:28.98, 45.80). In the multivariate linear regression analysis, hs-CRP and the homeostasis model assessment of insulin resistance index (HOMA2-IR) were independent predictors of the IP10 values, while FSH was inversely associated with the IP10.No significant association was observed between the IP10 and BMI, glucose, LH and TT. CONCLUSIONS The serum IP10 concentrations increase in women with PCOS, moreover, IP10 appears to be correlated with the inflammatory and IR statuses of PCOS. IP10 may be a potential biomarker to estimate the disease activity of PCOS.
Collapse
Affiliation(s)
- Hongli Deng
- a Department of Clinical Laboratory , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Changsha , Hunan , China
| | - Zhibo Li
- a Department of Clinical Laboratory , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Changsha , Hunan , China
| | - Guang Liu
- a Department of Clinical Laboratory , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Changsha , Hunan , China
| | - Xianhua Li
- a Department of Clinical Laboratory , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Changsha , Hunan , China
| | - Yong Chen
- b Department of Clinical Laboratory , Affiliated Changsha Hospital of University of South China, The First Hospital of Changsha City , Changsha , Hunan , China
| | - Yong Zhang
- c Department of Gastrointestinal Surgery , Affiliated Liuyang Hospital of University of South China, People's Hospital of Liuyang City , Chang sha , Hunan , China
| | - Yifan Sun
- d Department of Clinical Laboratory , Third Affiliated Hospital of Guangxi University of Chinese Medicine , Liuzhou , Guangxi , China , and
| | - Jinjian Fu
- e Department of Clinical Laboratory , Liuzhou Maternity and Child Health Care Hospital , Liuzhou , Guangxi , China
| |
Collapse
|
176
|
Cheng S, Shah SH, Corwin EJ, Fiehn O, Fitzgerald RL, Gerszten RE, Illig T, Rhee EP, Srinivas PR, Wang TJ, Jain M. Potential Impact and Study Considerations of Metabolomics in Cardiovascular Health and Disease: A Scientific Statement From the American Heart Association. ACTA ACUST UNITED AC 2017; 10:HCG.0000000000000032. [PMID: 28360086 DOI: 10.1161/hcg.0000000000000032] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Through the measure of thousands of small-molecule metabolites in diverse biological systems, metabolomics now offers the potential for new insights into the factors that contribute to complex human diseases such as cardiovascular disease. Targeted metabolomics methods have already identified new molecular markers and metabolomic signatures of cardiovascular disease risk (including branched-chain amino acids, select unsaturated lipid species, and trimethylamine-N-oxide), thus in effect linking diverse exposures such as those from dietary intake and the microbiota with cardiometabolic traits. As technologies for metabolomics continue to evolve, the depth and breadth of small-molecule metabolite profiling in complex systems continue to advance rapidly, along with prospects for ongoing discovery. Current challenges facing the field of metabolomics include scaling throughput and technical capacity for metabolomics approaches, bioinformatic and chemoinformatic tools for handling large-scale metabolomics data, methods for elucidating the biochemical structure and function of novel metabolites, and strategies for determining the true clinical relevance of metabolites observed in association with cardiovascular disease outcomes. Progress made in addressing these challenges will allow metabolomics the potential to substantially affect diagnostics and therapeutics in cardiovascular medicine.
Collapse
|
177
|
Gene expression profile of subcutaneous adipose tissue in BMI-discordant monozygotic twin pairs unravels molecular and clinical changes associated with sub-types of obesity. Int J Obes (Lond) 2017; 41:1176-1184. [PMID: 28439093 DOI: 10.1038/ijo.2017.95] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 12/11/2022]
|
178
|
Heianza Y, Qi L. Gene-Diet Interaction and Precision Nutrition in Obesity. Int J Mol Sci 2017; 18:ijms18040787. [PMID: 28387720 PMCID: PMC5412371 DOI: 10.3390/ijms18040787] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/30/2017] [Accepted: 04/03/2017] [Indexed: 02/06/2023] Open
Abstract
The rapid rise of obesity during the past decades has coincided with a profound shift of our living environment, including unhealthy dietary patterns, a sedentary lifestyle, and physical inactivity. Genetic predisposition to obesity may have interacted with such an obesogenic environment in determining the obesity epidemic. Growing studies have found that changes in adiposity and metabolic response to low-calorie weight loss diets might be modified by genetic variants related to obesity, metabolic status and preference to nutrients. This review summarized data from recent studies of gene-diet interactions, and discussed integration of research of metabolomics and gut microbiome, as well as potential application of the findings in precision nutrition.
Collapse
Affiliation(s)
- Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA.
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
179
|
Central adiposity-induced plasma-free amino acid alterations are associated with increased insulin resistance in healthy Singaporean adults. Eur J Clin Nutr 2017; 71:1080-1087. [DOI: 10.1038/ejcn.2017.34] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/06/2017] [Accepted: 02/16/2017] [Indexed: 12/25/2022]
|
180
|
Plasma-free amino acid profiles are predictors of cancer and diabetes development. Nutr Diabetes 2017; 7:e249. [PMID: 28287627 PMCID: PMC5380892 DOI: 10.1038/nutd.2016.55] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/17/2016] [Accepted: 11/24/2016] [Indexed: 12/29/2022] Open
Abstract
Type 2 diabetes (T2D) and cancers are two major causes of morbidity and mortality worldwide. Nowadays, there is convincing evidence of positive associations between T2D and the incidence or prognosis of a wide spectrum of cancers, for example, breast, colon, liver and pancreas. Many observational studies suggest that certain medications used to treat hyperglycemia (or T2D) may affect cancer cells directly or indirectly. The potential mechanisms of the direct T2D cancer links have been hypothesized to be hyperinsulinemia, hyperglycemia and chronic inflammation; however, the metabolic pathways that lead to T2D and cancers still remain elusive. Plasma-free amino acid (PFAA) profiles have been highlighted in their associations with the risks of developing T2D and cancers in individuals with different ethnic groups and degree of obesity. The alterations of PFAAs might be predominately caused by the metabolic shift resulted from insulin resistance. The underlying mechanisms have not been fully elucidated, in particular whether the amino acids are contributing to these diseases development in a causal manner. This review addresses the molecular and clinical associations between PFAA alterations and both T2D and cancers, and interprets possible mechanisms involved. Revealing these interactions and mechanisms may improve our understanding of the complex pathogenesis of diabetes and cancers and improve their treatment strategies.
Collapse
|
181
|
Dudzik D, Zorawski M, Skotnicki M, Zarzycki W, García A, Angulo S, Lorenzo MP, Barbas C, Ramos MP. GC-MS based Gestational Diabetes Mellitus longitudinal study: Identification of 2-and 3-hydroxybutyrate as potential prognostic biomarkers. J Pharm Biomed Anal 2017; 144:90-98. [PMID: 28314466 DOI: 10.1016/j.jpba.2017.02.056] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/04/2017] [Accepted: 02/28/2017] [Indexed: 01/03/2023]
Abstract
Gestational Diabetes Mellitus (GDM) causes severe short- and long-term complications for the mother, fetus and neonate, including type 2-diabetes (T2DM) later in life. In this pilot study, GC-Q/MS analysis was applied for plasma metabolomics fingerprinting of 24 healthy and 24 women with GDM at different stages of gestation (second and third trimester) and postpartum (one and three months). Multivariate (unsupervised and supervised) statistical analysis was performed to investigate variance in the data, identify outliers and for unbiased assessment of data quality. Plasma fingerprints allowed for the discrimination of GDM pregnant women from controls both in the 2nd and 3rd trimesters of gestation. However, metabolic profiles tended to be similar after delivery. Follow up of these women revealed that 4 of them developed T2DM within 2 years postpartum. Multivariate PLS-DA models limited to women with GDM showed clear separation 3 months postpartum. In the 2nd trimester of gestation there was also a clear separation between GDM women that were normoglycemic after pregnancy and those with recognized postpartum T2DM. Metabolites that had the strongest discriminative power between these groups in the 2nd trimester of gestation were 2-hydroxybutyrate, 3-hydroxybutyrate, and stearic acid. We have described, that early GDM comprises metabotypes that are associated with the risk of future complications, including postpartum T2DM. In this pilot study, we provide evidence that 2-hydroxybutyrate and 3-hydroxybutyrate may be considered as future prognostic biomarkers to predict the onset of diabetic complications in women with gestational diabetes after delivery.
Collapse
Affiliation(s)
- Danuta Dudzik
- Center for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain.
| | - Marcin Zorawski
- Department of Clinical Medicine, Faculty of Health Science, Medical University of Bialystok, 37 Szpitalna Street, 15-254, Bialystok, Poland.
| | - Mariusz Skotnicki
- Clinical Department of Perinatology, Public Clinic Hospital, Medical University of Bialystok, 24a Sklodowskiej-Curie Street, 15-276, Bialystok, Poland.
| | - Wieslaw Zarzycki
- Clinical Department of Endocrinology, Diabetology and Internal Diseases, Public Clinic Hospital, Medical University of Bialystok, 24a Sklodowskiej-Curie Street, 15-276, Bialystok, Poland.
| | - Antonia García
- Center for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain.
| | - Santiago Angulo
- Center for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain.
| | - M Paz Lorenzo
- Center for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain.
| | - Coral Barbas
- Center for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain.
| | - M Pilar Ramos
- Biochemistry and Molecular Biology, Facultad de Farmacia, Universidad San Pablo CEU, Boadilla del Monte, 28668, Madrid, Spain.
| |
Collapse
|
182
|
Connelly MA, Wolak-Dinsmore J, Dullaart RPF. Branched Chain Amino Acids Are Associated with Insulin Resistance Independent of Leptin and Adiponectin in Subjects with Varying Degrees of Glucose Tolerance. Metab Syndr Relat Disord 2017; 15:183-186. [PMID: 28437198 DOI: 10.1089/met.2016.0145] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Branched chain amino acids (BCAA) may be involved in the pathogenesis of insulin resistance and are associated with type 2 diabetes mellitus (T2DM) development. Adipokines such as leptin and adiponectin influence insulin resistance and reflect adipocyte dysfunction. We examined the extent to which the association of BCAA with insulin resistance is attributable to altered leptin and adiponectin levels in individuals with varying degrees of glucose tolerance. METHODS BCAA were measured by nuclear magnetic resonance, whereas leptin and adiponectin were measured by immunoassay, in subjects with normal fasting glucose (n = 30), impaired fasting glucose (n = 25), and T2DM (n = 15). Insulin resistance was estimated by homeostasis model assessment (HOMAir). RESULTS BCAA were higher in men than in women (P < 0.001) and tended to be higher in T2DM subjects (P = 0.10) compared to subjects with normal or impaired fasting glucose. In univariate regression analysis, BCAA were correlated with HOMAir (r = 0.46; P < 0.001) and inversely with adiponectin (r = -0.53; P < 0.001) but not with leptin (r = -0.08; P > 0.05). Multivariable linear regression analysis, adjusting for age, sex, T2DM, and body mass index (BMI), demonstrated that BCAA were positively associated with HOMAir (β = 0.242, P = 0.023). When BCAA, leptin, and adiponectin were included together, the positive relationship of HOMAir with BCAA (β = 0.275, P = 0.012) remained significant. CONCLUSIONS Insulin resistance was associated with BCAA. This association remained after adjusting for age, sex, T2DM, BMI, as well as leptin and adiponectin. It is unlikely that the relationship of insulin resistance with BCAA is to a major extent attributable to effects of leptin and adiponectin.
Collapse
Affiliation(s)
- Margery A Connelly
- 1 LipoScience, Laboratory Corporation of America® Holdings , Raleigh, North Carolina
| | | | - Robin P F Dullaart
- 2 Department of Endocrinology, University Medical Center Groningen, University of Groningen , Groningen, the Netherlands
| |
Collapse
|
183
|
Piperine regulates UCP1 through the AMPK pathway by generating intracellular lactate production in muscle cells. Sci Rep 2017; 7:41066. [PMID: 28117414 PMCID: PMC5259784 DOI: 10.1038/srep41066] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/14/2016] [Indexed: 12/19/2022] Open
Abstract
This study characterizes the human metabolic response to piperine, a curcumin extract, and the details of its underlying molecular mechanism. Using 1H-NMR-based metabolome analysis, we showed the metabolic effect of piperine on skeletal muscle and found that piperine increased the level of intracellular lactate, an important metabolic intermediate that controls expression of several genes involved in mitochondrial activity. Piperine also induced the phosphorylation of AMP-activated protein kinase (AMPK) and its downstream target, acetyl-CoA carboxylase (ACC), while additionally stimulating glucose uptake in an AMPK dependent manner. Piperine also stimulates the p38 mitogen-activated protein kinase (p38 MAPK), an effect that was reversed by pretreatment with compound C, an AMPK inhibitor. Inhibition of p38 MAPK resulted in no piperine-induced glucose uptake. Increased level of lactate resulted in increased expression of mitochondrial uncoupling protein 1 (UCP1), which regulates energy expenditure, thermogenesis, and fat browning. Knock-down of AMPK blocked piperine-induced UCP1 up-regulation, demonstrating the required role of AMPK in this effect. Taken together, these results suggest that piperine leads to benign metabolic effects by activating the AMPK-p38 MAPK signaling pathway and UCP1 expression by activating intracellular lactate production in skeletal muscle.
Collapse
|
184
|
Abstract
Metabolomics based on direct mass spectrometry (MS) analysis, either by direct infusion or flow injection of crude sample extracts, shows a great potential for metabolic fingerprinting because of its high-throughput screening capability, wide metabolite coverage and reduced time of analysis. Considering that numerous metabolic pathways are significantly perturbed during the initiation and progression of diseases, these metabolomic tools can be used to get a deeper understanding about disease pathogenesis and discover potential biomarkers for early diagnosis. In this work, we describe the most common metabolomic platforms used in biomedical research, with special focus on strategies based on direct MS analysis. Then, a comprehensive review on the application of direct MS fingerprinting in clinical issues is provided.
Collapse
|
185
|
Abstract
Metabolomics, or the comprehensive profiling of small molecule metabolites in cells, tissues, or whole organisms, has undergone a rapid technological evolution in the past two decades. These advances have led to the application of metabolomics to defining predictive biomarkers for incident cardiometabolic diseases and, increasingly, as a blueprint for understanding those diseases' pathophysiologic mechanisms. Progress in this area and challenges for the future are reviewed here.
Collapse
Affiliation(s)
- Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC 27701, USA.
| |
Collapse
|
186
|
Geidenstam N, Magnusson M, Danielsson APH, Gerszten RE, Wang TJ, Reinius LE, Mulder H, Melander O, Ridderstråle M. Amino Acid Signatures to Evaluate the Beneficial Effects of Weight Loss. Int J Endocrinol 2017; 2017:6490473. [PMID: 28484491 PMCID: PMC5412138 DOI: 10.1155/2017/6490473] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/10/2017] [Accepted: 02/27/2017] [Indexed: 11/17/2022] Open
Abstract
Aims. We investigated the relationship between circulating amino acid levels and obesity; to what extent weight loss followed by weight maintenance can correct amino acid abnormalities; and whether amino acids are related to weight loss. Methods. Amino acids associated with waist circumference (WC) and BMI were studied in 804 participants from the Malmö Diet and Cancer Cardiovascular Cohort (MDC-CC). Changes in amino acid levels were analyzed after weight loss and weight maintenance in 12 obese subjects and evaluated in a replication cohort (n = 83). Results. Out of the eight identified BMI-associated amino acids from the MDC-CC, alanine, isoleucine, tyrosine, phenylalanine, and glutamate decreased after weight loss, while asparagine increased after weight maintenance. These changes were validated in the replication cohort. Scores that were constructed based on obesity-associated amino acids and known risk factors decreased in the ≥10% weight loss group with an associated change in BMI (R2 = 0.16-0.22, p < 0.002), whereas the scores increased in the <10% weight loss group (p < 0.0004). Conclusions. Weight loss followed by weight maintenance leads to differential changes in amino acid levels associated with obesity. Treatment modifiable scores based on epidemiological and interventional data may be used to evaluate the potential metabolic benefit of weight loss.
Collapse
Affiliation(s)
- Nina Geidenstam
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
- *Nina Geidenstam:
| | - Martin Magnusson
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Anders P. H. Danielsson
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Robert E. Gerszten
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas J. Wang
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lovisa E. Reinius
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hindrik Mulder
- Department of Clinical Sciences Malmö, Molecular Metabolism, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
- Center of Emergency Medicine, Skåne University Hospital, Malmö, Sweden
| | - Martin Ridderstråle
- Department of Clinical Sciences Malmö, Clinical Obesity, Lund University Diabetes Center, Lund University, Malmö, Sweden
- Steno Diabetes Center A/S, Gentofte, Denmark
| |
Collapse
|
187
|
Chronic Diseases and Lifestyle Biomarkers Identification by Metabolomics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 965:235-263. [DOI: 10.1007/978-3-319-47656-8_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
188
|
Tang R, Ding C, Ma Y, Wang J, Zhang T, Wang X. Time-dependent responses of earthworms to soil contaminated with low levels of lead as detected using1H NMR metabolomics. RSC Adv 2017. [DOI: 10.1039/c7ra04393g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
1H NMR-based metabolomics was used to profile the time-dependent metabolic responses of earthworms (Eisenia fetida) that were exposed to low-Pb-contaminated-soil (L-Pb-CS) for 28 days using an indoor culture.
Collapse
Affiliation(s)
- Ronggui Tang
- Key Laboratory of Soil Environment and Pollution Remediation
- Institute of Soil Science
- Chinese Academy of Sciences
- Nanjing
- People's Republic of China
| | - Changfeng Ding
- Key Laboratory of Soil Environment and Pollution Remediation
- Institute of Soil Science
- Chinese Academy of Sciences
- Nanjing
- People's Republic of China
| | - Yibing Ma
- Institute of Agricultural Resources and Regional Planning
- Chinese Academy of Agricultural Sciences
- Beijing
- People's Republic of China
| | - Junsong Wang
- Center for Molecular Metabolism
- School of Environmental and Biological Engineering
- Nanjing University of Science and Technology
- Nanjing
- People's Republic of China
| | - Taolin Zhang
- Key Laboratory of Soil Environment and Pollution Remediation
- Institute of Soil Science
- Chinese Academy of Sciences
- Nanjing
- People's Republic of China
| | - Xingxiang Wang
- Key Laboratory of Soil Environment and Pollution Remediation
- Institute of Soil Science
- Chinese Academy of Sciences
- Nanjing
- People's Republic of China
| |
Collapse
|
189
|
Weisenberger MM, Bowser MT. In Vivo Monitoring of Amino Acid Biomarkers from Inguinal Adipose Tissue Using Online Microdialysis-Capillary Electrophoresis. Anal Chem 2016; 89:1009-1014. [DOI: 10.1021/acs.analchem.6b04516] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Megan M. Weisenberger
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Michael T. Bowser
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
190
|
Kim M, Lee SH, Lee JH. Global Metabolic Profiling of Plasma Shows that Three-Year Mild-Caloric Restriction Lessens an Age-Related Increase in Sphingomyelin and Reduces L-leucine and L-phenylalanine in Overweight and Obese Subjects. Aging Dis 2016; 7:721-733. [PMID: 28053823 PMCID: PMC5198864 DOI: 10.14336/ad.2016.0330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/30/2016] [Indexed: 12/21/2022] Open
Abstract
The effect of weight loss from long-term, mild-calorie diets (MCD) on plasma metabolites is unknown. This study was to examine whether MCD-induced weight reduction caused changes in the extended plasma metabolites. Overweight and obese subjects aged 40-59 years consumed a MCD (approximately 100 kcal/day deficit, n=47) or a weight-maintenance diet (control, n=47) in a randomized, controlled design with a three-year clinical intervention period and plasma samples were analyzed by using UPLC-LTQ-Orbitrap mass spectrometry. The three-year MCD intervention resulted in weight loss (-8.87%) and significant decreases in HOMA-IR and TG. The three-year follow-up of the MCD group showed reductions in the following 13 metabolites: L-leucine; L-phenylalanine; 9 lysoPCs; PC (18:0/20:4); and SM (d18:0/16:1). The three-year MCD group follow-up identified increases in palmitic amide, oleamide, and PC (18:2/18:2). Considering the age-related alterations in the identified metabolites, the MCD group showed a greater decrease in L-leucine, L-phenylalanine, and SM (d18:0/16:1) compared with those of the control group. Overall, the change (Δ) in BMI positively correlated with the ΔTG, ΔHOMA-IR, ΔL-leucine, and ΔSM (d18:0/16:1). The ΔHOMA-IR positively correlated with ΔTG, ΔL-leucine, ΔL-phenylalanine, and ΔSM (d18:0/16:1). The weight loss resulting from three-year mild-caloric restriction lessens the age-related increase in SM and reduces L-leucine and L-phenylalanine in overweight and obese subjects. These changes were coupled with improved insulin resistance (ClinicalTrials.gov: NCT02081898).
Collapse
Affiliation(s)
- Minjoo Kim
- 1National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea; 2Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Sang-Hyun Lee
- 3Department of Family Practice, National Health Insurance Corporation Ilsan Hospital, Goyang, 10444, Korea
| | - Jong Ho Lee
- 1National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea; 2Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea; 4Research Institute of Science for Aging, Yonsei University, Seoul, 03722, Korea
| |
Collapse
|
191
|
Tulipani S, Griffin J, Palau-Rodriguez M, Mora-Cubillos X, Bernal-Lopez RM, Tinahones FJ, Corkey BE, Andres-Lacueva C. Metabolomics-guided insights on bariatric surgery versus behavioral interventions for weight loss. Obesity (Silver Spring) 2016; 24:2451-2466. [PMID: 27891833 DOI: 10.1002/oby.21686] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 08/30/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To review the metabolomic studies carried out so far to identify metabolic markers associated with surgical and dietary treatments for weight loss in subjects with obesity. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. RESULTS Thirty-two studies successfully met the eligibility criteria. The metabolic adaptations shared by surgical and dietary interventions mirrored a state of starvation ketoacidosis (increase of circulating ketone bodies), an increase of acylcarnitines and fatty acid β-oxidation, a decrease of specific amino acids including branched-chain amino acids (BCAA) and (lyso)glycerophospholipids previously associated with obesity, and adipose tissue expansion. The metabolic footprint of bariatric procedures was specifically characterized by an increase of bile acid circulating pools and a decrease of ceramide levels, a greater perioperative decline in BCAA, and the rise of circulating serine and glycine, mirroring glycemic control and inflammation improvement. In one study, 3-hydroxybutyrate was particularly identified as an early metabolic marker of long-term prognosis after surgery and proposed to increase current prognostic modalities and contribute to personalized treatment. CONCLUSIONS Metabolomics helped in deciphering the metabolic response to weight loss treatments. Moving from association to causation is the next challenge to move to a further level of clinical application.
Collapse
Affiliation(s)
- Sara Tulipani
- Department of Nutrition, Food Sciences and Gastronomy, Biomarkers & Nutrimetabolomic Lab, XaRTA, INSA, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga, Malaga, Spain
| | - Jules Griffin
- MRC Human Nutrition Research, Elsie Widdowson Laboratory, Cambridge, UK
- Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Magali Palau-Rodriguez
- Department of Nutrition, Food Sciences and Gastronomy, Biomarkers & Nutrimetabolomic Lab, XaRTA, INSA, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Ximena Mora-Cubillos
- Department of Nutrition, Food Sciences and Gastronomy, Biomarkers & Nutrimetabolomic Lab, XaRTA, INSA, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Rosa M Bernal-Lopez
- Biomedical Research Institute (IBIMA), Service of Internal Medicine, Malaga Hospital Complex (Hospital Regional Universitario de Malaga), University of Malaga, Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Francisco J Tinahones
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga, Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Barbara E Corkey
- School of Medicine, Obesity Research Center, Boston University, Boston, Massachusetts, USA
| | - Cristina Andres-Lacueva
- Department of Nutrition, Food Sciences and Gastronomy, Biomarkers & Nutrimetabolomic Lab, XaRTA, INSA, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| |
Collapse
|
192
|
Fukai K, Harada S, Iida M, Kurihara A, Takeuchi A, Kuwabara K, Sugiyama D, Okamura T, Akiyama M, Nishiwaki Y, Oguma Y, Suzuki A, Suzuki C, Hirayama A, Sugimoto M, Soga T, Tomita M, Takebayashi T. Metabolic Profiling of Total Physical Activity and Sedentary Behavior in Community-Dwelling Men. PLoS One 2016; 11:e0164877. [PMID: 27741291 PMCID: PMC5065216 DOI: 10.1371/journal.pone.0164877] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 10/03/2016] [Indexed: 01/22/2023] Open
Abstract
Objective Physical activity is known to be preventive against various non-communicable diseases. We investigated the relationship between daily physical activity level and plasma metabolites using a targeted metabolomics approach in a population-based study. Methods A total of 1,193 participants (male, aged 35 to 74 years) with fasting blood samples were selected from the baseline survey of a cohort study. Information on daily total physical activity, classified into four levels by quartile of metabolic equivalent scores, and sedentary behavior, defined as hours of sitting per day, was collected through a self-administered questionnaire. Plasma metabolite concentrations were quantified by capillary electrophoresis mass spectrometry method. We performed linear regression analysis models with multivariable adjustment and corrected p-values for multiple testing in the original population (n = 808). The robustness of the results was confirmed by replication analysis in a separate population (n = 385) created by random allocation. Results Higher levels of total physical activity were associated with various metabolite concentrations, including lower concentrations of amino acids and their derivatives, and higher concentrations of pipecolate (FDR p <0.05 in original population). The findings persisted after adjustment for age, body mass index, smoking, alcohol intake, and energy intake. Isoleucine, leucine, valine, 4-methyl-2-oxoisopentanoate, 2-oxoisopentanoate, alanine, and proline concentrations were lower with a shorter sitting time. Conclusions Physical activity is related to various plasma metabolites, including known biomarkers for future insulin resistance or type 2 diabetes. These metabolites might potentially play a key role in the protective effects of higher physical activity and/or less sedentary behavior on non-communicable diseases.
Collapse
Affiliation(s)
- Kota Fukai
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Sei Harada
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Miho Iida
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Ayako Kurihara
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Ayano Takeuchi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyo Kuwabara
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Sugiyama
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Tomonori Okamura
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Miki Akiyama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Faculty of Environment and Information Studies, Keio University, Fujisawa, Japan
| | - Yuji Nishiwaki
- Division of Environmental and Occupational Health, Department of Social Medicine, Faculty of Medicine, Toho University, Tokyo, Japan
| | - Yuko Oguma
- Sports Medicine Research Center, Keio University, Yokohama, Japan
- Graduate School of Health Management, Keio University, Fujisawa, Japan
| | - Asako Suzuki
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Chizuru Suzuki
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Faculty of Environment and Information Studies, Keio University, Fujisawa, Japan
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Faculty of Environment and Information Studies, Keio University, Fujisawa, Japan
| | - Toru Takebayashi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Graduate School of Health Management, Keio University, Fujisawa, Japan
- * E-mail:
| |
Collapse
|
193
|
Lerin C, Goldfine AB, Boes T, Liu M, Kasif S, Dreyfuss JM, De Sousa-Coelho AL, Daher G, Manoli I, Sysol JR, Isganaitis E, Jessen N, Goodyear LJ, Beebe K, Gall W, Venditti CP, Patti ME. Defects in muscle branched-chain amino acid oxidation contribute to impaired lipid metabolism. Mol Metab 2016; 5:926-936. [PMID: 27689005 PMCID: PMC5034611 DOI: 10.1016/j.molmet.2016.08.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Plasma levels of branched-chain amino acids (BCAA) are consistently elevated in obesity and type 2 diabetes (T2D) and can also prospectively predict T2D. However, the role of BCAA in the pathogenesis of insulin resistance and T2D remains unclear. METHODS To identify pathways related to insulin resistance, we performed comprehensive gene expression and metabolomics analyses in skeletal muscle from 41 humans with normal glucose tolerance and 11 with T2D across a range of insulin sensitivity (SI, 0.49 to 14.28). We studied both cultured cells and mice heterozygous for the BCAA enzyme methylmalonyl-CoA mutase (Mut) and assessed the effects of altered BCAA flux on lipid and glucose homeostasis. RESULTS Our data demonstrate perturbed BCAA metabolism and fatty acid oxidation in muscle from insulin resistant humans. Experimental alterations in BCAA flux in cultured cells similarly modulate fatty acid oxidation. Mut heterozygosity in mice alters muscle lipid metabolism in vivo, resulting in increased muscle triglyceride accumulation, increased plasma glucose, hyperinsulinemia, and increased body weight after high-fat feeding. CONCLUSIONS Our data indicate that impaired muscle BCAA catabolism may contribute to the development of insulin resistance by perturbing both amino acid and fatty acid metabolism and suggest that targeting BCAA metabolism may hold promise for prevention or treatment of T2D.
Collapse
Affiliation(s)
- Carles Lerin
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA; Endocrinology Department, Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, Barcelona 08950, Spain.
| | - Allison B Goldfine
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA
| | - Tanner Boes
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Manway Liu
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Simon Kasif
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Jonathan M Dreyfuss
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Ana Luisa De Sousa-Coelho
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA
| | - Grace Daher
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | - Irini Manoli
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Justin R Sysol
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Elvira Isganaitis
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA
| | - Niels Jessen
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | | | | | - Walt Gall
- Metabolon, Inc., Durham, NC 27723, USA
| | - Charles P Venditti
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mary-Elizabeth Patti
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
194
|
Justice JN, Johnson LC, DeVan AE, Cruickshank-Quinn C, Reisdorph N, Bassett CJ, Evans TD, Brooks FA, Bryan NS, Chonchol MB, Giordano T, McQueen MB, Seals DR. Improved motor and cognitive performance with sodium nitrite supplementation is related to small metabolite signatures: a pilot trial in middle-aged and older adults. Aging (Albany NY) 2016; 7:1004-21. [PMID: 26626856 PMCID: PMC4694069 DOI: 10.18632/aging.100842] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Advancing age is associated with reductions in nitric oxide bioavailability and changes in metabolic activity, which are implicated in declines in motor and cognitive function. In preclinical models, sodium nitrite supplementation (SN) increases plasma nitrite and improves motor function, whereas other nitric oxide-boosting agents improve cognitive function. This pilot study was designed to translate these findings to middle-aged and older (MA/O) humans to provide proof-of-concept support for larger trials. SN (10 weeks, 80 or 160 mg/day capsules, TheraVasc, Inc.) acutely and chronically increased plasma nitrite and improved performance on measures of motor and cognitive outcomes (all p<0.05 or better) in healthy MA/O adults (62 ± 7 years). Untargeted metabolomics analysis revealed that SN significantly altered 33 (160 mg/day) to 45 (80 mg/day) different metabolites, 13 of which were related to changes in functional outcomes; baseline concentrations of 99 different metabolites predicted functional improvements with SN. This pilot study provides the first evidence that SN improves aspects of motor and cognitive function in healthy MA/O adults, and that these improvements are associated with, and predicted by, the plasma metabolome. Our findings provide the necessary support for larger clinical trials on this promising pharmacological strategy for preserving physiological function with aging.
Collapse
Affiliation(s)
- Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Lawrence C Johnson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Allison E DeVan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Charmion Cruickshank-Quinn
- Integrated Department of Immunology, University of Colorado Anschutz Medical Campus and National Jewish Hospital, Denver, CO 80045, USA
| | - Nichole Reisdorph
- Integrated Department of Immunology, University of Colorado Anschutz Medical Campus and National Jewish Hospital, Denver, CO 80045, USA
| | - Candace J Bassett
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Trent D Evans
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Forrest A Brooks
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | | | - Michel B Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO 80045, USA
| | | | - Matthew B McQueen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
195
|
Narath SH, Mautner SI, Svehlikova E, Schultes B, Pieber TR, Sinner FM, Gander E, Libiseller G, Schimek MG, Sourij H, Magnes C. An Untargeted Metabolomics Approach to Characterize Short-Term and Long-Term Metabolic Changes after Bariatric Surgery. PLoS One 2016; 11:e0161425. [PMID: 27584017 PMCID: PMC5008721 DOI: 10.1371/journal.pone.0161425] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 07/20/2016] [Indexed: 12/28/2022] Open
Abstract
Bariatric surgery is currently one of the most effective treatments for obesity and leads to significant weight reduction, improved cardiovascular risk factors and overall survival in treated patients. To date, most studies focused on short-term effects of bariatric surgery on the metabolic profile and found high variation in the individual responses to surgery. The aim of this study was to identify relevant metabolic changes not only shortly after bariatric surgery (Roux-en-Y gastric bypass) but also up to one year after the intervention by using untargeted metabolomics. 132 serum samples taken from 44 patients before surgery, after hospital discharge (1-3 weeks after surgery) and at a 1-year follow-up during a prospective study (NCT01271062) performed at two study centers (Austria and Switzerland). The samples included 24 patients with type 2 diabetes at baseline, thereof 9 with diabetes remission after one year. The samples were analyzed by using liquid chromatography coupled to high resolution mass spectrometry (LC-HRMS, HILIC-QExactive). Raw data was processed with XCMS and drift-corrected through quantile regression based on quality controls. 177 relevant metabolic features were selected through Random Forests and univariate testing and 36 metabolites were identified. Identified metabolites included trimethylamine-N-oxide, alanine, phenylalanine and indoxyl-sulfate which are known markers for cardiovascular risk. In addition we found a significant decrease in alanine after one year in the group of patients with diabetes remission relative to non-remission. Our analysis highlights the importance of assessing multiple points in time in subjects undergoing bariatric surgery to enable the identification of biomarkers for treatment response, cardiovascular benefit and diabetes remission. Key-findings include different trend pattern over time for various metabolites and demonstrated that short term changes should not necessarily be used to identify important long term effects of bariatric surgery.
Collapse
Affiliation(s)
- Sophie H. Narath
- JOANNEUM RESEARCH Forschungsgesellschaft mbH HEALTH Institute for Biomedicine and Health Sciences, Graz, Austria
| | - Selma I. Mautner
- JOANNEUM RESEARCH Forschungsgesellschaft mbH HEALTH Institute for Biomedicine and Health Sciences, Graz, Austria
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
- CBmed – Center of Biomarker Research in Medicine, Stiftingtalstrasse 5, 8010 Graz, Austria
| | - Eva Svehlikova
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
| | - Bernd Schultes
- eSwiss Medical & Surgical Center, St. Gallen, Switzerland
| | - Thomas R. Pieber
- JOANNEUM RESEARCH Forschungsgesellschaft mbH HEALTH Institute for Biomedicine and Health Sciences, Graz, Austria
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
- CBmed – Center of Biomarker Research in Medicine, Stiftingtalstrasse 5, 8010 Graz, Austria
| | - Frank M. Sinner
- JOANNEUM RESEARCH Forschungsgesellschaft mbH HEALTH Institute for Biomedicine and Health Sciences, Graz, Austria
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
| | - Edgar Gander
- JOANNEUM RESEARCH Forschungsgesellschaft mbH HEALTH Institute for Biomedicine and Health Sciences, Graz, Austria
| | - Gunnar Libiseller
- JOANNEUM RESEARCH Forschungsgesellschaft mbH HEALTH Institute for Biomedicine and Health Sciences, Graz, Austria
| | - Michael G. Schimek
- Institute for Medical Informatics, Statistics and Documentation Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
- CBmed – Center of Biomarker Research in Medicine, Stiftingtalstrasse 5, 8010 Graz, Austria
- * E-mail:
| | - Christoph Magnes
- JOANNEUM RESEARCH Forschungsgesellschaft mbH HEALTH Institute for Biomedicine and Health Sciences, Graz, Austria
| |
Collapse
|
196
|
Yang P, Hu W, Fu Z, Sun L, Zhou Y, Gong Y, Yang T, Zhou H. The positive association of branched-chain amino acids and metabolic dyslipidemia in Chinese Han population. Lipids Health Dis 2016; 15:120. [PMID: 27457614 PMCID: PMC4960685 DOI: 10.1186/s12944-016-0291-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/14/2016] [Indexed: 12/19/2022] Open
Abstract
Background It has been suggested that serum branched-chain amino acids (BCAAs) are associated with the incident, progression and prognostic of type 2 diabetes. However, the role of BCAAs in metabolic dyslipidemia (raised triglycerides (TG) and reduced high-density lipoprotein cholesterol (HDL-C)) remains poorly understood. This study aims to investigate 1) the association of serum BCAAs with total cholesterol (TC), TG, HDL-C and low-density lipoprotein cholesterol (LDL-C) and 2) the association between serum BCAAs levels and risk of metabolic dyslipidemia in a community population with different glucose homeostasis. Methods Demographics data and blood samples were collected from 2251 Chinese subjects from the Huaian Diabetes Protective Program (HADPP) study. After exclusion for cardiovascular disease (CVD), serious hepatic or nephritic diseases and others, 1320 subjects remained for analysis (789 subjects with hemoglobin A1c (HbA1c) > 5.7, 521 with HbA1c ≤ 5.7). Serum BCAAs level was measured by liquid chromatography-tandem mass spectrometry (LC MS/MS). The association of BCAAs with lipids or with the risk of metabolic dyslipidemia was analyzed. Results Elevated serum BCAAs (both total and individual BCAA) were positively associated with TG and inversely associated with HDL-C in the whole population. These correlations were still significant even after adjustment for confounding factors (r = 0.165, p < 0.001 for TG; and r = -0.126, p < 0.001 for HDL-C). For reduced HDL-C, we found higher odds risk (OR) of Valine (Val) in high HbA1c group than in the low one (OR = 1.055, p < 0.001 vs OR = 1.032, p = 0.059). Compared with that in the first quartile, the multivariable-adjusted OR (95 % CI) of the 4th quartile of serum total BCAAs level for reduced HDL-C was 3.689 (2.325, 5.854) in high HbA1c group and 2.329 (1.284, 4.227) in low group, for raised TG was 3.305 (2.120, 5.152) and 2.972 (1.706, 5.176), and for metabolic dyslipidemia was 3.703 (2.261, 6.065) and 3.702 (1.877, 7.304), respectively (all p < 0.01). Conclusion Elevated serum BCAAs level are positively associated with incident metabolic dyslipidemia. In addition, glucose homeostasis could play a certain role in BCAAs-related dyslipidemia.
Collapse
Affiliation(s)
- Panpan Yang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Wen Hu
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.,Department of Endocrinology and Metabolism, Huaian Hospital Affiliated to Xuzhou Medical College and Huaian Second People's Hospital, Huaian, 223002, China
| | - Zhenzhen Fu
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Luning Sun
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Ying Zhou
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Yingyun Gong
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Tao Yang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Hongwen Zhou
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
197
|
Zheng Y, Li Y, Qi Q, Hruby A, Manson JE, Willett WC, Wolpin BM, Hu FB, Qi L. Cumulative consumption of branched-chain amino acids and incidence of type 2 diabetes. Int J Epidemiol 2016; 45:1482-1492. [PMID: 27413102 DOI: 10.1093/ije/dyw143] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2016] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Plasma branched-chain amino acids (BCAAs, including leucine, isoleucine and valine) were recently related to risk of type 2 diabetes (T2D). Dietary intake is the only source of BCAAs; however, little is known about whether habitual dietary intake of BCAAs affects risk of T2D. METHODS We assessed associations between cumulative consumption of BCAAs and risk of T2D among participants from three prospective cohorts: the Nurses' Health Study (NHS; followed from 1980 to 2012); NHS II (followed from 1991 to 2011); and the Health Professionals Follow-up Study (HPFS; followed from 1986 to 2010). RESULTS We documented 16 097 incident T2D events during up to 32 years of follow-up. After adjustment for demographics and traditional risk factors, higher total BCAA intake was associated with an increased risk of T2D in men and women. In the meta-analysis of all cohorts, comparing participants in the highest quintile with those in the lowest quintile of intake, hazard ratios (95%confidence intervals) were for leucine 1.13 (1.07-1.19), for isoleucine 1.13 (1.07-1.19) and for valine 1.11 (1.05-1.17) (all P for trend < 0.001). In a healthy subsample, higher dietary BCAAs were significantly associated with higher plasma levels of these amino acids (P for trend = 0.01). CONCLUSIONS Our data suggest that high consumption of BCAAs is associated with an increased risk of T2D.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yanping Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Qibin Qi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Adela Hruby
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - JoAnn E Manson
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brian M Wolpin
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lu Qi
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA .,Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| |
Collapse
|
198
|
Bioactivity of Polyphenols: Preventive and Adjuvant Strategies toward Reducing Inflammatory Bowel Diseases-Promises, Perspectives, and Pitfalls. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9346470. [PMID: 27478535 PMCID: PMC4958438 DOI: 10.1155/2016/9346470] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/13/2016] [Accepted: 05/24/2016] [Indexed: 12/13/2022]
Abstract
Inflammatory bowel diseases (IBDs) are characterized by autoimmune and inflammation-related complications of the large intestine (ulcerative colitis) and additional parts of the digestive tract (Crohn's disease). Complications include pain, diarrhoea, chronic inflammation, and cancer. IBD prevalence has increased during the past decades, especially in Westernized countries, being as high as 1%. As prognosis is poor and medication often ineffective or causing side effects, additional preventive/adjuvant strategies are sought. A possible approach is via diets rich in protective constituents. Polyphenols, the most abundant phytochemicals, have been associated with anti-inflammatory, antioxidant, immunomodulatory, and apoptotic properties. Locally reducing oxidative stress, they can further act on cellular targets, altering gene expression related to inflammation, including NF-κB, Nrf-2, Jak/STAT, and MAPKs, suppressing downstream cytokine formation (e.g., IL-8, IL-1β, and TNF-α), and boosting the bodies' own antioxidant status (HO-1, SOD, and GPx). Moreover, they may promote, as prebiotics, healthy microbiota (e.g., Bifidobacteria, Akkermansia), short-chain fatty acid formation, and reduced gut permeability/improved tight junction stability. However, potential adverse effects such as acting as prooxidants, or perturbations of efflux transporters and phase I/II metabolizing enzymes, with increased uptake of undesired xenobiotics, should also be considered. In this review, we summarize current knowledge around preventive and arbitrary actions of polyphenols targeting IBD.
Collapse
|
199
|
Identification of key metabolic changes in renal interstitial fibrosis rats using metabonomics and pharmacology. Sci Rep 2016; 6:27194. [PMID: 27256510 PMCID: PMC4891668 DOI: 10.1038/srep27194] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 05/16/2016] [Indexed: 01/17/2023] Open
Abstract
Renal fibrosis is one of the important pathways involved in end-stage renal failure. Investigating the metabolic changes in the progression of disease may enhance the understanding of its pathogenesis and therapeutic information. In this study, (1)H-nuclear magnetic resonance (NMR)-based metabonomics was firstly used to screen the metabolic changes in urine and kidney tissues of renal interstitial fibrotic rats induced by unilateral ureteral obstruction (UUO), at 7, 14, 21, and 28 days after operation, respectively. The results revealed that reduced levels of bioenergy synthesis and branched chain amino acids (BCAAs), as well as elevated levels of indoxyl sulfate (IS) are involved in metabolic alterations of renal fibrosis rats. Next, by pharmacological treatment we found that reduction of IS levels could prevent the renal fibrotic symptoms. Therefore, we suggested that urinary IS may be used as a potential biomarker for the diagnosis of renal fibrosis, and a therapeutic target for drugs. Novel attempt combining metabonomics and pharmacology was established that have ability to provide more systematic diagnostic and therapeutic information of diseases.
Collapse
|
200
|
Ang JE, Pandher R, Ang JC, Asad YJ, Henley AT, Valenti M, Box G, de Haven Brandon A, Baird RD, Friedman L, Derynck M, Vanhaesebroeck B, Eccles SA, Kaye SB, Workman P, de Bono JS, Raynaud FI. Plasma Metabolomic Changes following PI3K Inhibition as Pharmacodynamic Biomarkers: Preclinical Discovery to Phase I Trial Evaluation. Mol Cancer Ther 2016; 15:1412-24. [PMID: 27048952 PMCID: PMC5321508 DOI: 10.1158/1535-7163.mct-15-0815] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/29/2016] [Indexed: 12/30/2022]
Abstract
PI3K plays a key role in cellular metabolism and cancer. Using a mass spectrometry-based metabolomics platform, we discovered that plasma concentrations of 26 metabolites, including amino acids, acylcarnitines, and phosphatidylcholines, were decreased in mice bearing PTEN-deficient tumors compared with non-tumor-bearing controls and in addition were increased following dosing with class I PI3K inhibitor pictilisib (GDC-0941). These candidate metabolomics biomarkers were evaluated in a phase I dose-escalation clinical trial of pictilisib. Time- and dose-dependent effects were observed in patients for 22 plasma metabolites. The changes exceeded baseline variability, resolved after drug washout, and were recapitulated on continuous dosing. Our study provides a link between modulation of the PI3K pathway and changes in the plasma metabolome and demonstrates that plasma metabolomics is a feasible and promising strategy for biomarker evaluation. Also, our findings provide additional support for an association between insulin resistance, branched-chain amino acids, and related metabolites following PI3K inhibition. Mol Cancer Ther; 15(6); 1412-24. ©2016 AACR.
Collapse
Affiliation(s)
- Joo Ern Ang
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom. Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Rupinder Pandher
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Joo Chew Ang
- School of Physics, University of Melbourne, Melbourne, Victoria, Australia
| | - Yasmin J Asad
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Alan T Henley
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Melanie Valenti
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Gary Box
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Alexis de Haven Brandon
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Richard D Baird
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom. Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | | | | | | | - Suzanne A Eccles
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Stan B Kaye
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom. Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Johann S de Bono
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom. Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom
| | - Florence I Raynaud
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom. Drug Development Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom.
| |
Collapse
|