1
|
Bork J, Markus MRP, Ewert R, Nauck M, Templin C, Völzke H, Kastenmüller G, Artati A, Adamski J, Dörr M, Friedrich N, Bahls M. The Metabolic Signature of Cardiorespiratory Fitness. Scand J Med Sci Sports 2025; 35:e70034. [PMID: 40072034 PMCID: PMC11899505 DOI: 10.1111/sms.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
High cardiorespiratory fitness (CRF) is associated with better overall health. This study aimed to find a metabolic signature associated with CRF to identify health-promoting effects. CRF based on cardiopulmonary exercise testing, targeted and untargeted metabolomics approaches based on mass spectrometry, and clinical data from two independent cohorts of the Study of Health in Pomerania (SHIP) were used. Sex-stratified linear regression models were adjusted for age, smoking, and height to relate CRF with individual metabolites. A total of 132 (SHIP-START-2: 483 men with a median age of 58 years and 450 women with a median age of 56 years) and 118 (SHIP-TREND-0: 341 men and 371 women both with a median age of 51 years) metabolites were associated with CRF. Lipids showed bidirectional relations to CRF independent of sex. Specific subsets of sphingomyelins were positively related to CRF in men (SM (OH) C14:1, SM(OH)C22:2 SM C16:0, SM C20:2 SM(OH)C24:1) and inversely in women (SM C16:1, SM C18:0, SM C18:1). Metabolites involved in energy production (citrate and succinylcarnitine) were only associated with CRF in men. In women, xenobiotics (hippurate, stachydrine) were related to CRF. The sex-specific metabolic signature of CRF is influenced by sphingomyelins, energy substrates, and xenobiotics. The greater effect estimates seen in women may emphasize the important role of CRF in maintaining metabolic health. Future research should explore how this profile changes with different types of exercise interventions or diseases in diverse populations and how these metabolites could be implemented in primary prevention settings.
Collapse
Affiliation(s)
- Julia Bork
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
| | - Marcello R. P. Markus
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
- German Center for Diabetes Research (DZD)Partner Site GreifswaldGreifswaldGermany
| | - Ralf Ewert
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
| | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medicine GreifswaldGreifswaldGermany
| | - Christian Templin
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
- Institute for Community Medicine, SHIP‐KEFUniversity Medicine GreifswaldGreifswaldGermany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems BiologyHelmholtz Zentrum MünchenNeuherbergGermany
| | - Anna Artati
- Metabolomics and Proteomics CoreHelmholtz Center MunichNeuherbergGermany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Department of Biochemistry, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute of Biochemistry, Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Marcus Dörr
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
| | - Nele Friedrich
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medicine GreifswaldGreifswaldGermany
| | - Martin Bahls
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
| |
Collapse
|
2
|
Healy DR, Zarei I, Mikkonen S, Soininen S, Viitasalo A, Haapala EA, Auriola S, Hanhineva K, Kolehmainen M, Lakka TA. Longitudinal associations of an exposome score with serum metabolites from childhood to adolescence. Commun Biol 2024; 7:890. [PMID: 39039257 PMCID: PMC11263428 DOI: 10.1038/s42003-024-06146-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 04/05/2024] [Indexed: 07/24/2024] Open
Abstract
Environmental and lifestyle factors, including air pollution, impaired diet, and low physical activity, have been associated with cardiometabolic risk factors in childhood and adolescence. However, environmental and lifestyle exposures do not exert their physiological effects in isolation. This study investigated associations between an exposome score to measure the impact of multiple exposures, including diet, physical activity, sleep duration, air pollution, and socioeconomic status, and serum metabolites measured using LC-MS and NMR, compared to the individual components of the score. A general population of 504 children aged 6-9 years at baseline was followed up for eight years. Data were analysed with linear mixed-effects models using the R software. The exposome score was associated with 31 metabolites, of which 12 metabolites were not associated with any individual exposure category. These findings highlight the value of a composite score to predict metabolic changes associated with multiple environmental and lifestyle exposures since childhood.
Collapse
Affiliation(s)
- Darren R Healy
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio Campus, Finland.
| | - Iman Zarei
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio Campus, Finland
| | - Santtu Mikkonen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio Campus, Finland
- Department of Technical Physics, University of Eastern Finland, Kuopio Campus, Finland
| | - Sonja Soininen
- Institute of Biomedicine, University of Eastern Finland, Kuopio Campus, Finland
- Physician and Nursing Services, Health and Social Services Centre, Wellbeing Services County of North Savo, Varkaus, Finland
| | - Anna Viitasalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio Campus, Finland
| | - Eero A Haapala
- Institute of Biomedicine, University of Eastern Finland, Kuopio Campus, Finland
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Seppo Auriola
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio Campus, Finland
- LC-MS Metabolomics Center, Biocenter Kuopio, Kuopio, Finland
| | - Kati Hanhineva
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio Campus, Finland
- Food Sciences Unit, Department of Life Technologies, University of Turku, Turku, Finland
| | - Marjukka Kolehmainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio Campus, Finland
| | - Timo A Lakka
- Institute of Biomedicine, University of Eastern Finland, Kuopio Campus, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| |
Collapse
|
3
|
Masenga SK, Povia JP, Lwiindi PC, Kirabo A. Recent Advances in Microbiota-Associated Metabolites in Heart Failure. Biomedicines 2023; 11:2313. [PMID: 37626809 PMCID: PMC10452327 DOI: 10.3390/biomedicines11082313] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Heart failure is a risk factor for adverse events such as sudden cardiac arrest, liver and kidney failure and death. The gut microbiota and its metabolites are directly linked to the pathogenesis of heart failure. As emerging studies have increased in the literature on the role of specific gut microbiota metabolites in heart failure development, this review highlights and summarizes the current evidence and underlying mechanisms associated with the pathogenesis of heart failure. We found that gut microbiota-derived metabolites such as short chain fatty acids, bile acids, branched-chain amino acids, tryptophan and indole derivatives as well as trimethylamine-derived metabolite, trimethylamine N-oxide, play critical roles in promoting heart failure through various mechanisms. Mainly, they modulate complex signaling pathways such as nuclear factor kappa-light-chain-enhancer of activated B cells, Bcl-2 interacting protein 3, NLR Family Pyrin Domain Containing inflammasome, and Protein kinase RNA-like endoplasmic reticulum kinase. We have also highlighted the beneficial role of other gut metabolites in heart failure and other cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Joreen P. Povia
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
| | - Propheria C. Lwiindi
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| |
Collapse
|
4
|
McCullough D, Harrison T, Enright KJ, Amirabdollahian F, Mazidi M, Lane KE, Stewart CE, Davies IG. The Effect of Carbohydrate Restriction on Lipids, Lipoproteins, and Nuclear Magnetic Resonance-Based Metabolites: CALIBER, a Randomised Parallel Trial. Nutrients 2023; 15:3002. [PMID: 37447328 DOI: 10.3390/nu15133002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Low-carbohydrate high-fat (LCHF) diets can be just as effective as high-carbohydrate, lower-fat (HCLF) diets for improving cardiovascular disease risk markers. Few studies have compared the effects of the UK HCLF dietary guidelines with an LCHF diet on lipids and lipoprotein metabolism using high-throughput NMR spectroscopy. This study aimed to explore the effect of an ad libitum 8-week LCHF diet compared to an HCLF diet on lipids and lipoprotein metabolism and CVD risk factors. For 8 weeks, n = 16 adults were randomly assigned to follow either an LCHF (n = 8, <50 g CHO p/day) or an HCLF diet (n = 8). Fasted blood samples at weeks 0, 4, and 8 were collected and analysed for lipids, lipoprotein subclasses, and energy-related metabolism markers via NMR spectroscopy. The LCHF diet increased (p < 0.05) very small VLDL, IDL, and large HDL cholesterol levels, whereas the HCLF diet increased (p < 0.05) IDL and large LDL cholesterol levels. Following the LCHF diet alone, triglycerides in VLDL and HDL lipoproteins significantly (p < 0.05) decreased, and HDL phospholipids significantly (p < 0.05) increased. Furthermore, the LCHF diet significantly (p < 0.05) increased the large and small HDL particle concentrations compared to the HCLF diet. In conclusion, the LCHF diet may reduce CVD risk factors by reducing triglyceride-rich lipoproteins and improving HDL functionality.
Collapse
Affiliation(s)
- D McCullough
- Carnegie School of Sport, Leeds Beckett University, Leeds LS6 3QS, UK
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - T Harrison
- Department of Clinical Sciences and Nutrition, University of Chester, Chester CH1 4BJ, UK
| | - K J Enright
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - F Amirabdollahian
- School of Health and Society, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - M Mazidi
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford OX1 7LF, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- Department of Twin Research & Genetic Epidemiology, South Wing St Thomas', King's College London, London SE1 7EH, UK
| | - K E Lane
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - C E Stewart
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - I G Davies
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool L3 3AF, UK
| |
Collapse
|
5
|
Zhao H, Ren Q, Wang HY, Zong Y, Zhao W, Wang Y, Qu M, Wang J. Alterations in gut microbiota and urine metabolomics in infants with yin-deficiency constitution aged 0–2 years. Heliyon 2023; 9:e14684. [PMID: 37064462 PMCID: PMC10102239 DOI: 10.1016/j.heliyon.2023.e14684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Background Based on the constitution theroy, infants are classified into balanced constitution (BC) and unbalanced constitution. Yin-deficiency constitution (YINDC) is a common type of unbalanced constitutions in Chinese infants. An infant's gut microbiota directly affects the child's health and has long-term effects on the maturation of the immune and endocrine systems throughout life. However, the gut microbiota of infants with YINDC remains unknown. Herein, we aimed to evaluate the intestinal flora profiles and urinary metabolites in infant with YINDC, find biomarkers to identify YINDC, and promote our understanding of infant constitution classification. Methods Constitutional Medicine Questionnaires were used to assess the infants' constitution types. 47 infants with 21 cases of YINDC and 26 cases of BC were included, and a cross-sectional sampling of stool and urine was conducted. Fecal microbiota was characterized using 16S rRNA sequencing, and urinary metabolomics was profiled using UPLC-Q-TOF/MS method. YINDC markers with high accuracy were identified using receiver operating characteristic (ROC) analysis. Results The diversity and composition of intestinal flora and urinary metabolites differed significantly between the YINDC and BC groups. A total of 13 obviously different genera and 55 altered metabolites were identified. Stool microbiome shifts were associated with urine metabolite changes. A combined marker comprising two genera may have a high potential to identify YINDC with an AUC of 0.845. Conclusions Infants with YINDC had a unique gut microbiota and metabolomic profile resulting in a constitutional microclassification. The altered gut microbiome in YINDC may account for the higher risk of cardiovascular diseases. Metabolomic analysis of urine showed that metabolic pathways, including histidine metabolism, proximal tubule bicarbonate reclamation, arginine biosynthesis, and steroid hormone biosynthesis, were altered in infants with YINDC. Additionally, the combined bacterial biomarker had the ability to identify YINDC. Identifying YINDC in infancy and intervening at an early stage is crucial for preventing cardiovascular diseases.
Collapse
|
6
|
Shen J, Guo H, Liu S, Jin W, Zhang ZW, Zhang Y, Liu K, Mao S, Zhou Z, Xie L, Wang G, Hao H, Liang Y. Aberrant branched-chain amino acid accumulation along the microbiota-gut-brain axis: Crucial targets affecting the occurrence and treatment of ischaemic stroke. Br J Pharmacol 2023; 180:347-368. [PMID: 36181407 DOI: 10.1111/bph.15965] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Although increasing evidence illustrated that the bidirectional communication between the brain and the gut is closely related to the occurrence of various complex diseases. Limited effort has been made to explore the influence of intestinal flora on the risk of ischaemic stroke. The present study aims to identify microbiota and specialized microbiota metabolites related to the occurrence and treatment of ischaemic stroke. EXPERIMENTAL APPROACH The role of microbiota in the occurrence and the treatment of ischaemic stroke was evaluated on ischaemia/reperfusion (I/R), pseudo-germ-free and faecal transplantation animals. The target microbiota and specialized metabolites were identified by comparing their distribution in flora and metabolomic profiles in ischaemic stroke patients and animals with compared with healthy controls. The effects and mechanisms involved of the targeted metabolites in ischaemic stroke were explored in ischaemia/reperfusion rats, hypoxia/reoxygenation PC12 cells and LPS-induced inflammatory BV2 cells. KEY RESULTS Both ischaemic stroke patients and I/R rats had significant accumulation of branched-chain amino acids, which were closely associated with gut microflora dysbiosis and the development of ischaemic stroke. Lactobacillus helveticus (L.hel) and Lactobacillus brevis (L.bre) are identified as the microbiota most affected by ischaemia/reperfusion modelling and treatment. L.hel and L.bre colonization exhibited significant neuroprotective activity and could greatly alleviate the accumulation of branched-chain amino acids. In addition, branched-chain amino acid (BCAA) accumulation was shown to exacerbate microglia-induced neuroinflammation by activating AKT/STAT3/NF-κB signalling. CONCLUSION AND IMPLICATIONS Our findings demonstrated the crucial role of intestinal flora and microbiota metabolites in the occurrence and treatment of ischaemic stroke.
Collapse
Affiliation(s)
- Jiajia Shen
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Huimin Guo
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Shijia Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Jin
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhi-Wei Zhang
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yong Zhang
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Keanqi Liu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Shuying Mao
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhihao Zhou
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lin Xie
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haiping Hao
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yan Liang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
7
|
Shen M, Xie Q, Zhang R, Yu C, Xiao P. Metabolite-assisted models improve risk prediction of coronary heart disease in patients with diabetes. Front Pharmacol 2023; 14:1175021. [PMID: 37033607 PMCID: PMC10081143 DOI: 10.3389/fphar.2023.1175021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Background: Patients with diabetes have a two-to four-fold increased incidence of cardiovascular diseases compared with non-diabetics. Currently, there is no recognized model to predict the occurrence and progression of CVDs in diabetics. Objective: This work aimed to develop a metabolic biomarker-assisted model, a combination of metabolic markers with clinical variables, for risk prediction of CVDs in diabetics. Methods: A total of 475 patients with diabetes were studied. Each patient underwent coronary angiography. Plasma samples were analyzed by liquid chromatography-quadrupole time-of-flight mass spectrometry. Ordinal logistic regression and random forest were used to screen metabolites. Receiver operating characteristic (ROC) curve, nomogram, and decision curve analysis (DCA) were employed to evaluate their prediction performances. Results: Ordinal logistic regression screened out 34 differential metabolites (adjusted-false discovery rate p < 0.05) from 2059 ion features by comparisons of diabetics with and without CVDs. Random forest identified methylglutarylcarnitine and lysoPC (18:0) as the metabolic markers (mean decrease gini >1.0) for non-significant CVDs (nos-CVDs) versus normal coronary artery (NCA), 1,3-Octadiene and 3-Octanone for acute coronary syndrome (ACS) versus nos-CVDs, and lysoPC (18:0) for acute coronary syndrome versus normal coronary artery. For risk prediction, the metabolic marker-assisted models provided areas under the curve of 0.962-0.979 by ROC (0.576-0.779 for the base models), and c-indices of 0.8477-0.9537 by nomogram analysis (0.1514-0.5196 for the base models). Decision curve analysis (DCA) showed that the models produced greater benefits throughout a wide range of risk probabilities compared with the base model. Conclusion: Metabolic biomarker-assisted model remarkably improved risk prediction of cardiovascular disease in diabetics (>90%).
Collapse
Affiliation(s)
- Min Shen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qingya Xie
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Ruizhe Zhang
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
- *Correspondence: Chunjing Yu, ; Pingxi Xiao,
| | - Pingxi Xiao
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Chunjing Yu, ; Pingxi Xiao,
| |
Collapse
|
8
|
Zhang W, Wan Z, Li X, Li R, Luo L, Song Z, Miao Y, Li Z, Wang S, Shan Y, Li Y, Chen B, Zhen H, Sun Y, Fang M, Ding J, Yan Y, Zong Y, Wang Z, Zhang W, Yang H, Yang S, Wang J, Jin X, Wang R, Chen P, Min J, Zeng Y, Li T, Xu X, Nie C. A population-based study of precision health assessments using multi-omics network-derived biological functional modules. Cell Rep Med 2022; 3:100847. [PMID: 36493776 PMCID: PMC9798030 DOI: 10.1016/j.xcrm.2022.100847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 10/05/2022] [Accepted: 11/11/2022] [Indexed: 12/13/2022]
Abstract
Recent technological advances in multi-omics and bioinformatics provide an opportunity to develop precision health assessments, which require big data and relevant bioinformatic methods. Here we collect multi-omics data from 4,277 individuals. We calculate the correlations between pairwise features from cross-sectional data and then generate 11 biological functional modules (BFMs) in males and 12 BFMs in females using a community detection algorithm. Using the features in the BFM associated with cardiometabolic health, carotid plaques can be predicted accurately in an independent dataset. We developed a model by comparing individual data with the health baseline in BFMs to assess health status (BFM-ash). Then we apply the model to chronic patients and modify the BFM-ash model to assess the effects of consuming grape seed extract as a dietary supplement. Finally, anomalous BFMs are identified for each subject. Our BFMs and BFM-ash model have huge prospects for application in precision health assessment.
Collapse
Affiliation(s)
- Wei Zhang
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Ziyun Wan
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Xiaoyu Li
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China,BGI Education Center, University of the Chinese Academy of Sciences, Shenzhen 518083, China
| | - Rui Li
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Lihua Luo
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China,BGI Education Center, University of the Chinese Academy of Sciences, Shenzhen 518083, China
| | - Zijun Song
- The First Affiliated Hospital, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Miao
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China,BGI Education Center, University of the Chinese Academy of Sciences, Shenzhen 518083, China
| | - Zhiming Li
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Shiyu Wang
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China,BGI Education Center, University of the Chinese Academy of Sciences, Shenzhen 518083, China
| | - Ying Shan
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Yan Li
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Bangwei Chen
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China,School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Hefu Zhen
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Yuzhe Sun
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Mingyan Fang
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Jiahong Ding
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Yizhen Yan
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Yang Zong
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Zhen Wang
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Wenwei Zhang
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China,James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Shuang Yang
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China,James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Ru Wang
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| | - Peijie Chen
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Zeng
- Center for Healthy Aging and Development Studies, National School of Development, Peking University, Beijing, China
| | - Tao Li
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China
| | - Chao Nie
- BGI-Shenzhen, Shenzhen 518083, China,China National GeneBank, Shenzhen 518120, China,Corresponding author
| |
Collapse
|
9
|
Caballero FF, Lana A, Struijk EA, Arias-Fernández L, Yévenes-Briones H, Cárdenas-Valladolid J, Salinero-Fort MÁ, Banegas JR, Rodríguez-Artalejo F, Lopez-Garcia E. Prospective Association Between Plasma Amino Acids And Multimorbidity In Older Adults. J Gerontol A Biol Sci Med Sci 2022; 78:637-644. [PMID: 35876753 DOI: 10.1093/gerona/glac144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Some amino acids have been associated with aging-related disorders and risk of physical impairment. The aim of this study was to assess the association between plasma concentrations of nine amino acids, including branched-chain and aromatic amino acids, and multimorbidity. METHODS This research uses longitudinal data from the Seniors-ENRICA 2 study, a population-based cohort from Spain which comprises non-institutionalized adults older than 65. Blood samples were extracted at baseline and after a follow-up period of two years for a total of 1488 subjects. Participants' information was linked with electronic health records. Chronic diseases were grouped into a list of 60 mutually exclusive conditions. A quantitative measure of multimorbidity, weighting morbidities by their regression coefficients on physical functioning, was employed and ranged from 0 to 100. Generalized estimating equation models were used to explore the relationship between plasma amino acids and multimorbidity, adjusting for sociodemographics, socioeconomic status and lifestyle behaviors. RESULTS The mean age of participants at baseline was 73.6 (SD = 4.2) years, 49.6% were women. Higher concentrations of glutamine [coef. per mmol/l (95% confidence interval = 10.1 (3.7, 16.6)], isoleucine [50.3 (21.7, 78.9)] and valine [15.5 (3.1, 28.0)] were significantly associated with higher multimorbidity scores, after adjusting for potential confounders. Body mass index could have influenced the relationship between isoleucine and multimorbidity (p = 0.016). CONCLUSIONS Amino acids could play a role in regulating aging-related diseases. Glutamine and branched-chain amino acids as isoleucine and valine are prospectively associated and could serve as risk markers for multimorbidity in older adults.
Collapse
Affiliation(s)
- Francisco Félix Caballero
- Department of Preventive Medicine and Public Health. Universidad Autónoma de Madrid and CIBER of Epidemiology and Public Health, Madrid
| | - Alberto Lana
- Department of Medicine. Universidad de Oviedo/ISPA, Oviedo
| | - Ellen A Struijk
- Department of Preventive Medicine and Public Health. Universidad Autónoma de Madrid and CIBER of Epidemiology and Public Health, Madrid
| | | | - Humberto Yévenes-Briones
- Department of Preventive Medicine and Public Health. Universidad Autónoma de Madrid and CIBER of Epidemiology and Public Health, Madrid
| | - Juan Cárdenas-Valladolid
- Dirección Técnica de Sistemas de Información. Gerencia Asistencial de Atención Primaria, Servicio Madrileño de Salud, Madrid.,Fundación de Investigación e Innovación Biosanitaria de Atención Primaria, Madrid.,Enfermería. Universidad Alfonso X El Sabio, Villanueva de la Cañada
| | - Miguel Ángel Salinero-Fort
- Fundación de Investigación e Innovación Biosanitaria de Atención Primaria, Madrid.,Subdirección General de Investigación Sanitaria. Consejería de Sanidad, Madrid.,Red de Investigación en Servicios de Salud en Enfermedades Crónicas.,Grupo de Envejecimiento y Fragilidad de las personas mayores. IdIPAZ, Madrid
| | - José R Banegas
- Department of Preventive Medicine and Public Health. Universidad Autónoma de Madrid and CIBER of Epidemiology and Public Health, Madrid
| | - Fernando Rodríguez-Artalejo
- Department of Preventive Medicine and Public Health. Universidad Autónoma de Madrid and CIBER of Epidemiology and Public Health, Madrid.,IMDEA-Food Institute. CEI UAM+CSIC, Madrid
| | - Esther Lopez-Garcia
- Department of Preventive Medicine and Public Health. Universidad Autónoma de Madrid and CIBER of Epidemiology and Public Health, Madrid.,IMDEA-Food Institute. CEI UAM+CSIC, Madrid
| |
Collapse
|
10
|
Hu ZK, Niu JL, Lin JJ, Guo Y, Dong LH. Proteomics of restenosis model in LDLR-deficient hamsters coupled with the proliferative rat vascular smooth muscle cells reveals a new mechanism of vascular remodeling diseases. J Proteomics 2022; 264:104634. [PMID: 35661764 DOI: 10.1016/j.jprot.2022.104634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/21/2022]
Abstract
A major pathological mechanism involved in vascular remodeling diseases is the proliferation and migration of vascular smooth muscle cells. The lipid distribution of golden hamsters is similar to that of humans, which makes them an excellent study model for studying the pathogenesis and molecular characteristics of vascular remodeling diseases. We performed proteomic analysis on Sprague Dawley rat VSMCs (rVSMCs) and restenosis hamsters with low-density lipoprotein receptor (LDLR) deficiency as part of this study. We have also performed the enrichment analysis of differentially modified proteins in regards to Gene Ontology, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and protein domain. 1070 differentially abundant proteins were assessed in rVSMCs before and after platelet-derived growth factor-BB (PDGF-BB) stimulation. Specifically, 1246 proteins displayed significant differences in the restenosis model in LDLR-deficient hamsters. An analysis of crosstalk between LDLR+/- hamsters artery restenosis and proliferating rVSMCs revealed 130 differentially expressed proteins, including 67 up-regulated proteins and 63 downregulated proteins. Enrichment analysis with KEGG showed differential proteins to be mainly concentrated in metabolic pathways. There are numerous differentially abundant proteins but particularly two proteins (phosphofructokinase 1 of liver type and lactate dehydrogenase A) were found to be up-regulated by PDGF-BB stimulation of rVSMCs and in a restenosis model of hamsters with LDLR+/- expression. SIGNIFICANCE: Based on bioinformatics, we have found glycolysis pathway plays an important role in both the LDLR+/- hamsters restenosis model and the proliferation of rVSMCs. Some key glycolysis enzymes may likely be developed either as new biomarkers or drug targets for vascular remodeling diseases.
Collapse
Affiliation(s)
- Zhao-Kun Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Medical Biotechnology of Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiang-Ling Niu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Medical Biotechnology of Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jia-Jie Lin
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Medical Biotechnology of Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yu Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Medical Biotechnology of Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Li-Hua Dong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Medical Biotechnology of Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
11
|
Amino Acid-Related Metabolic Signature in Obese Children and Adolescents. Nutrients 2022; 14:nu14071454. [PMID: 35406066 PMCID: PMC9003189 DOI: 10.3390/nu14071454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
The growing interest in metabolomics has spread to the search for suitable predictive biomarkers for complications related to the emerging issue of pediatric obesity and its related cardiovascular risk and metabolic alteration. Indeed, several studies have investigated the association between metabolic disorders and amino acids, in particular branched-chain amino acids (BCAAs). We have performed a revision of the literature to assess the role of BCAAs in children and adolescents' metabolism, focusing on the molecular pathways involved. We searched on Pubmed/Medline, including articles published until February 2022. The results have shown that plasmatic levels of BCAAs are impaired already in obese children and adolescents. The relationship between BCAAs, obesity and the related metabolic disorders is explained on one side by the activation of the mTORC1 complex-that may promote insulin resistance-and on the other, by the accumulation of toxic metabolites, which may lead to mitochondrial dysfunction, stress kinase activation and damage of pancreatic cells. These compounds may help in the precocious identification of many complications of pediatric obesity. However, further studies are still needed to better assess if BCAAs may be used to screen these conditions and if any other metabolomic compound may be useful to achieve this goal.
Collapse
|
12
|
Remme CA. Sudden cardiac death in diabetes and obesity: mechanisms and therapeutic strategies. Can J Cardiol 2022; 38:418-426. [PMID: 35017043 DOI: 10.1016/j.cjca.2022.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 02/07/2023] Open
Abstract
Ventricular arrhythmias and sudden cardiac death (SCD) occur most frequently in the setting of coronary artery disease, cardiomyopathy and heart failure, but are also increasingly observed in individuals suffering from diabetes mellitus and obesity. The incidence of these metabolic disorders is rising in Western countries, but adequate prevention and treatment of arrhythmias and SCD in affected patients is limited due to our incomplete knowledge of the underlying disease mechanisms. Here, an overview is presented of the prevalence of electrophysiological disturbances, ventricular arrhythmias and SCD in the clinical setting of diabetes and obesity. Experimental studies are reviewed, which have identified disease pathways and associated modulatory factors, in addition to pro-arrhythmic mechanisms. Key processes are discussed, including mitochondrial dysfunction, oxidative stress, cardiac structural derangements, abnormal cardiac conduction, ion channel dysfunction, prolonged repolarization and dysregulation of intracellular sodium and calcium homeostasis. In addition, the recently identified pro-arrhythmic effects of dysregulated branched chain amino acid metabolism, a common feature in patients with metabolic disorders, are addressed. Finally, current management options are discussed, in addition to the potential development of novel preventive and therapeutic strategies based on recent insight gained from translational studies.
Collapse
Affiliation(s)
- Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
13
|
De Spiegeleer M, De Paepe E, Van Meulebroek L, Gies I, De Schepper J, Vanhaecke L. Paediatric obesity: a systematic review and pathway mapping of metabolic alterations underlying early disease processes. Mol Med 2021; 27:145. [PMID: 34742239 PMCID: PMC8571978 DOI: 10.1186/s10020-021-00394-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The alarming trend of paediatric obesity deserves our greatest awareness to hinder the early onset of metabolic complications impacting growth and functionality. Presently, insight into molecular mechanisms of childhood obesity and associated metabolic comorbidities is limited. This systematic review aimed at scrutinising what has been reported on putative metabolites distinctive for metabolic abnormalities manifesting at young age by searching three literature databases (Web of Science, Pubmed and EMBASE) during the last 6 years (January 2015-January 2021). Global metabolomic profiling of paediatric obesity was performed (multiple biological matrices: blood, urine, saliva and adipose tissue) to enable overarching pathway analysis and network mapping. Among 2792 screened Q1 articles, 40 met the eligibility criteria and were included to build a database on metabolite markers involved in the spectrum of childhood obesity. Differential alterations in multiple pathways linked to lipid, carbohydrate and amino acid metabolisms were observed. High levels of lactate, pyruvate, alanine and acetate marked a pronounced shift towards hypoxic conditions in children with obesity, and, together with distinct alterations in lipid metabolism, pointed towards dysbiosis and immunometabolism occurring early in life. Additionally, aberrant levels of several amino acids, most notably belonging to tryptophan metabolism including the kynurenine pathway and its relation to histidine, phenylalanine and purine metabolism were displayed. Moreover, branched-chain amino acids were linked to lipid, carbohydrate, amino acid and microbial metabolism, inferring a key role in obesity-associated insulin resistance. CONCLUSIONS This systematic review revealed that the main metabolites at the crossroad of dysregulated metabolic pathways underlying childhood obesity could be tracked down to one central disturbance, i.e. impending insulin resistance for which reference values and standardised measures still are lacking. In essence, glycolytic metabolism was evinced as driving energy source, coupled to impaired Krebs cycle flux and ß-oxidation. Applying metabolomics enabled to retrieve distinct metabolite alterations in childhood obesity(-related insulin resistance) and associated pathways at early age and thus could provide a timely indication of risk by elucidating early-stage biomarkers as hallmarks of future metabolically unhealthy phenotypes.
Collapse
Affiliation(s)
- Margot De Spiegeleer
- Laboratory of Chemical Analysis, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Ellen De Paepe
- Laboratory of Chemical Analysis, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Lieven Van Meulebroek
- Laboratory of Chemical Analysis, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Inge Gies
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussel, Belgium
| | - Jean De Schepper
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussel, Belgium.,Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Lynn Vanhaecke
- Laboratory of Chemical Analysis, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium. .,Institute for Global Food Security, School of Biological Sciences, Queen's University, University Road, Belfast, BT7 1NN, UK.
| |
Collapse
|
14
|
Supruniuk E, Żebrowska E, Chabowski A. Branched chain amino acids-friend or foe in the control of energy substrate turnover and insulin sensitivity? Crit Rev Food Sci Nutr 2021; 63:2559-2597. [PMID: 34542351 DOI: 10.1080/10408398.2021.1977910] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Branched chain amino acids (BCAA) and their derivatives are bioactive molecules with pleiotropic functions in the human body. Elevated fasting blood BCAA concentrations are considered as a metabolic hallmark of obesity, insulin resistance, dyslipidaemia, nonalcoholic fatty liver disease, type 2 diabetes and cardiovascular disease. However, since increased BCAA amount is observed both in metabolically healthy and obese subjects, a question whether BCAA are mechanistic drivers of insulin resistance and its morbidities or only markers of metabolic dysregulation, still remains open. The beneficial effects of BCAA on body weight and composition, aerobic capacity, insulin secretion and sensitivity demand high catabolic potential toward amino acids and/or adequate BCAA intake. On the opposite, BCAA-related inhibition of lipogenesis and lipolysis enhancement may preclude impairment in insulin sensitivity. Thereby, the following review addresses various strategies pertaining to the modulation of BCAA catabolism and the possible roles of BCAA in energy homeostasis. We also aim to elucidate mechanisms behind the heterogeneity of ramifications associated with BCAA modulation.
Collapse
Affiliation(s)
- Elżbieta Supruniuk
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Żebrowska
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
15
|
Hübel C, Herle M, Santos Ferreira DL, Abdulkadir M, Bryant-Waugh R, Loos RJF, Bulik CM, Lawlor DA, Micali N. Childhood overeating is associated with adverse cardiometabolic and inflammatory profiles in adolescence. Sci Rep 2021; 11:12478. [PMID: 34127697 PMCID: PMC8203659 DOI: 10.1038/s41598-021-90644-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
Childhood eating behaviour contributes to the rise of obesity and related noncommunicable disease worldwide. However, we lack a deep understanding of biochemical alterations that can arise from aberrant eating behaviour. In this study, we prospectively associate longitudinal trajectories of childhood overeating, undereating, and fussy eating with metabolic markers at age 16 years to explore adolescent metabolic alterations related to specific eating patterns in the first 10 years of life. Data are from the Avon Longitudinal Study of Parents and Children (n = 3104). We measure 158 metabolic markers with a high-throughput (1H) NMR metabolomics platform. Increasing childhood overeating is prospectively associated with an adverse cardiometabolic profile (i.e., hyperlipidemia, hypercholesterolemia, hyperlipoproteinemia) in adolescence; whereas undereating and fussy eating are associated with lower concentrations of the amino acids glutamine and valine, suggesting a potential lack of micronutrients. Here, we show associations between early behavioural indicators of eating and metabolic markers.
Collapse
Affiliation(s)
- Christopher Hübel
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- UK National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health, South London and Maudsley Hospital, London, UK
- National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, Aarhus, Denmark
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Moritz Herle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Diana L Santos Ferreira
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Mohamed Abdulkadir
- Department of Pediatrics Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Rachel Bryant-Waugh
- Maudsley Centre for Child and Adolescent Eating Disorders, Michael Rutter Centre for Children and Young People, Maudsley Hospital, London, UK
| | - Ruth J F Loos
- Icahn School of Medicine At Mount Sinai, New York, NY, USA
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
| | - Deborah A Lawlor
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol National Institute of Health Research Biomedical Research Centre, Bristol, UK
| | - Nadia Micali
- Department of Pediatrics Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Great Ormond Street Institute of Child Health, University College London, London, UK.
| |
Collapse
|
16
|
Asghari G, Teymoori F, Farhadnejad H, Mirmiran P, Azizi F. Dietary Amino Acid Patterns Are Associated With Incidence of Chronic Kidney Disease. J Ren Nutr 2021; 32:312-318. [PMID: 34053818 DOI: 10.1053/j.jrn.2021.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/18/2021] [Accepted: 04/17/2021] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES No findings are available regarding the association of the dietary amino acid (AA) intakes with the risk of chronic kidney disease (CKD) in adults. We aimed to assess the association between dietary AA patterns and incidence of CKD in adults. DESIGN AND METHODS This study was conducted within the framework of Tehran Lipid and Glucose Study on 4,233 adults, who were free of CKD at baseline (2009-2011) and were followed for 3.1 years (2011-2014). The principal component analysis was used based on 8 AA groups to characterize major AA patterns. Multivariable logistic regression models were used to estimate the risk of CKD across quartiles of dietary AA pattern scores. RESULTS Three major AA patterns were extracted: (1) higher loads of branched-chain, alcoholic, and aromatic AAs; (2) higher loads of acidic AAs, proline, and lower load of alkaline AAs and small AAs; and (3) higher loads of sulfuric AAs and small AAs. The mean ± standard deviation age of participants (45.9% male) was 39.4 ± 11.7 at baseline and the incidence of CKD was 12.1% (513 cases) after 3.1 years of follow-up. The highest score of first dietary AA pattern tended to be associated with an increased risk of CKD (odds ratio = 1.43, 95% confidence interval1.06-1.93); however, the second pattern was related to a decreased risk of CKD (odds ratio = 0.71, 95% confidence interval: 0.52-0.97) in the multivariable model. CONCLUSION These novel findings suggest that the dietary AA patterns may modify risk of CKD.
Collapse
Affiliation(s)
- Golaleh Asghari
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farshad Teymoori
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Farhadnejad
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Lischka J, Schanzer A, Hojreh A, Ba Ssalamah A, Item CB, de Gier C, Walleczek N, Metz TF, Jakober I, Greber‐Platzer S, Zeyda M. A branched-chain amino acid-based metabolic score can predict liver fat in children and adolescents with severe obesity. Pediatr Obes 2021; 16:e12739. [PMID: 33058486 PMCID: PMC7988615 DOI: 10.1111/ijpo.12739] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Eighty percent of adolescents with severe obesity suffer from non-alcoholic fatty liver disease (NAFLD). Non-invasive prediction models have been tested in adults, however, they performed poorly in paediatric populations. OBJECTIVE This study aimed to investigate novel biomarkers for NAFLD and to develop a score that predicts liver fat in youth with severe obesity. METHODS From a population with a BMI >97th percentile aged 9-19 years (n = 68), clinically thoroughly characterized including MRI-derived proton density fat fraction (MRI-PDFF), amino acids and acylcarnitines were measured by HPLC-MS. RESULTS In children with NAFLD, higher levels of plasma branched-chain amino acids (BCAA) were determined. BCAAs correlated with MRI-PDFF (R = 0.46, p < .01). We identified a linear regression model adjusted for age, sex and pubertal stage consisting of BCAAs, ALT, GGT, ferritin and insulin that predicted MRI-PDFF (R = 0.75, p < .01). ROC analysis of this model revealed AUCs of 0.85, 0.85 and 0.92 for the detection of any, moderate and severe steatosis, respectively, thus markedly outperforming previously published scores. CONCLUSION BCAAs could be an important link between obesity and other metabolic pathways. A BCAA-based metabolic score can predict steatosis grade in high-risk children and adolescents and may provide a feasible alternative to sophisticated methods like MRI or biopsy in the future.
Collapse
Affiliation(s)
- Julia Lischka
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria,Comprehensive Center for Pediatrics, Medical University of ViennaViennaAustria
| | - Andrea Schanzer
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria,Comprehensive Center for Pediatrics, Medical University of ViennaViennaAustria
| | - Azadeh Hojreh
- Comprehensive Center for Pediatrics, Medical University of ViennaViennaAustria,Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Ahmed Ba Ssalamah
- Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Chike Bellarmine Item
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
| | - Charlotte de Gier
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria,Comprehensive Center for Pediatrics, Medical University of ViennaViennaAustria
| | - Nina‐Katharina Walleczek
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria,Comprehensive Center for Pediatrics, Medical University of ViennaViennaAustria
| | - Thomas F. Metz
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
| | - Ivana Jakober
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
| | - Susanne Greber‐Platzer
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria,Comprehensive Center for Pediatrics, Medical University of ViennaViennaAustria
| | - Maximilian Zeyda
- Clinical Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria,Comprehensive Center for Pediatrics, Medical University of ViennaViennaAustria
| |
Collapse
|
18
|
Enko D, Moro T, Holasek S, Baranyi A, Schnedl WJ, Zelzer S, Mangge H, Herrmann M, Meinitzer A. Branched-chain amino acids are linked with iron metabolism. ANNALS OF TRANSLATIONAL MEDICINE 2021; 8:1569. [PMID: 33437768 PMCID: PMC7791222 DOI: 10.21037/atm-20-624a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background The branched-chain amino acids (BCAAs) valine, leucine and isoleucine are reported to influence erythropoiesis and the human iron status. Large study cohorts encompassing biomarkers of iron metabolism and BCAAs are still lacking. Methods We investigated potential interactions between blood concentrations of all three BCAAs valine, leucine and isoleucine and biomarkers of iron metabolism [i.e., hemoglobin (Hb), mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), iron, transferrin, ferritin, transferrin saturation, soluble transferrin receptor (sTfR)] in 430 outpatients referred for a medical health check-up. Linear regression models were performed to assess possible associations between variables. Results All three BCAAs were positively correlated with Hb, ferritin and the sTfR (r-values: 0.145–0.382; P values: <0.001–0.003). The strongest correlation was observed between valine and Hb (r=0.382; P value <0.001). Linear regression models showed a statistically significant influence of all three BCAAs on Hb and ferritin (β-coefficients: 0.173–0.351; all P values: <0.001). Seventeen patients with anemia (4%) were found with significantly lower serum BCAA concentrations compared to 413 non-anemic individuals (P<0.05). Conclusions These data indicate a pathophysiological link between the three BCAAs valine, leucine and isoleucine and the human iron indicators Hb and ferritin. Further studies are needed to clarify the exact causal mechanisms of these findings.
Collapse
Affiliation(s)
- Dietmar Enko
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Hochsteiermark, Leoben, Austria
| | - Thomas Moro
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Sandra Holasek
- Department of Immunology and Pathophysiology, Medical University of Graz, Otto Loewi Research Center, Graz, Austria
| | - Andreas Baranyi
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | | | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
19
|
Wang F, Wang B, Chen X, Liu W, Wang G, Li X, Liu X, Li N, Zhang J, Yin T, Jing J, Chang X, Jin Y, Zhang Y, Zhao Y. Association Between Blood Pressure and Branched-Chain/Aromatic Amino Acid Excretion Rate in 24-Hour Urine Samples from Elderly Hypertension Patients. Diabetes Metab Syndr Obes 2021; 14:3965-3973. [PMID: 34531669 PMCID: PMC8439623 DOI: 10.2147/dmso.s324153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Recently, the association between lifestyle-related diseases and free amino acids in the blood plasma-especially branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs)-has been highlighted. However, few studies have been carried out on 24-hour urine samples. Therefore, we aimed to explore the relationships between 24-hour urinary BCAAs and AAAs excretion rate and blood pressure (BP) in elderly patients with hypertension. METHODS AND RESULTS Each of the 322 elderly patients with hypertension completed an in-person questionnaire interview, underwent a physical examination, and provided a 24-hour urine specimen. We measured their BCAAs and AAAs excretion rate, and used multiple linear regression analysis with variable selection to construct models describing the relationships between their BCAAs and AAAs excretion rate and BP. After adjusted for age, gender, height, and weight, valine was inversely associated with both systolic blood pressure (SBP) (β: -0.232, 95% CI: -0.16, -0.006) and diastolic blood pressure (DBP) (β: -0.144, 95% CI: -0.089, -0.005). These findings were invariant even following adjustment for urine volume and drugs history, and Ile was positively associated with DBP (β: 0.170, 95% CI: 0.001, 0.066). CONCLUSION The data revealed that the excretion rate of 24-hour urinary BCAAs was closely related to BP in elderly hypertension patients, and these findings will provide new insights into the association between BACC metabolism and BP.
Collapse
Affiliation(s)
- Faxuan Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Binxia Wang
- Second People’s Hospital of Gansu Province, Lanzhou, Gansu, People’s Republic of China
| | - Xiyuan Chen
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Wanlu Liu
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Guoqi Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Xiaoxia Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Xiuying Liu
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Nan Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Jiaxing Zhang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Ting Yin
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Jinyun Jing
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Xiaoyu Chang
- Editorial Department of the Journal of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Yanan Jin
- Centers for Disease Control and Prevention in Ningxia, Yinchuan, Ningxia, People’s Republic of China
| | - Yuhong Zhang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, Ningxia, People’s Republic of China
| | - Yi Zhao
- School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Correspondence: Yi Zhao School of Public Health and Management, Ningxia Medical UniversityTel +86 139 9501 1690Fax +86 951-6980144 Email
| |
Collapse
|
20
|
Liu L, Jin X, Wu Y, Yang M, Xu T, Li X, Ren J, Yan LL. A Novel Dried Blood Spot Detection Strategy for Characterizing Cardiovascular Diseases. Front Cardiovasc Med 2020; 7:542519. [PMID: 33195447 PMCID: PMC7583634 DOI: 10.3389/fcvm.2020.542519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/21/2020] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in China. Conventional diagnostic methods are dependent on advanced instruments, which are expensive, inaccessible, and inconvenient in underdeveloped areas. To build a novel dried blood spot (DBS) detection strategy for imaging CVDs, in this study, a total of 12 compounds, including seven amino acids [homocysteine (Hcy), isoleucine (Ile), leucine (Leu), valine (Val), phenylalanine (Phe), tyrosine (Tyr), and tryptophan (Trp)], three amino acid derivatives [choline, betaine, and trimethylamine N-oxide (TMAO)], L-carnitine, and creatinine, were screened for their ability to identify CVD. A rapid and reliable method was established for the quantitative analysis of the 12 compounds in DBS. A total of 526 CVD patients and 200 healthy volunteers in five provinces of China were recruited and divided into the following groups: stroke, coronary heart disease, diabetes, and high blood pressure. The orthogonal projection to latent structures-discriminant analysis (OPLSDA) model was used to characterize the difference between each CVD group. Marked differences between the groups based on the OPLSDA model were observed. Based on the model, the patients in the three training sets were mostly accurately categorized into the appropriate group. In addition, the receiver operating characteristic (ROC) curves and logistic regression of each metabolite chosen by the OPLSDA model had an excellent predictive value in both the test and validation groups. DBS detection of 12 biomarkers was sensitive and powerful for characterizing different types of CVD. Such differentiation may reduce unnecessary invasive coronary angiography, enhance predictive value, and complement current diagnostic methods.
Collapse
Affiliation(s)
- Linsheng Liu
- Clinical Pharmacology Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xurui Jin
- Global Health Research Center, Duke Kunshan University, Kunshan, China
| | - Yangfeng Wu
- Peking University Clinical Research Institute, Beijing, China
| | - Mei Yang
- Suzhou BioNovoGene Metabolomics Platform, Suzhou, China
| | - Tao Xu
- The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, China.,The Therapeutic Antibody Research Center of SEU-Alphamab, Southeast University, Nanjing, China
| | - Xianglian Li
- Clinical Pharmacology Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianhong Ren
- Suzhou BioNovoGene Metabolomics Platform, Suzhou, China
| | - Lijing L Yan
- Global Health Research Center, Duke Kunshan University, Kunshan, China
| |
Collapse
|
21
|
Proffitt C, Bidkhori G, Moyes D, Shoaie S. Disease, Drugs and Dysbiosis: Understanding Microbial Signatures in Metabolic Disease and Medical Interventions. Microorganisms 2020; 8:microorganisms8091381. [PMID: 32916966 PMCID: PMC7565856 DOI: 10.3390/microorganisms8091381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Since the discovery of the potential role for the gut microbiota in health and disease, many studies have gone on to report its impact in various pathologies. These studies have fuelled interest in the microbiome as a potential new target for treating disease Here, we reviewed the key metabolic diseases, obesity, type 2 diabetes and atherosclerosis and the role of the microbiome in their pathogenesis. In particular, we will discuss disease associated microbial dysbiosis; the shift in the microbiome caused by medical interventions and the altered metabolite levels between diseases and interventions. The microbial dysbiosis seen was compared between diseases including Crohn’s disease and ulcerative colitis, non-alcoholic fatty liver disease, liver cirrhosis and neurodegenerative diseases, Alzheimer’s and Parkinson’s. This review highlights the commonalities and differences in dysbiosis of the gut between diseases, along with metabolite levels in metabolic disease vs. the levels reported after an intervention. We identify the need for further analysis using systems biology approaches and discuss the potential need for treatments to consider their impact on the microbiome.
Collapse
Affiliation(s)
- Ceri Proffitt
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
- Correspondence: (C.P.); (S.S.)
| | - Gholamreza Bidkhori
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
| | - David Moyes
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
| | - Saeed Shoaie
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden
- Correspondence: (C.P.); (S.S.)
| |
Collapse
|
22
|
Telle-Hansen VH, Christensen JJ, Formo GA, Holven KB, Ulven SM. A comprehensive metabolic profiling of the metabolically healthy obesity phenotype. Lipids Health Dis 2020; 19:90. [PMID: 32386512 PMCID: PMC7211343 DOI: 10.1186/s12944-020-01273-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/29/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The ever-increasing prevalence of obesity constitutes a major health problem worldwide. A subgroup of obese individuals has been described as "metabolically healthy obese" (MHO). In contrast to metabolically unhealthy obese (MUO), the MHO phenotype has a favorable risk profile. Despite this, the MHO phenotype is still sub-optimally characterized with respect to a comprehensive risk assessment. Our aim was to increase the understanding of metabolic alterations associated with healthy and unhealthy obesity. METHODS In this cross-sectional study, men and women (18-70 years) with obesity (body mass index (BMI) ≥ 30 kg/m2) or normal weight (NW) (BMI ≤ 25 kg/m2) were classified with MHO (n = 9), MUO (n = 10) or NW (n = 11) according to weight, lipid profile and glycemic regulation. We characterized individuals by comprehensive metabolic profiling using a commercial available high-throughput proton NMR metabolomics platform. Plasma fatty acid profile, including short chain fatty acids, was measured using gas chromatography. RESULTS The concentrations of very low density lipoprotein (VLDL), intermediate density lipoprotein (IDL) and low density lipoprotein (LDL) subclasses were overall significantly higher, and high density lipoprotein (HDL) subclasses lower in MUO compared with MHO. VLDL and IDL subclasses were significantly lower and HDL subclasses were higher in NW compared with MHO. The concentration of isoleucine, leucine and valine was significantly higher in MUO compared with MHO, and the concentration phenylalanine was lower in NW subjects compared with MHO. The fatty acid profile in MHO was overall more favorable compared with MUO. CONCLUSIONS Comprehensive metabolic profiling supports that MHO subjects have intermediate-stage cardiovascular disease risk marker profile compared with NW and MUO subjects. CLINICAL TRIAL REGISTRATION NUMBER NCT01034436, Fatty acid quality and overweight (FO-study).
Collapse
Affiliation(s)
- Vibeke H Telle-Hansen
- Faculty of Health Sciences, Oslo Metropolitan University, P.O. Box 4, St. Olavsplass, 0130, Oslo, Norway.
| | - Jacob J Christensen
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital Rikshospitalet, P.O. Box 4950, Nydalen, 0424, Oslo, Norway.,Departmentof Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway
| | - Gulla Aase Formo
- Departmentof Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway
| | - Kirsten B Holven
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital Rikshospitalet, P.O. Box 4950, Nydalen, 0424, Oslo, Norway.,Departmentof Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway
| | - Stine M Ulven
- Departmentof Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1046, Blindern, 0317, Oslo, Norway
| |
Collapse
|
23
|
Mangge H, Herrmann M, Almer G, Zelzer S, Moeller R, Horejsi R, Renner W. Telomere shortening associates with elevated insulin and nuchal fat accumulation. Sci Rep 2020; 10:6863. [PMID: 32322021 PMCID: PMC7176638 DOI: 10.1038/s41598-020-63916-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/19/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity and relative leucocyte telomere length (RTL) are both linked to accelerated aging and premature mortality. We examined if nuchal subcutaneous adipose tissue (SAT) thickness, a surrogate marker of central trunk-weighted obesity, is an independent predictor of RTL that provides information beyond BMI, metabolic and inflammatory markers. RTL and nuchal SAT thickness were determined in 362 participants of the STYJOBS/EDECTA study (STYrian Juvenile Obesity Study, Early DEteCTion of atherosclerosis), which included overweight individuals and matched eutrophic controls. Fasting plasma samples were used for the measurement of leptin, resistin, adiponectin, glucose, insulin, high-sensitivity C-reactive protein (hs-CRP), interleukin-6 (IL-6), liver enzymes, creatinine, cholesterol, high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, oxidized LDL, triglycerides, homocysteine and uric acid. Furthermore, all participants underwent carotid artery ultrasound. Obese individuals had markedly higher body mass index (BMI), nuchal SAT thickness, hip and waist circumferences and carotid intima media thickness (IMT) than eutrophic controls. In addition, they showed typical biochemical abnormalities related to energy metabolism, systemic inflammation and liver function. RTL was inversely correlated with nuchal SAT thickness, IMT, hs-CRP, alkaline phosphatase, insulin, resistin, and leptin. Positive correlations were seen with homocysteine and creatinine. Stepwise linear regression analyses identified nuchal SAT thickness and insulin as the only significant predictors of RTL. In conclusion, nuchal SAT thickness is a robust predictor of RTL that provides information beyond traditional obesity-related metabolic and inflammatory biomarkers. This suggests an important role of fat depots at the neck for accelerated telomere shortening.
Collapse
Affiliation(s)
- Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Gunter Almer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Reinhard Moeller
- Otto Loewi Research Center (for Vascular Biology, Immunology and Inflammation), Medical University of Graz, Graz, Austria
| | - Renate Horejsi
- Otto Loewi Research Center (for Vascular Biology, Immunology and Inflammation), Medical University of Graz, Graz, Austria
| | - Wilfried Renner
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
24
|
Biswas D, Tozer K, Dao KT, Perez LJ, Mercer A, Brown A, Hossain I, Yip AM, Aguiar C, Motawea H, Brunt KR, Shea J, Legare JF, Hassan A, Kienesberger PC, Pulinilkunnil T. Adverse Outcomes in Obese Cardiac Surgery Patients Correlates With Altered Branched-Chain Amino Acid Catabolism in Adipose Tissue and Heart. Front Endocrinol (Lausanne) 2020; 11:534. [PMID: 32903728 PMCID: PMC7438793 DOI: 10.3389/fendo.2020.00534] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/30/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Predicting relapses of post-operative complications in obese patients who undergo cardiac surgery is significantly complicated by persistent metabolic maladaptation associated with obesity. Despite studies supporting the linkages of increased systemic branched-chain amino acids (BCAAs) driving the pathogenesis of obesity, metabolome wide studies have either supported or challenged association of circulating BCAAs with cardiovascular diseases (CVDs). Objective: We interrogated whether BCAA catabolic changes precipitated by obesity in the heart and adipose tissue can be reliable prognosticators of adverse outcomes following cardiac surgery. Our study specifically clarified the correlation between BCAA catabolizing enzymes, cellular BCAAs and branched-chain keto acids (BCKAs) with the severity of cardiometabolic outcomes in obese patients pre and post cardiac surgery. Methods: Male and female patients of ages between 44 and 75 were stratified across different body mass index (BMI) (non-obese = 17, pre-obese = 19, obese class I = 14, class II = 17, class III = 12) and blood, atrial appendage (AA), and subcutaneous adipose tissue (SAT) collected during cardiac surgery. Plasma and intracellular BCAAs and BC ketoacids (BCKAs), tissue mRNA and protein expression and activity of BCAA catabolizing enzymes were assessed and correlated with clinical parameters. Results: Intramyocellular, but not systemic, BCAAs increased with BMI in cardiac surgery patients. In SAT, from class III obese patients, mRNA and protein expression of BCAA catabolic enzymes and BCKA dehydrogenase (BCKDH) enzyme activity was decreased. Within AA, a concomitant increase in mRNA levels of BCAA metabolizing enzymes was observed, independent of changes in BCKDH protein expression or activity. BMI, indices of tissue dysfunction and duration of hospital stay following surgery correlated with BCAA metabolizing enzyme expression and metabolite levels in AA and SAT. Conclusion: This study proposes that in a setting of obesity, dysregulated BCAA catabolism could be an effective surrogate to determine cardiac surgery outcomes and plausibly predict premature re-hospitalization.
Collapse
Affiliation(s)
- Dipsikha Biswas
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
- IMPART Investigator Team Canada, Saint John, NB, Canada
| | - Kathleen Tozer
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Khoi T. Dao
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Lester J. Perez
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Angella Mercer
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
- IMPART Investigator Team Canada, Saint John, NB, Canada
| | - Amy Brown
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Intekhab Hossain
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Alexandra M. Yip
- New Brunswick Heart Centre, Saint John Regional Hospital, Saint John, NB, Canada
| | - Christie Aguiar
- IMPART Investigator Team Canada, Saint John, NB, Canada
- New Brunswick Heart Centre, Saint John Regional Hospital, Saint John, NB, Canada
| | - Hany Motawea
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
- IMPART Investigator Team Canada, Saint John, NB, Canada
| | - Keith R. Brunt
- IMPART Investigator Team Canada, Saint John, NB, Canada
- Department of Pharmacology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jennifer Shea
- Department of Pathology, Dalhousie University, Saint John, NB, Canada
- Department of Laboratory Medicine, Saint John Regional Hospital, Saint John, NB, Canada
| | - Jean F. Legare
- IMPART Investigator Team Canada, Saint John, NB, Canada
- New Brunswick Heart Centre, Saint John Regional Hospital, Saint John, NB, Canada
| | - Ansar Hassan
- IMPART Investigator Team Canada, Saint John, NB, Canada
- New Brunswick Heart Centre, Saint John Regional Hospital, Saint John, NB, Canada
| | - Petra C. Kienesberger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
- IMPART Investigator Team Canada, Saint John, NB, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Dalhousie University, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
- IMPART Investigator Team Canada, Saint John, NB, Canada
- *Correspondence: Thomas Pulinilkunnil
| |
Collapse
|
25
|
ALMEIDA APD, FORTES FS, SILVEIRA BKS, REIS NDA, HERMSDORFf HHM. Branched-Chain amino acids intake is negatively related to body adiposity in individuals at cardiometabolic risk. REV NUTR 2020. [DOI: 10.1590/1678-9865202033e190208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT Objective To assess the relationship between branched-chain amino acids intake in the current diet and the metabolic and body adiposity markers in a population at cardiovascular risk. Methods This is a cross-sectional study with 282 adults and elderly people from the Cardiovascular Health Care Program of the Universidade Federal de Viçosa. Sociodemographic, anthropometric and body composition data, as well as metabolic biomarkers were collected using standardized protocols. Dietary intake of branched amino acids was assessed using a 24-hour recall. Results Individuals with a higher branched-chain amino acids intake (≥2.6g/day, median value) had lower body fat (29.6 vs 32.2%; p=0.019), and higher serum ferritin (113.2 vs. 60.1mg/dL; p=0.006) and uric acid concentrations (4.4 vs. 4.0; p=0.023). In addition, a lower prevalence of overweight and excessive abdominal fat (p<0.05) was found in the individuals with higher branched-chain amino acids intake. They also had a higher daily intake of fiber, copper, zinc, magnesium, and iron, as well as a lower intake of total lipids. Conclusion In the present study, the intake of branched amino acids is negatively related to total and central adiposity, but more studies are needed to fully elucidate this possible relationship. (Brazilian Registry of Clinical Trials, code RBR-5n4y2g).
Collapse
|
26
|
Fellendorf FT, Platzer M, Pilz R, Rieger A, Kapfhammer HP, Mangge H, Dalkner N, Zelzer S, Meinitzer A, Birner A, Bengesser SA, Queissner R, Hamm C, Hartleb R, Reininghaus EZ. Branched-chain amino acids are associated with metabolic parameters in bipolar disorder. World J Biol Psychiatry 2019; 20:821-826. [PMID: 29898625 DOI: 10.1080/15622975.2018.1487077] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Objectives: An important aspect of bipolar disorder (BD) research is the identification of biomarkers pertaining to the somatic health state. The branched-chain essential amino acids (BCAAs), viz valine, leucine and isoleucine, have been proposed as biomarkers of an individual's health state, given their influence on protein synthesis and gluconeogenesis inhibition.Methods: BCAA levels of 141 euthymic/subsyndromal individuals with BD and 141 matched healthy controls (HC) were analysed by high-pressure lipid chromatography and correlated with clinical psychiatric, anthropometric and metabolic parameters.Results: BD and HC did not differ in valine and isoleucine, whereas leucine was significantly lower in BD. Furthermore, correlations were found between BCAAs and anthropometric and glucose metabolism data. All BCAAs correlated with lipid metabolism parameters in females. There were no associations between BCAAs and long-term clinical parameters of BD. A negative correlation was found between valine and Hamilton Depression-Scale, and Beck Depression Inventory II, in male individualsConclusions: Our results indicate the utility of BCAAs as biomarkers for the current state of health, also in BD. As BD individuals have a high risk for overweight/obesity, in association with comorbid medical conditions (e.g. cardiovascular diseases or insulin resistance), health state markers are urgently required. However, no illness-specific associations were found in this euthymic/subsyndromal BD group.
Collapse
Affiliation(s)
- Frederike T Fellendorf
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Martina Platzer
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Rene Pilz
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Alexandra Rieger
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Hans-Peter Kapfhammer
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Nina Dalkner
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Armin Birner
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Susanne A Bengesser
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Robert Queissner
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Carlo Hamm
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Riccarda Hartleb
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Eva Z Reininghaus
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
27
|
Suzuki Y, Kido J, Matsumoto S, Shimizu K, Nakamura K. Associations among amino acid, lipid, and glucose metabolic profiles in childhood obesity. BMC Pediatr 2019; 19:273. [PMID: 31387549 PMCID: PMC6683574 DOI: 10.1186/s12887-019-1647-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023] Open
Abstract
Background Plasma-free amino acid profiles have been reported to correlate with obesity and glucose metabolism, and have been studied as potentially useful biomarkers of lifestyle-related diseases affecting metabolism in adulthood. However, knowledge of these relationships is lacking in children, despite the growing public health problem posed by childhood obesity. The aim of this study was to assess whether plasma-free amino acid profiles can serve as useful biomarkers of lifestyle-related diseases in children with obesity. Methods This retrospective study used the medical records of 26 patients (15 male, 11 female) aged 9 or 10 years presenting with moderate to severe obesity and hyperlipidemia between April 2015 and March 2017. A degree of obesity of 30% or more was defined as moderate or severe. Amino acid levels were compared between obese children with and without impaired glucose tolerance using a t-test or Mann–Whitney U test. In addition, the influence of factors such as intima media thickness, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, amino acids, and homeostasis model assessment-insulin resistance (HOMA-IR) were analyzed pairwise using Pearson’s correlation or Spearman’s rank correlation. Results HOMA-IR was positively correlated with valine, leucine (Leu), isoleucine, phenylalanine, tryptophan, methionine, threonine, lysine, alanine, tyrosine, glutamate (Glu), proline, arginine, ornithine, total free amino acids (all P < 0.01), and aspartate (P = 0.010). Moreover, blood uric acid levels were positively correlated with Leu (P = 0.005) and Glu (P = 0.019), and negatively correlated with serine, glycine, and asparagine (P = 0.007, P = 0.003, and P = 0.013, respectively). Conclusions Amino acid profile reflects impaired glucose tolerance and hyperuricemia at an early stage of obesity. It is therefore a useful marker to inform early intervention in children with obesity, as in adults. Electronic supplementary material The online version of this article (10.1186/s12887-019-1647-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yosuke Suzuki
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| | - Jun Kido
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| | - Shirou Matsumoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan.
| | - Kie Shimizu
- Department of Central Radiology, Kumamoto University Hospital, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| |
Collapse
|
28
|
Tobias DK, Lawler PR, Harada PH, Demler OV, Ridker PM, Manson JE, Cheng S, Mora S. Circulating Branched-Chain Amino Acids and Incident Cardiovascular Disease in a Prospective Cohort of US Women. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002157. [PMID: 29572205 DOI: 10.1161/circgen.118.002157] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Circulating branched-chain amino acids (BCAAs; isoleucine, leucine, and valine) are strong predictors of type 2 diabetes mellitus (T2D), but their association with cardiovascular disease (CVD) is uncertain. We hypothesized that plasma BCAAs are positively associated with CVD risk and evaluated whether this was dependent on an intermediate diagnosis of T2D. METHODS Participants in the Women's Health Study prospective cohort were eligible if free of CVD at baseline blood collection (n=27 041). Plasma metabolites were measured via nuclear magnetic resonance spectroscopy. Multivariable Cox regression models estimated hazard ratios (HRs) and 95% confidence intervals (CIs) for BCAAs with incident CVD (myocardial infarction, stroke, and coronary revascularization). RESULTS We confirmed 2207 CVD events over a mean 18.6 years of follow-up. Adjusting for age, body mass index, and other established CVD risk factors, total BCAAs were positively associated with CVD (per SD: HR, 1.13; 95% CI, 1.08-1.18), comparable to LDL-C (low-density lipoprotein cholesterol) with CVD (per SD: HR, 1.12; 95% CI, 1.07-1.17). BCAAs were associated with coronary events (myocardial infarction: HR, 1.16; 95% CI, 1.06-1.26; revascularization: HR, 1.17; 95% CI, 1.11-1.25), and borderline significant association with stroke (HR, 1.07; 95% CI, 0.99-1.15). The BCAA-CVD association was greater (P interaction=0.036) among women who developed T2D before CVD (HR, 1.20; 95% CI, 1.08-1.32) versus women without T2D (HR, 1.08; 95% CI, 1.03-1.14). Adjusting for LDL-C, an established CVD risk factor, did not attenuate these findings; however, adjusting for HbA1c and insulin resistance eliminated the associations of BCAAs with CVD. CONCLUSIONS Circulating plasma BCAAs were positively associated with incident CVD in women. Impaired BCAA metabolism may capture the long-term risk of the common cause underlying T2D and CVD.
Collapse
Affiliation(s)
- Deirdre K Tobias
- Division of Preventive Medicine, Department of Medicine (D.K.T., P.H.H., O.V.D., P.M.R., J.E.M., S.C., S.M.), Center for Lipid Metabolomics (P.H.H., O.V.D., S.M.), Division of Cardiovascular Medicine (P.M.R., S.C., S.M.), Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA (J.E.M.); Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, ON, Canada (P.R.L.); Center for Clinical and Epidemiological Research, Hospital Universitario at University of Sao Paulo, Brazil (P.H.H.); and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (J.E.M.)
| | - Patrick R Lawler
- Division of Preventive Medicine, Department of Medicine (D.K.T., P.H.H., O.V.D., P.M.R., J.E.M., S.C., S.M.), Center for Lipid Metabolomics (P.H.H., O.V.D., S.M.), Division of Cardiovascular Medicine (P.M.R., S.C., S.M.), Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA (J.E.M.); Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, ON, Canada (P.R.L.); Center for Clinical and Epidemiological Research, Hospital Universitario at University of Sao Paulo, Brazil (P.H.H.); and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (J.E.M.)
| | - Paulo H Harada
- Division of Preventive Medicine, Department of Medicine (D.K.T., P.H.H., O.V.D., P.M.R., J.E.M., S.C., S.M.), Center for Lipid Metabolomics (P.H.H., O.V.D., S.M.), Division of Cardiovascular Medicine (P.M.R., S.C., S.M.), Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA (J.E.M.); Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, ON, Canada (P.R.L.); Center for Clinical and Epidemiological Research, Hospital Universitario at University of Sao Paulo, Brazil (P.H.H.); and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (J.E.M.)
| | - Olga V Demler
- Division of Preventive Medicine, Department of Medicine (D.K.T., P.H.H., O.V.D., P.M.R., J.E.M., S.C., S.M.), Center for Lipid Metabolomics (P.H.H., O.V.D., S.M.), Division of Cardiovascular Medicine (P.M.R., S.C., S.M.), Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA (J.E.M.); Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, ON, Canada (P.R.L.); Center for Clinical and Epidemiological Research, Hospital Universitario at University of Sao Paulo, Brazil (P.H.H.); and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (J.E.M.)
| | - Paul M Ridker
- Division of Preventive Medicine, Department of Medicine (D.K.T., P.H.H., O.V.D., P.M.R., J.E.M., S.C., S.M.), Center for Lipid Metabolomics (P.H.H., O.V.D., S.M.), Division of Cardiovascular Medicine (P.M.R., S.C., S.M.), Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA (J.E.M.); Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, ON, Canada (P.R.L.); Center for Clinical and Epidemiological Research, Hospital Universitario at University of Sao Paulo, Brazil (P.H.H.); and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (J.E.M.)
| | - JoAnn E Manson
- Division of Preventive Medicine, Department of Medicine (D.K.T., P.H.H., O.V.D., P.M.R., J.E.M., S.C., S.M.), Center for Lipid Metabolomics (P.H.H., O.V.D., S.M.), Division of Cardiovascular Medicine (P.M.R., S.C., S.M.), Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA (J.E.M.); Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, ON, Canada (P.R.L.); Center for Clinical and Epidemiological Research, Hospital Universitario at University of Sao Paulo, Brazil (P.H.H.); and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (J.E.M.)
| | - Susan Cheng
- Division of Preventive Medicine, Department of Medicine (D.K.T., P.H.H., O.V.D., P.M.R., J.E.M., S.C., S.M.), Center for Lipid Metabolomics (P.H.H., O.V.D., S.M.), Division of Cardiovascular Medicine (P.M.R., S.C., S.M.), Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA (J.E.M.); Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, ON, Canada (P.R.L.); Center for Clinical and Epidemiological Research, Hospital Universitario at University of Sao Paulo, Brazil (P.H.H.); and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (J.E.M.)
| | - Samia Mora
- Division of Preventive Medicine, Department of Medicine (D.K.T., P.H.H., O.V.D., P.M.R., J.E.M., S.C., S.M.), Center for Lipid Metabolomics (P.H.H., O.V.D., S.M.), Division of Cardiovascular Medicine (P.M.R., S.C., S.M.), Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA (J.E.M.); Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, ON, Canada (P.R.L.); Center for Clinical and Epidemiological Research, Hospital Universitario at University of Sao Paulo, Brazil (P.H.H.); and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (J.E.M.)
| |
Collapse
|
29
|
Yang L, Li Z, Song Y, Liu Y, Zhao H, Liu Y, Zhang T, Yuan Y, Cai X, Wang S, Wang P, Gao S, Li L, Li Y, Yu C. Study on urine metabolic profiling and pathogenesis of hyperlipidemia. Clin Chim Acta 2019; 495:365-373. [PMID: 31059703 DOI: 10.1016/j.cca.2019.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 04/14/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND As a recognized risk factor for cardiovascular disease (CVD), hyperlipidemia (HLP) has developed into a high incidence disease that seriously threatens human health. Finding a new target for effective treatment of HLP will be a powerful way to reduce the incidence of CVD. The purpose of this study was to find potential biomarkers in urine of HLP patients and analyze their metabolic pathways to study the pathogenesis of HLP. METHODS An UPLC-Q-TOF/MS technology was used to detect the metabolites in urine of 60 HLP patients and 60 normal controls. Based on PLS-DA pattern recognition, potential biomarkers related to HLP were screened out. RESULTS 22 potential biomarkers related to HLP were identified, which involved amino acid metabolism, fatty acid metabolism, nucleotide metabolism, steroid hormone metabolism and intestinal flora metabolism, and their possible pathogenesis was found to be related to inflammatory reaction and oxidative stress. CONCLUSION The non-targeted metabolomic method based on UPLC-Q-TOF/MS technology can effectively identify potential biomarkers in the urine of HLP patients and explore the possible pathogenesis. Our research will lay a foundation for finding new targets for the treatment of HLP and provide a basis for clinical research on the treatment of HLP.
Collapse
Affiliation(s)
- Liu Yang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Zhu Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Yanqi Song
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Yijia Liu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Huan Zhao
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Yuechen Liu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Tianpu Zhang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Yu Yuan
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Xuemeng Cai
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Shuo Wang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Pengwei Wang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Shan Gao
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China.
| | - Yubo Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China.
| | - Chunquan Yu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China.
| |
Collapse
|
30
|
Lau CHE, Siskos AP, Maitre L, Robinson O, Athersuch TJ, Want EJ, Urquiza J, Casas M, Vafeiadi M, Roumeliotaki T, McEachan RRC, Azad R, Haug LS, Meltzer HM, Andrusaityte S, Petraviciene I, Grazuleviciene R, Thomsen C, Wright J, Slama R, Chatzi L, Vrijheid M, Keun HC, Coen M. Determinants of the urinary and serum metabolome in children from six European populations. BMC Med 2018; 16:202. [PMID: 30404627 PMCID: PMC6223046 DOI: 10.1186/s12916-018-1190-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/10/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Environment and diet in early life can affect development and health throughout the life course. Metabolic phenotyping of urine and serum represents a complementary systems-wide approach to elucidate environment-health interactions. However, large-scale metabolome studies in children combining analyses of these biological fluids are lacking. Here, we sought to characterise the major determinants of the child metabolome and to define metabolite associations with age, sex, BMI and dietary habits in European children, by exploiting a unique biobank established as part of the Human Early-Life Exposome project ( http://www.projecthelix.eu ). METHODS Metabolic phenotypes of matched urine and serum samples from 1192 children (aged 6-11) recruited from birth cohorts in six European countries were measured using high-throughput 1H nuclear magnetic resonance (NMR) spectroscopy and a targeted LC-MS/MS metabolomic assay (Biocrates AbsoluteIDQ p180 kit). RESULTS We identified both urinary and serum creatinine to be positively associated with age. Metabolic associations to BMI z-score included a novel association with urinary 4-deoxyerythreonic acid in addition to valine, serum carnitine, short-chain acylcarnitines (C3, C5), glutamate, BCAAs, lysophosphatidylcholines (lysoPC a C14:0, lysoPC a C16:1, lysoPC a C18:1, lysoPC a C18:2) and sphingolipids (SM C16:0, SM C16:1, SM C18:1). Dietary-metabolite associations included urinary creatine and serum phosphatidylcholines (4) with meat intake, serum phosphatidylcholines (12) with fish, urinary hippurate with vegetables, and urinary proline betaine and hippurate with fruit intake. Population-specific variance (age, sex, BMI, ethnicity, dietary and country of origin) was better captured in the serum than in the urine profile; these factors explained a median of 9.0% variance amongst serum metabolites versus a median of 5.1% amongst urinary metabolites. Metabolic pathway correlations were identified, and concentrations of corresponding metabolites were significantly correlated (r > 0.18) between urine and serum. CONCLUSIONS We have established a pan-European reference metabolome for urine and serum of healthy children and gathered critical resources not previously available for future investigations into the influence of the metabolome on child health. The six European cohort populations studied share common metabolic associations with age, sex, BMI z-score and main dietary habits. Furthermore, we have identified a novel metabolic association between threonine catabolism and BMI of children.
Collapse
Affiliation(s)
- Chung-Ho E Lau
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK.
| | - Alexandros P Siskos
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK.,Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Léa Maitre
- ISGlobal, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiologa y Salud Pública (CIBERESP), Madrid, Spain
| | - Oliver Robinson
- MRC-PHE Centre for Environment and Health, School of Public Health, Faculty of Medicine, Imperial College London, London, W2 1PG, UK
| | - Toby J Athersuch
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK.,MRC-PHE Centre for Environment and Health, School of Public Health, Faculty of Medicine, Imperial College London, London, W2 1PG, UK
| | - Elizabeth J Want
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Jose Urquiza
- ISGlobal, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiologa y Salud Pública (CIBERESP), Madrid, Spain
| | - Maribel Casas
- ISGlobal, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiologa y Salud Pública (CIBERESP), Madrid, Spain
| | - Marina Vafeiadi
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Theano Roumeliotaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Rosemary R C McEachan
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Rafaq Azad
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Line S Haug
- Norwegian Institute of Public Health, Oslo, Norway
| | | | - Sandra Andrusaityte
- Department of Environmental Sciences, Vytautas Magnus University, Kaunas, Lithuania
| | - Inga Petraviciene
- Department of Environmental Sciences, Vytautas Magnus University, Kaunas, Lithuania
| | | | | | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Remy Slama
- Inserm, Univ. Grenoble Alpes, CNRS, IAB (Institute of Advanced Biosciences), Grenoble, France
| | - Leda Chatzi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiologa y Salud Pública (CIBERESP), Madrid, Spain
| | - Hector C Keun
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Muireann Coen
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK. .,Oncology Safety, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK.
| |
Collapse
|
31
|
Libert DM, Nowacki AS, Natowicz MR. Metabolomic analysis of obesity, metabolic syndrome, and type 2 diabetes: amino acid and acylcarnitine levels change along a spectrum of metabolic wellness. PeerJ 2018; 6:e5410. [PMID: 30186675 PMCID: PMC6120443 DOI: 10.7717/peerj.5410] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/18/2018] [Indexed: 12/12/2022] Open
Abstract
Background Metabolic syndrome (MS) is a construct used to separate “healthy” from “unhealthy” obese patients, and is a major risk factor for type 2 diabetes (T2D) and cardiovascular disease. There is controversy over whether obese “metabolically well” persons have a higher morbidity and mortality than lean counterparts, suggesting that MS criteria do not completely describe physiologic risk factors or consequences of obesity. We hypothesized that metabolomic analysis of plasma would distinguish obese individuals with and without MS and T2D along a spectrum of obesity-associated metabolic derangements, supporting metabolomic analysis as a tool for a more detailed assessment of metabolic wellness than currently used MS criteria. Methods Fasting plasma samples from 90 adults were assigned to groups based on BMI and ATP III criteria for MS: (1) lean metabolically well (LMW; n = 24); (2) obese metabolically well (OBMW; n = 26); (3) obese metabolically unwell (OBMUW; n = 20); and (4) obese metabolically unwell with T2D (OBDM; n = 20). Forty-one amino acids/dipeptides, 33 acylcarnitines and 21 ratios were measured. Obesity and T2D effects were analyzed by Wilcoxon rank-sum tests comparing obese nondiabetics vs LMW, and OBDM vs nondiabetics, respectively. Metabolic unwellness was analyzed by Jonckheere-Terpstra trend tests, assuming worsening health from LMW → OBMW → OBMUW. To adjust for multiple comparisons, statistical significance was set at p < 0.005. K-means cluster analysis of aggregated amino acid and acylcarnitine data was also performed. Results Analytes and ratios significantly increasing in obesity, T2D, and with worsening health include: branched-chain amino acids (BCAAs), cystine, alpha-aminoadipic acid, phenylalanine, leucine + lysine, and short-chain acylcarnitines/total carnitines. Tyrosine, alanine and propionylcarnitine increase with obesity and metabolic unwellness. Asparagine and the tryptophan/large neutral amino acid ratio decrease with T2D and metabolic unwellness. Malonylcarnitine decreases in obesity and 3-OHbutyrylcarnitine increases in T2D; neither correlates with unwellness. Cluster analysis did not separate subjects into discreet groups based on metabolic wellness. Discussion Levels of 15 species and metabolite ratios trend significantly with worsening metabolic health; some are newly recognized. BCAAs, aromatic amino acids, lysine, and its metabolite, alpha-aminoadipate, increase with worsening health. The lysine pathway is distinct from BCAA metabolism, indicating that biochemical derangements associated with MS involve pathways besides those affected by BCAAs. Even those considered “obese, metabolically well” had metabolite levels which significantly trended towards those found in obese diabetics. Overall, this analysis yields a more granular view of metabolic wellness than the sole use of cardiometabolic MS parameters. This, in turn, suggests the possible utility of plasma metabolomic analysis for research and public health applications.
Collapse
Affiliation(s)
- Diane M Libert
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Amy S Nowacki
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America.,Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, United States of America
| | - Marvin R Natowicz
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America.,Pathology and Laboratory Medicine, Genomic Medicine, Pediatrics and Neurological Institutes, Cleveland Clinic, Cleveland, OH, United States of America
| |
Collapse
|
32
|
Huang J, Weinstein SJ, Moore SC, Derkach A, Hua X, Liao LM, Gu F, Mondul AM, Sampson JN, Albanes D. Serum Metabolomic Profiling of All-Cause Mortality: A Prospective Analysis in the Alpha-Tocopherol, Beta-Carotene Cancer Prevention (ATBC) Study Cohort. Am J Epidemiol 2018; 187:1721-1732. [PMID: 29390044 DOI: 10.1093/aje/kwy017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022] Open
Abstract
Tobacco use, hypertension, hyperglycemia, overweight, and inactivity are leading causes of overall and cardiovascular disease (CVD) mortality worldwide, yet the relevant metabolic alterations responsible are largely unknown. We conducted a serum metabolomic analysis of 620 men in the Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study (1985-2013). During 28 years of follow-up, there were 435 deaths (197 CVD and 107 cancer). The analysis included 406 known metabolites measured with ultra-high-performance liquid chromatography/mass spectrometry-gas chromatography/mass spectrometry. We used Cox regression to estimate mortality hazard ratios for a 1-standard-deviation difference in metabolite signals. The strongest associations with overall mortality were N-acetylvaline (hazard ratio (HR) = 1.28; P < 4.1 × 10-5, below Bonferroni statistical threshold) and dimethylglycine, 7-methylguanine, C-glycosyltryptophan, taurocholate, and N-acetyltryptophan (1.23 ≤ HR ≤ 1.32; 5 × 10-5 ≤ P ≤ 1 × 10-4). C-Glycosyltryptophan, 7-methylguanine, and 4-androsten-3β,17β-diol disulfate were statistically significantly associated with CVD mortality (1.49 ≤ HR ≤ 1.62, P < 4.1 × 10-5). No metabolite was associated with cancer mortality, at a false discovery rate of <0.1. Individuals with a 1-standard-deviation higher metabolite risk score had increased all-cause and CVD mortality in the test set (HR = 1.4, P = 0.05; HR = 1.8, P = 0.003, respectively). The several serum metabolites and their composite risk score independently associated with all-cause and CVD mortality may provide potential leads regarding the molecular basis of mortality.
Collapse
Affiliation(s)
- Jiaqi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Andriy Derkach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Xing Hua
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Linda M Liao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Fangyi Gu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
| | - Alison M Mondul
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
33
|
Grajeda-Iglesias C, Aviram M. Specific Amino Acids Affect Cardiovascular Diseases and Atherogenesis via Protection against Macrophage Foam Cell Formation: Review Article. Rambam Maimonides Med J 2018; 9:RMMJ.10337. [PMID: 29944113 PMCID: PMC6115485 DOI: 10.5041/rmmj.10337] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The strong relationship between cardiovascular diseases (CVD), atherosclerosis, and endogenous or exogenous lipids has been recognized for decades, underestimating the contribution of other dietary components, such as amino acids, to the initiation of the underlying inflammatory disease. Recently, specific amino acids have been associated with incident cardiovascular disorders, suggesting their significant role in the pathogenesis of CVD. Special attention has been paid to the group of branched-chain amino acids (BCAA), leucine, isoleucine, and valine, since their plasma values are frequently found in high concentrations in individuals with CVD risk. Nevertheless, dietary BCAA, leucine in particular, have been associated with improved indicators of atherosclerosis. Therefore, their potential role in the process of atherogenesis and concomitant CVD development remains unclear. Macrophages play pivotal roles in the development of atherosclerosis. They can accumulate high amounts of circulating lipids, through a process known as macrophage foam cell formation, and initiate the atherogenesis process. We have recently screened for anti- or pro-atherogenic amino acids in the macrophage model system. Our study showed that glycine, cysteine, alanine, leucine, glutamate, and glutamine significantly affected macrophage atherogenicity mainly through modulation of the cellular triglyceride metabolism. The anti-atherogenic properties of glycine and leucine, and the pro-atherogenic effects of glutamine, were also confirmed in vivo. Further investigation is warranted to define the role of these amino acids in atherosclerosis and CVD, which may serve as a basis for the development of anti-atherogenic nutritional and therapeutic approaches.
Collapse
|
34
|
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
|
35
|
Enko D, Wagner H, Kriegshäuser G, Brandmayr W, Halwachs-Baumann G, Schnedl WJ, Zelzer S, Mangge H, Meinitzer A. Assessment of tryptophan metabolism and signs of depression in individuals with carbohydrate malabsorption. Psychiatry Res 2018; 262:595-599. [PMID: 28965810 DOI: 10.1016/j.psychres.2017.09.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/17/2017] [Accepted: 09/21/2017] [Indexed: 01/16/2023]
Abstract
This prospective cross-sectional study aimed to investigate the potential association between primary-adult lactose malabsorption, fructose malabsorption, tryptophan (TRP) metabolism and the presence of depressive signs. Overall 251 patients, who were referred for lactase gene C/T-13910 polymorphism genotyping and fructose hydrogen/methane breath testing, were included. All participants filled out the Beck Depression Inventory (BDI II). Serum concentrations of tryptophan (TRP), kynurenine (KYN), kynuric acid (KYNA), and TRP competing amino acids (leucine, isoleucine, valine, phenylalanine, tyrosine) were measured by high-pressure liquid-chromatography. Logistic regression analysis was performed with lactose malabsorption, fructose malabsorption and all potential biomarkers of TRP metabolism to assess the effect on signs of depression, defined as a BDI II score > 13. Primary-adult lactose malabsorption and fructose malabsorption was detected in 65 (25.90%) and 65 (25.90%) patients, respectively. Fructose malabsorption was significantly associated with BDI II score, whereas no such relationship was found for lactose malabsorption. Serum levels of TRP and TRP metabolites were no predictors of depression. The authors suggest to conduct further prospective longitudinal studies in order to get further insight of associations between carbohydrate malabsorption, biomarkers and mood disorders.
Collapse
Affiliation(s)
- Dietmar Enko
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Steyr, Steyr, Austria; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.
| | - Helga Wagner
- Department of Applied Statistics, Johannes Kepler University Linz, Linz, Austria.
| | - Gernot Kriegshäuser
- Institute of Clinical Chemistry and Laboratory Medicine, General Hospital Steyr, Steyr, Austria; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.
| | - Wolfgang Brandmayr
- Department of Psychiatry and Psychotherapeutic Medicine, General Hospital Steyr, Steyr, Austria.
| | | | | | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.
| |
Collapse
|
36
|
Affiliation(s)
- Claudia Grajeda-Iglesias
- Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel, Institute of Technology, Haifa, Israel
| | - Oren Rom
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Michael Aviram
- Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel, Institute of Technology, Haifa, Israel
| |
Collapse
|
37
|
Guevara-Cruz M, Vargas-Morales JM, Méndez-García AL, López-Barradas AM, Granados-Portillo O, Ordaz-Nava G, Rocha-Viggiano AK, Gutierrez-Leyte CA, Medina-Cerda E, Rosado JL, Morales JC, Torres N, Tovar AR, Noriega LG. Amino acid profiles of young adults differ by sex, body mass index and insulin resistance. Nutr Metab Cardiovasc Dis 2018; 28:393-401. [PMID: 29422298 DOI: 10.1016/j.numecd.2018.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/29/2017] [Accepted: 01/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS An increase in plasma branched-chain amino acids is associated with a higher risk of developing type 2 diabetes and cardiovascular diseases. However, little is known about the basal plasma amino acid concentrations in young adults. Our aim was to determine the plasma amino acid profiles of young adults and to evaluate how these profiles were modified by sex, body mass index (BMI) and insulin resistance (IR). METHODS AND RESULTS We performed a transversal study with 608 Mexican young adults aged 19.9 ± 2.4 years who were applicants to the Universidad Autónoma de San Luis Potosí. The subjects underwent a physical examination and provided a clinical history and a blood sample for biochemical, hormonal and amino acid analyses. The women had higher levels of arginine, aspartate and serine and lower levels of α-aminoadipic acid, cysteine, isoleucine, leucine, methionine, proline, tryptophan, tyrosine, urea and valine than the men. The obese subjects had higher levels of alanine, aspartate, cysteine, ornithine, phenylalanine, proline and tyrosine and lower levels of glycine, ornithine and serine than the normal weight subjects. Subjects with IR (defined as HOMA > 2.5) had higher levels of arginine, alanine, aspartate, isoleucine, leucine, phenylalanine, proline, tyrosine, taurine and valine than the subjects without IR. Furthermore, we identified two main groups in the subjects with obesity and/or IR; one group was composed of amino acids that positively correlated with the clinical, biochemical and hormonal parameters, whereas the second group exhibited negative correlations. CONCLUSION This study demonstrates that young adults with obesity or IR have altered amino acid profiles characterized by an increase in alanine, aspartate, proline and tyrosine and a decrease in glycine.
Collapse
Affiliation(s)
- M Guevara-Cruz
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - J M Vargas-Morales
- Facultad de Ciencias Química, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - A L Méndez-García
- Facultad de Enfermería, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - A M López-Barradas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - O Granados-Portillo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - G Ordaz-Nava
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - A K Rocha-Viggiano
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - C A Gutierrez-Leyte
- Departamento de Ciencia y Tecnología de Alimentos, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - E Medina-Cerda
- Centro de Salud Universitario, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - J L Rosado
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Juriquilla, Querétaro, Mexico
| | - J C Morales
- Departamento de Ciencia y Tecnología de Alimentos, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - N Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - A R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico.
| | - L G Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico.
| |
Collapse
|
38
|
Nie C, He T, Zhang W, Zhang G, Ma X. Branched Chain Amino Acids: Beyond Nutrition Metabolism. Int J Mol Sci 2018; 19:E954. [PMID: 29570613 PMCID: PMC5979320 DOI: 10.3390/ijms19040954] [Citation(s) in RCA: 462] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/01/2018] [Accepted: 03/14/2018] [Indexed: 12/14/2022] Open
Abstract
Branched chain amino acids (BCAAs), including leucine (Leu), isoleucine (Ile), and valine (Val), play critical roles in the regulation of energy homeostasis, nutrition metabolism, gut health, immunity and disease in humans and animals. As the most abundant of essential amino acids (EAAs), BCAAs are not only the substrates for synthesis of nitrogenous compounds, they also serve as signaling molecules regulating metabolism of glucose, lipid, and protein synthesis, intestinal health, and immunity via special signaling network, especially phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signal pathway. Current evidence supports BCAAs and their derivatives as the potential biomarkers of diseases such as insulin resistance (IR), type 2 diabetes mellitus (T2DM), cancer, and cardiovascular diseases (CVDs). These diseases are closely associated with catabolism and balance of BCAAs. Hence, optimizing dietary BCAA levels should have a positive effect on the parameters associated with health and diseases. This review focuses on recent findings of BCAAs in metabolic pathways and regulation, and underlying the relationship of BCAAs to related disease processes.
Collapse
Affiliation(s)
- Cunxi Nie
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing 100193, China.
- College of Animal Science and Technology, Shihezi University, No. 221. Beisi Road, Shihezi, Xinjiang 832003, China.
| | - Ting He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing 100193, China.
| | - Wenju Zhang
- College of Animal Science and Technology, Shihezi University, No. 221. Beisi Road, Shihezi, Xinjiang 832003, China.
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA.
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing 100193, China.
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
39
|
Barceló A, Bauça JM, Peña-Zarza JA, Morell-Garcia D, Yáñez A, Pérez G, Piérola J, Toledo N, de la Peña M. Circulating branched-chain amino acids in children with obstructive sleep apnea. Pediatr Pulmonol 2017; 52:1085-1091. [PMID: 28672086 DOI: 10.1002/ppul.23753] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/02/2017] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The effects of obstructive sleep apnea (OSA) on the metabolic system are not well understood, especially in children. Recent studies have provided evidence of the modulation of insulin action by branched-chain amino acids (BCAAs) and suggested novel mechanistic relationships between glucose and amino acid metabolic pathways. We hypothesized that plasma BCAA levels may serve as biomarkers of insulin resistance and metabolic dysfunction in children with OSA. METHODS A polysomnography was conducted for the diagnosis of OSA in 90 snoring children, in a tertiary hospital. Anthropometric and clinical data were measured and venous blood samples were collected for the measurement of plasma glucose, insulin, HbA1c, and amino acids. RESULTS Children with OSA had significantly higher levels of BCAAs (leucine, isoleucine, and total BCAAs) compared with those without OSA (P = 0.024). A positive significant correlation was observed between insulin levels and both leucine and isoleucine (r = 0.232; P < 0.05). On multivariate regression analyses, the presence of OSA was significantly associated with leucine, isoleucine, and total BCAA concentrations (P = 0.028), whereas the arousal index was associated with leucine, valine, and total BCAA levels (P = 0.037). CONCLUSIONS The presence of OSA and sleep fragmentation may induce changes in branched-chain amino acid metabolism in snoring children, independently of obesity. These data may suggest a new mechanism linking OSA and glucose homeostasis.
Collapse
Affiliation(s)
- Antonia Barceló
- Servei Anàlisis Clíniques, Hospital Universitari Son Espases, Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain.,CIBER Enfermedades Respiratorias (CibeRes) (CB06/06), Spain
| | - Josep Miquel Bauça
- Servei Anàlisis Clíniques, Hospital Universitari Son Espases, Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
| | - José Antonio Peña-Zarza
- Sleep Unit, Department of Pediatrics, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Daniel Morell-Garcia
- Servei Anàlisis Clíniques, Hospital Universitari Son Espases, Palma de Mallorca, Spain.,Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
| | - Aina Yáñez
- Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
| | - Gerardo Pérez
- Servei Anàlisis Clíniques, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Javier Piérola
- Institut d'Investigació Sanitària de Palma (IdISPa), Palma de Mallorca, Spain
| | - Nuria Toledo
- Sleep Unit, Department of Respiratory Medicine, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Mónica de la Peña
- Sleep Unit, Department of Respiratory Medicine, Hospital Universitari Son Espases, Palma de Mallorca, Spain
| |
Collapse
|
40
|
Pallottini AC, Sales CH, Vieira DADS, Marchioni DM, Fisberg RM. Dietary BCAA Intake Is Associated with Demographic, Socioeconomic and Lifestyle Factors in Residents of São Paulo, Brazil. Nutrients 2017; 9:E449. [PMID: 28468321 PMCID: PMC5452179 DOI: 10.3390/nu9050449] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/21/2017] [Accepted: 04/26/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Identifying which risk groups have a higher intake of branched chain amino acids (BCAA) is important for the planning of public policies. This study was undertaken to investigate BCAA consumption, the foods contributing to that consumption and their association with demographic, socioeconomic and lifestyle factors. METHODS Data from the Health Survey of São Paulo, a cross-sectional population-based survey (n = 1662; age range 12-97 years), were used. Dietary intake was measured using 24-h dietary recalls. Baseline characteristics were collected. Associations between BCAA intake and demographic, socioeconomic and lifestyle factors were determined using linear regression. RESULTS Total BCAA intake was 217.14 mg/kg·day (Leu: 97.16 mg/kg·day; Ile: 56.44 mg/kg·day; Val: 63.54 mg/kg·day). BCAA intake was negatively associated with female sex in adolescents and adult groups, with no white race in adolescents, and with former smoker status in adults. Conversely, BCAA was positively associated with household per capita income in adolescents and adults. No associations were observed in the older adults group. Main food contributors to BCAA were unprocessed red meat, unprocessed poultry, bread and toast, beans and rice. CONCLUSIONS Adolescents and adults were the most vulnerable to having their BCCA intake influenced by demographic, socioeconomic and lifestyle factors.
Collapse
Affiliation(s)
- Ana Carolina Pallottini
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo CEP 01246-904, Brazil.
| | - Cristiane Hermes Sales
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo CEP 01246-904, Brazil.
| | - Diva Aliete Dos Santos Vieira
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo CEP 01246-904, Brazil.
| | - Dirce Maria Marchioni
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo CEP 01246-904, Brazil.
| | - Regina Mara Fisberg
- Department of Nutrition, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Cerqueira César, São Paulo CEP 01246-904, Brazil.
| |
Collapse
|
41
|
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
|
42
|
Yamaguchi N, Mahbub MH, Takahashi H, Hase R, Ishimaru Y, Sunagawa H, Amano H, Kobayashi-Miura M, Kanda H, Fujita Y, Yamamoto H, Yamamoto M, Kikuchi S, Ikeda A, Takasu M, Kageyama N, Nakamura M, Tanabe T. Plasma free amino acid profiles evaluate risk of metabolic syndrome, diabetes, dyslipidemia, and hypertension in a large Asian population. Environ Health Prev Med 2017; 22:35. [PMID: 29165132 PMCID: PMC5664911 DOI: 10.1186/s12199-017-0642-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/01/2017] [Indexed: 01/04/2023] Open
Abstract
Background Recently, the association of plasma free amino acid (PFAA) profile and lifestyle-related diseases has been reported. However, few studies have been reported in large Asian populations, about the usefulness of PFAAs for evaluating disease risks. We examined the ability of PFAA profiles to evaluate lifestyle-related diseases in so far the largest Asian population. Methods We examined plasma concentrations of 19 amino acids in 8589 Japanese subjects, and determined the association with variables associated with obesity, blood glucose, lipid, and blood pressure. We also evaluated the PFAA indexes that reflect visceral fat obesity and insulin resistance. The contribution of single PFAA level and relevant PFAA indexes was also examined in the risk assessment of lifestyle-related diseases. Results Of the 19 amino acids, branched-chain amino acids and aromatic amino acids showed association with obesity and lipid variables. The PFAA index related to visceral fat obesity showed relatively higher correlation with variables than that of any PFAA. In the evaluation of lifestyle-related disease risks, the odds ratios of the PFAA index related to visceral fat obesity or insulin resistance with the diseases were higher than most of those of individual amino acid levels even after adjusting for potential confounding factors. The association pattern of the indexes and PFAA with each lifestyle-related disease was distinct. Conclusions We confirmed the usefulness of PFAA profiles and indexes as markers for evaluating the risks of lifestyle-related diseases, including diabetes mellitus, metabolic syndrome, dyslipidemia, and hypertension in a large Asian population.
Collapse
Affiliation(s)
- Natsu Yamaguchi
- Department of Public Health and Preventive Medicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - M H Mahbub
- Department of Public Health and Preventive Medicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hidekazu Takahashi
- Department of Public Health and Preventive Medicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ryosuke Hase
- Department of Public Health and Preventive Medicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yasutaka Ishimaru
- Department of Public Health and Preventive Medicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hiroshi Sunagawa
- Department of Public Health and Preventive Medicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hiroki Amano
- Division of Health Administration and Promotion, Graduate School of Medicine, Tottori University, Yonago, Japan
| | | | - Hideyuki Kanda
- Department of Environmental Medicine and Public Health, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Yasuyuki Fujita
- Department of Environmental Medicine and Public Health, Faculty of Medicine, Shimane University, Izumo, Japan
| | | | - Mai Yamamoto
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Shinya Kikuchi
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Atsuko Ikeda
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Mariko Takasu
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Naoko Kageyama
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Mina Nakamura
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Tsuyoshi Tanabe
- Department of Public Health and Preventive Medicine, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|