1
|
Li C, Yang Q, Zhang L. Identification of putative allosteric inhibitors of BCKDK via virtual screening and biological evaluation. J Enzyme Inhib Med Chem 2024; 39:2290458. [PMID: 38059302 DOI: 10.1080/14756366.2023.2290458] [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: 06/24/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023] Open
Abstract
Abnormal accumulation of branched-chain amino acids (BCAAs) can lead to metabolic diseases and cancers. Branched-chain α-keto acid dehydrogenase kinase (BCKDK) is a key negative regulator of BCAA catabolism, and targeting BCKDK provides a promising therapeutic approach for diseases caused by BCAA accumulation. Here, we screened PPHN and POAB as novel putative allosteric inhibitors by integrating allosteric binding site prediction, large-scale ligand database virtual screening, and bioactivity evaluation assays. Both of them showed a high binding affinity to BCKDK, with Kd values of 3.9 μM and 1.86 μM, respectively. In vivo experiments, the inhibitors demonstrated superior kinase inhibitory activity and notable antiproliferative and proapoptotic effects on diverse cancer cells. Finally, bulk RNA-seq analysis revealed that PPHN and POAB suppressed cell growth through a range of signalling pathways. Taken together, our findings highlight two novel BCKDK inhibitors as potent therapeutic candidates for metabolic diseases and cancers associated with BCAA dysfunctional metabolism.
Collapse
Affiliation(s)
- Chunqiong Li
- Genomics Center, Chinese Institute for Brain Research, Beijing, China
| | - Quanjun Yang
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Zhang
- Genomics Center, Chinese Institute for Brain Research, Beijing, China
| |
Collapse
|
2
|
Manninen S, Tilles-Tirkkonen T, Aittola K, Männikkö R, Karhunen L, Kolehmainen M, Schwab U, Lindström J, Lakka T, Pihlajamäki J. Associations of Lifestyle Patterns with Glucose and Lipid Metabolism in Finnish Adults at Increased Risk of Type 2 Diabetes. Mol Nutr Food Res 2024; 68:e2300338. [PMID: 38308150 DOI: 10.1002/mnfr.202300338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/18/2023] [Indexed: 02/04/2024]
Abstract
SCOPE Various lifestyle and sociodemographic factors have been associated with risk factors for type 2 diabetes (T2D). However, their combined associations with T2D risk factors have been studied much less. MATERIALS AND RESULTS This study investigates cross-sectional associations of lifestyle patterns with T2D risk factors among 2925 adults at increased risk participating in the Stop Diabetes study. Lifestyle patterns are determined using principal component analysis (PCA) with several lifestyle and sociodemographic factors. The associations of lifestyle patterns with measures of glucose and lipid metabolism and serum metabolites analyzed by nuclear magnetic resonance (NMR) spectroscopy are studied using linear regression analysis. "Healthy eating" pattern is associated with better glucose and insulin metabolism, more favorable lipoprotein and fatty acid profiles and lower serum concentrations of metabolites related to inflammation, insulin resistance, and T2D. "High socioeconomic status and low physical activity" pattern is associated with increased serum concentrations of branched-chain amino acids, as are "Meat and poultry" and "Sleeping hours" patterns. "Snacks" pattern is associated with lower serum concentrations of ketone bodies. CONCLUSIONS Our results show, in large scale primary care setting, that healthy eating is associated with better glucose and lipid metabolism and reveal novel associations of lifestyle patterns with metabolites related to glucose metabolism.
Collapse
Affiliation(s)
- Suvi Manninen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, 70211, Finland
| | - Tanja Tilles-Tirkkonen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, 70211, Finland
| | - Kirsikka Aittola
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, 70211, Finland
| | - Reija Männikkö
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, 70211, Finland
| | - Leila Karhunen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, 70211, Finland
| | - Marjukka Kolehmainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, 70211, Finland
| | - Ursula Schwab
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, 70211, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, KYS, 70029, Finland
| | - Jaana Lindström
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Timo Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, 70211, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, KYS, 70029, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, 70100, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, 70211, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, KYS, 70029, Finland
| |
Collapse
|
3
|
Torki SA, Bahadori E, Aghakhaninejad Z, Mohseni GK, Tajadod S, Rajabi Harsini A, Azaryan F, Saeedirad Z, Askarpour SA, Mahmoudi Z, Khoshdooz S, Bahar B, Shafaei H, Mosavi Jarrahi SA, Doaei S, Nazemi S, Gholamalizadeh M. Association between type 2 diabetes and branched chain amino acids (BCAA); a case-control study. J Diabetes Metab Disord 2023; 22:1291-1297. [PMID: 37975111 PMCID: PMC10638320 DOI: 10.1007/s40200-023-01247-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/03/2023] [Indexed: 11/19/2023]
Abstract
Background Several amino acids and their derivatives have been implicated in insulin resistance (IR) and Type 2 Diabetes Mellitus (T2DM). This research sought to establish a relationship between the dietary levels of branched-chain amino acids (BCAA) and the risk of T2DM. Methods This case-control study was carried out on 4200 participants consisting of 589 people with T2DM and 3611 non-diabetic aged 35 to 70 years residents in Sabzevar, Iran. Data on the economic-social, employment status, medical history, lifestyle, and sleep habits were collected via interview. The food frequency questionnaire (FFQ) was used to check the nutritional status. Participants' dietary BCAA consumption was estimated using Nutritionist IV software. Results A significant negative association between the incidence of T2DM and the dietary levels of BCAAs after adjustment for age and sex (OR = 0.972, CI 95%:0.648-0.996, P = 0.022). The negative association remained significant after additional adjustments for body mass index (BMI) and physical activity (OR = 0.967, CI 95%: 0.943-0.992, P = 0.010). Interestingly, a positive association was found between T2DM and total BCAAs (OR = 1.067, CI 95%: 1.017-1.119, P = 0.008), Isoleucine (OR = 1.248, CI 95%: 1.043-1.494, P = 0.016), Leucine (OR = 1.165, CI 95%: 1.046-1.299, P = 0.006) and Valine (OR = 1.274, CI 95%: 1.088-1.492, P = 0.003) after further adjustment for calorie intake. Conclusions Our results demonstrate branched-chain amino acids (BCAAs) including isoleucine, leucine, and valine are negatively associated with the incidence of type 2 diabetes (T2DM) after adjusting for age and sex, BMI, and physical activity. However, adjusting for calorie intake reversed the association between T2DM and BCAAs. These findings suggest that the association between BCAAs and T2DM may be influenced by calorie intake. Future longitudinal studies are warranted. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-023-01247-9.
Collapse
Affiliation(s)
- Saheb Abbas Torki
- Department of Nutrition, Faculty of Nutrition Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Effat Bahadori
- Department of Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zohreh Aghakhaninejad
- Department of Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Golsa Khalatbari Mohseni
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Tajadod
- Department of Nutrition, School of Public Health, International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Asma Rajabi Harsini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Azaryan
- Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Zahra Saeedirad
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Askarpour
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Mahmoudi
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Bojlul Bahar
- Nutrition Sciences and Applied Food Safety Studies, Research Centre for Global Development, School of Sport and Health Sciences, University of Central Lancashire, Preston, UK
| | - Hanieh Shafaei
- Nursing and Midwifery School, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Saeid Doaei
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samad Nazemi
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Sabzevar University of Medical Science, Sabzevar, Iran
| | - Maryam Gholamalizadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Jin A, Kan Z, Tan Q, Shao J, Han Q, Chang Y, An N, Yi M. Supplementation with food-derived oligopeptides promotes lipid metabolism in young male cyclists: a randomized controlled crossover trial. J Int Soc Sports Nutr 2023; 20:2254741. [PMID: 37674290 PMCID: PMC10486287 DOI: 10.1080/15502783.2023.2254741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Accumulation of body fat and dyslipidemia are associated with the development of obesity and cardiometabolic diseases. Moreover, the degree to which lipids can be metabolized has been cited as a determinant of cardiometabolic health and prolonged endurance capacity. In the backdrop of increasing obesity and cardiometabolic diseases, lipid metabolism and its modulation by physical activity, dietary adjustments, and supplementation play a significant role in maintaining health and endurance. Food-derived oligopeptides, such as rice and soybean peptides, have been shown to directly regulate abnormal lipid metabolism or promote hypolipidemia and fat oxidation in cell culture models, animal models, and human studies. However, whether supplementation with oligopeptides derived from multiple food sources can promote lipid degradation and fat oxidation in athletes remains unclear. Therefore, in a randomized controlled crossover trial, we investigated the impact of food-derived oligopeptide supplementation before and during exercise on lipid metabolism in young male cyclists. METHODS Sixteen young male cyclists (age: 17.0 ± 1.0 years; height: 178.4 ± 6.9 cm; body mass: 68.7 ± 12.7 kg, body mass index: 21.5 ± 3.4 kg/m2; maximum oxygen uptake: 56.3 ± 5.8 mL/min/kg) participated in this randomized controlled crossover trial. Each participant drank two beverages, one containing a blend of three food-derived oligopeptides (treatment, 0.5 g/kg body weight in total) and the other without (control), with a 2-week washout period between two experiments. The cyclists completed a one-day pattern protocol that consisted of intraday fasting, 30 min of sitting still, 85 min of prolonged exercise plus a 5-min sprint (PE), a short recovery period of 60 min, a 20-min time trial (TT), and recovery till next morning. Blood samples were collected for biochemical analyses of serum lipids and other biomarkers. We analyzed plasma triglyceride species (TGs), free amino acids (FAAs), and tricarboxylic acid (TCA) cycle intermediates using omics methods. In addition, exhaled gas was collected to assess the fat oxidation rate. RESULTS Five of 20 plasma FAAs were elevated pre-exercise (pre-Ex) only 20 min after oligopeptide ingestion, and most FAAs were markedly increased post PE and TT. Serum levels of TG and non-esterified fatty acids were lower in the experimental condition than in the control condition at the post PE and TT assessments, respectively. Further, the omics analysis of plasma TGs for the experimental condition demonstrated that most TGs were lower post PE and at the next fasting when compared with control levels. Simultaneously, the fat oxidation rate began to increase only 20 min after ingestion and during the preceding 85 min of PE. Levels of TCA cycle intermediates did not differ between the conditions. CONCLUSIONS The study noted that continuous ingestion of food-derived oligopeptides accelerated total body triglyceride breakdown, non-esterified fatty acid uptake, and fat oxidation during both sedentary and exercise states. Elevated circulating and intracellular FAA flux may modulate the selection of substrates for metabolic pathways in conjunction with the release of neuroendocrinological factors that slow down carbohydrate metabolism via acetyl coenzyme A feedback inhibition. This may increase the availability of fatty acids for energy production, with FAAs supplying more substrates for the TCA cycle. The findings of this study provide novel insight into strategies for promoting lipid metabolism in populations with dyslipidemia-related metabolic disorders such as obesity and for improving physiological functioning during endurance training. However, the absence of a non-exercising control group and verification of long-term supplementation effects was a limitation. Future studies will emphasize the impacts of whole protein supplementation as a control and of combined food-derived peptides or oligopeptides with probiotics and healthy food components on lipid metabolism in individuals who exercise.
Collapse
Affiliation(s)
- Aina Jin
- Beijing Sport University, Exercise Biochemistry, Beijing, China
- National Institute of Sports Medicine, Center for Sports Nutrition, Beijing, China
| | - Zhaobo Kan
- National Institute of Sports Medicine, Center for Sports Nutrition, Beijing, China
| | - Qiushi Tan
- National Institute of Sports Medicine, Center for Sports Nutrition, Beijing, China
| | - Jing Shao
- National Institute of Sports Medicine, Center for Sports Nutrition, Beijing, China
| | - Qi Han
- National Institute of Sports Medicine, Center for Sports Nutrition, Beijing, China
| | - Yashan Chang
- National Institute of Sports Medicine, Center for Sports Nutrition, Beijing, China
| | - Nan An
- National Institute of Sports Medicine, Center for Sports Nutrition, Beijing, China
| | - Muqing Yi
- National Institute of Sports Medicine, Center for Sports Nutrition, Beijing, China
| |
Collapse
|
5
|
Huang J, Li J, Ning Y, Ren Y, Shao Y, Zhang H, Zong X, Shi H. Enhancement of PPARα-Inhibited Leucine Metabolism-Stimulated β-Casein Synthesis and Fatty Acid Synthesis in Primary Bovine Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16184-16193. [PMID: 37853551 DOI: 10.1021/acs.jafc.3c00712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Leucine, a kind of branched-chain amino acid, plays a regulatory role in the milk production of mammalian mammary glands, but its regulatory functions and underlying molecular mechanisms remain unknown. This work showed that a leucine-enriched mixture (LEUem) supplementation increased the levels of milk protein and milk fat synthesis in primary bovine mammary epithelial cells (BMECs). RNA-seq of leucine-treated BMECs indicated alterations in lipid metabolism, translation, ribosomal structure and biogenesis, and inflammatory response signaling pathways. Meanwhile, the supplementation of leucine resulted in mTOR activation and increased the expression of BCKDHA, FASN, ACC, and SCD1. Interestingly, the expression of PPARα was independently correlated with the leucine-supplemented dose. PPARα activated by WY-14643 caused significant suppression of lipogenic genes expression. Furthermore, WY-14643 attenuated leucine-induced β-casein synthesis and enhanced the level of BCKDHA expression. Moreover, promoter analysis revealed a peroxisome-proliferator-response element (PPRE) site in the bovine BCKDHA promoter, and WY-14643 promoted the recruitment of PPARα onto the BCKDHA promoter. Together, the present data indicate that leucine promotes the synthesis of β-casein and fatty acid and that PPARα-involved leucine catabolism is the key target.
Collapse
Affiliation(s)
- Jiangtao Huang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jintao Li
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yong Ning
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yalun Ren
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yuexin Shao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Huawen Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xueyang Zong
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Huaiping Shi
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
6
|
Han S, Wu Q, Wang M, Yang M, Sun C, Liang J, Guo X, Zhang Z, Xu J, Qiu X, Xie C, Chen S, Gao Y, Meng ZX. An integrative profiling of metabolome and transcriptome in the plasma and skeletal muscle following an exercise intervention in diet-induced obese mice. J Mol Cell Biol 2023; 15:mjad016. [PMID: 36882217 PMCID: PMC10576543 DOI: 10.1093/jmcb/mjad016] [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: 10/10/2022] [Revised: 02/02/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023] Open
Abstract
Exercise intervention at the early stage of type 2 diabetes mellitus (T2DM) can aid in the maintenance of blood glucose homeostasis and prevent the development of macrovascular and microvascular complications. However, the exercise-regulated pathways that prevent the development of T2DM remain largely unclear. In this study, two forms of exercise intervention, treadmill training and voluntary wheel running, were conducted for high-fat diet (HFD)-induced obese mice. We observed that both forms of exercise intervention alleviated HFD-induced insulin resistance and glucose intolerance. Skeletal muscle is recognized as the primary site for postprandial glucose uptake and for responsive alteration beyond exercise training. Metabolomic profiling of the plasma and skeletal muscle in Chow, HFD, and HFD-exercise groups revealed robust alterations in metabolic pathways by exercise intervention in both cases. Overlapping analysis identified nine metabolites, including beta-alanine, leucine, valine, and tryptophan, which were reversed by exercise treatment in both the plasma and skeletal muscle. Transcriptomic analysis of gene expression profiles in the skeletal muscle revealed several key pathways involved in the beneficial effects of exercise on metabolic homeostasis. In addition, integrative transcriptomic and metabolomic analyses uncovered strong correlations between the concentrations of bioactive metabolites and the expression levels of genes involved in energy metabolism, insulin sensitivity, and immune response in the skeletal muscle. This work established two models of exercise intervention in obese mice and provided mechanistic insights into the beneficial effects of exercise intervention on systemic energy homeostasis.
Collapse
Affiliation(s)
- Shuang Han
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Qingqian Wu
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mengying Wang
- Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Miqi Yang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chen Sun
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaqi Liang
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medical, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zheyu Zhang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingya Xu
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xinyuan Qiu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha 410073, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medical, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yue Gao
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
7
|
Doestzada M, Zhernakova DV, C L van den Munckhof I, Wang D, Kurilshikov A, Chen L, Bloks VW, van Faassen M, Rutten JHW, Joosten LAB, Netea MG, Wijmenga C, Riksen NP, Zhernakova A, Kuipers F, Fu J. Systematic analysis of relationships between plasma branched-chain amino acid concentrations and cardiometabolic parameters: an association and Mendelian randomization study. BMC Med 2022; 20:485. [PMID: 36522747 PMCID: PMC9753387 DOI: 10.1186/s12916-022-02688-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Branched-chain amino acids (BCAAs; valine, leucine, and isoleucine) are essential amino acids that are associated with an increased risk of cardiometabolic diseases (CMD). However, there are still only limited insights into potential direct associations between BCAAs and a wide range of CMD parameters, especially those remaining after correcting for covariates and underlying causal relationships. METHODS To shed light on these relationships, we systematically characterized the associations between plasma BCAA concentrations and a large panel of 537 CMD parameters (including atherosclerosis-related parameters, fat distribution, plasma cytokine concentrations and cell counts, circulating concentrations of cardiovascular-related proteins and plasma metabolites) in 1400 individuals from the Dutch population cohort LifeLines DEEP and 294 overweight individuals from the 300OB cohort. After correcting for age, sex, and BMI, we assessed associations between individual BCAAs and CMD parameters. We further assessed the underlying causality using Mendelian randomization. RESULTS A total of 838 significant associations were detected for 409 CMD parameters. BCAAs showed both common and specific associations, with the most specific associations being detected for isoleucine. Further, we found that obesity status substantially affected the strength and direction of associations for valine, which cannot be corrected for using BMI as a covariate. Subsequent univariable Mendelian randomization (UVMR), after removing BMI-associated SNPs, identified seven significant causal relationships from four CMD traits to BCAA levels, mostly for diabetes-related parameters. However, no causal effects of BCAAs on CMD parameters were supported. CONCLUSIONS Our cross-sectional association study reports a large number of associations between BCAAs and CMD parameters. Our results highlight some specific associations for isoleucine, as well as obesity-specific effects for valine. MR-based causality analysis suggests that altered BCAA levels can be a consequence of diabetes and alteration in lipid metabolism. We found no MR evidence to support a causal role for BCAAs in CMD. These findings provide evidence to (re)evaluate the clinical importance of individual BCAAs in CMD diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Marwah Doestzada
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daria V Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Laboratory of Genomic Diversity, Center for Computer Technologies, ITMO University, St. Petersburg, Russia
| | - Inge C L van den Munckhof
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Daoming Wang
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lianmin Chen
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martijn van Faassen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.,Department for Genomics Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany.,Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Niels P Riksen
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,University of Groningen, University Medical Center Groningen, European Institute of Healthy Ageing (ERIBA), Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands. .,Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
8
|
Vanweert F, Schrauwen P, Phielix E. Role of branched-chain amino acid metabolism in the pathogenesis of obesity and type 2 diabetes-related metabolic disturbances BCAA metabolism in type 2 diabetes. Nutr Diabetes 2022; 12:35. [PMID: 35931683 PMCID: PMC9356071 DOI: 10.1038/s41387-022-00213-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022] Open
Abstract
Branched-chain amino acid (BCAA) catabolism has been considered to have an emerging role in the pathogenesis of metabolic disturbances in obesity and type 2 diabetes (T2D). Several studies showed elevated plasma BCAA levels in humans with insulin resistance and patients with T2D, although the underlying reason is unknown. Dysfunctional BCAA catabolism could theoretically be an underlying factor. In vitro and animal work collectively show that modulation of the BCAA catabolic pathway alters key metabolic processes affecting glucose homeostasis, although an integrated understanding of tissue-specific BCAA catabolism remains largely unknown, especially in humans. Proof-of-concept studies in rodents -and to a lesser extent in humans – strongly suggest that enhancing BCAA catabolism improves glucose homeostasis in metabolic disorders, such as obesity and T2D. In this review, we discuss several hypothesized mechanistic links between BCAA catabolism and insulin resistance and overview current available tools to modulate BCAA catabolism in vivo. Furthermore, this review considers whether enhancing BCAA catabolism forms a potential future treatment strategy to promote metabolic health in insulin resistance and T2D.
Collapse
Affiliation(s)
- Froukje Vanweert
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
9
|
Hamaya R, Mora S, Lawler PR, Cook NR, Buring JE, Lee IM, Manson JE, Tobias DK. Association of Modifiable Lifestyle Factors with Plasma Branched-Chain Amino Acid Metabolites in Women. J Nutr 2022; 152:1515-1524. [PMID: 35259270 PMCID: PMC9178956 DOI: 10.1093/jn/nxac056] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/04/2022] [Accepted: 03/04/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Circulating branched-chain amino acids (BCAAs-isoleucine, leucine, and valine) are strongly associated with higher risk of incident type 2 diabetes (T2D); however, determinants of elevated fasting BCAA concentrations are largely unknown. OBJECTIVES We aimed to characterize the modifiable lifestyle factors related to plasma BCAAs. METHODS We performed a cross-sectional analysis among n = 18,897 women (mean ± SD age: 54.9 ± 7.2 y) in the Women's Health Study, free of T2D and cardiovascular disease at baseline blood draw. Lifestyle factors, weight, and height were self-reported via questionnaire, including smoking status, alcohol, leisure-time physical activity (LTPA), diet quality scores [2010 Alternative Healthy Eating Index (without alcohol) (aHEI); alternate Mediterranean Diet (aMED)], and dietary sources of BCAAs. Plasma BCAAs were quantified via NMR spectroscopy. We calculated multivariable-adjusted percentage mean differences (95% CIs) and P values for linear trend of BCAAs stratified by categoric lifestyle factors. We estimated R2 from univariate cubic spline regression models to estimate the variability in BCAAs explained. RESULTS Compared with women with BMI (in kg/m2) <25.0, BCAAs were 8.6% (95% CI: 8.0%, 9.3%), 15.3% (95% CI: 14.4%, 16.3%), and 21.0% (95% CI: 18.2%, 23.9%) higher for the BMI strata 25.0-29.9, 30.0-39.9, and ≥40.0, respectively (P-trend < 0.0001). Women with higher LTPA and higher alcohol intake compared with lower had modestly (∼1%) lower plasma BCAAs (P-trend = 0.014 and 0.0003, respectively). Differences in smoking status, aHEI, and aMED score were not related to plasma BCAAs. Women with higher dietary BCAAs had dose-response higher plasma BCAA concentrations, 3.4% (95% CI: 2.5%, 4.4%) higher when comparing the highest with the lowest quintile (P-trend < 0.0001). BMI explained 11.6% of the variability of BCAAs, whereas other factors explained between 0.1% and 1%. CONCLUSIONS Our findings among a large cohort of US women indicate that BMI, but less so diet, physical activity, and other lifestyle factors, is related to plasma BCAAs.This trial was registered at clinicaltrials.gov as NCT00000479.
Collapse
Affiliation(s)
- Rikuta Hamaya
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Samia Mora
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Lipid Metabolomics and Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, University Health Network, and Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
| | - Nancy R Cook
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Julie E Buring
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - I-Min Lee
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - JoAnn E Manson
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Deirdre K Tobias
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
10
|
Yu L, Zhu Q, Li Y, Song P, Zhang J. Dietary Branched-Chain Amino Acids (BCAAs) and Risk of Dyslipidemia in a Chinese Population. Nutrients 2022; 14:nu14091824. [PMID: 35565798 PMCID: PMC9103899 DOI: 10.3390/nu14091824] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 12/10/2022] Open
Abstract
This study aimed to explore the association between dietary BCAAs, blood lipid levels and risk of dyslipidemia. In this case−control study, a total of 9541 subjects with normal blood lipids were included as a control group, and 9792 patients with dyslipidemia were included as a case group. Dietary BCAA intake data were measured using 3-day 24 h meal recalls and household condiment weighing. All samples were from China Nutrition and Health Surveillance (2015). Generalized linear model, logistic regression, and restricted cubic spline (RCS) were used to evaluate the relationship between dietary BCAAs, blood lipids and dyslipidemia. After adjusting for confounding factors, dietary BCAAs were positively correlated with TC and LDL-C (p < 0.05). Higher dietary BCAAs were associated with higher OR for Hypercholesteremia (Q4 vs. Q1, OR = 1.29, 95% CI: 1.05−1.58, p-trend = 0.034). The ORs of Hyper-LDL-cholesterolemia showed inverted U-shaped with increasing dietary BCAAs (Q3 vs. Q1, OR = 1.20, 95% CI: 1.03−1.39; Q2 vs. Q1, OR = 1.05, 95% CI: 1.01−1.31). The relationship between dietary BCAAs and the risk of Hypercholesteremia and Hyper-LDL-cholesterolemia were both nonlinear (p nonlinearity = 0.0059, 0.0198). Our study reveals that dietary BCAAs are associated with specific types of lipids and risk of dyslipidemia, some of which may be non-linear.
Collapse
Affiliation(s)
- Lianlong Yu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (L.Y.); (Q.Z.); (Y.L.); (P.S.)
| | - Qianrang Zhu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (L.Y.); (Q.Z.); (Y.L.); (P.S.)
| | - Yuqian Li
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (L.Y.); (Q.Z.); (Y.L.); (P.S.)
- NHC Key Laboratory of Trace Element Nutrition, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Pengkun Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (L.Y.); (Q.Z.); (Y.L.); (P.S.)
| | - Jian Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China; (L.Y.); (Q.Z.); (Y.L.); (P.S.)
- Correspondence: ; Tel.: +86-010-6623-7174
| |
Collapse
|
11
|
Hall ECR, Semenova EA, Bondareva EA, Andryushchenko LB, Larin AK, Cięszczyk P, Generozov EV, Ahmetov II. Association of Genetically Predicted BCAA Levels with Muscle Fiber Size in Athletes Consuming Protein. Genes (Basel) 2022; 13:genes13030397. [PMID: 35327951 PMCID: PMC8955300 DOI: 10.3390/genes13030397] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Branched-chain amino acid (BCAA) levels are associated with skeletal muscle cross-sectional area (CSA). Serum BCAA levels are enhanced by whey protein supplementation (WPS), and evidence in clinical populations suggests an association of single nucleotide polymorphisms (SNPs) with BCAA metabolite levels. It is not known whether the same SNPs are associated with the ability to catabolise BCAAs from exogenous sources, such as WPS. The present study investigated whether possessing a higher number of alleles associated with increased BCAA metabolites correlates with muscle fiber CSA of m. vastus lateralis in physically active participants, and whether any relationship is enhanced by WPS. Endurance-trained participants (n = 75) were grouped by self-reported habitual WPS consumption and genotyped for five SNPs (PPM1K rs1440580, APOA5 rs2072560, CBLN1 rs1420601, DDX19B rs12325419, and TRMT61A rs58101275). Body mass, BMI, and fat percentage were significantly lower and muscle mass higher in the WPS group compared to Non-WPS. The number of BCAA-increasing alleles was correlated with fiber CSA in the WPS group (r = 0.75, p < 0.0001) and was stronger for fast-twitch fibers (p = 0.001) than slow-twitch fibers (p = 0.048). Similar results remained when corrected for multiple covariates (age, physical activity, and meat and dairy intake). No correlation was found in the Non-WPS group. This study presents novel evidence of a positive relationship between BCAA-increasing alleles and muscle fiber CSA in athletes habitually consuming WPS. We suggest that a high number of BCAA-increasing alleles improves the efficiency of WPS by stimulation of muscle protein synthesis, and contributes to greater fiber CSA.
Collapse
Affiliation(s)
- Elliott C. R. Hall
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 5AF, UK;
| | - Ekaterina A. Semenova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (E.A.B.); (A.K.L.); (E.V.G.)
- Research Institute of Physical Culture and Sport, Volga Region State University of Physical Culture, Sport and Tourism, 420010 Kazan, Russia
| | - Elvira A. Bondareva
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (E.A.B.); (A.K.L.); (E.V.G.)
| | - Liliya B. Andryushchenko
- Department of Physical Education, Plekhanov Russian University of Economics, 115093 Moscow, Russia;
| | - Andrey K. Larin
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (E.A.B.); (A.K.L.); (E.V.G.)
| | - Pawel Cięszczyk
- Faculty of Physical Education, Gdańsk University of Physical Education and Sport, 80-854 Gdańsk, Poland;
| | - Edward V. Generozov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (E.A.B.); (A.K.L.); (E.V.G.)
| | - Ildus I. Ahmetov
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 5AF, UK;
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia; (E.A.S.); (E.A.B.); (A.K.L.); (E.V.G.)
- Department of Physical Education, Plekhanov Russian University of Economics, 115093 Moscow, Russia;
- Laboratory of Molecular Genetics, Kazan State Medical University, 420012 Kazan, Russia
- Correspondence:
| |
Collapse
|
12
|
Untargeted Metabolomics Analysis of the Serum Metabolic Signature of Childhood Obesity. Nutrients 2022; 14:nu14010214. [PMID: 35011090 PMCID: PMC8747180 DOI: 10.3390/nu14010214] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Obesity rates among children are growing rapidly worldwide, placing massive pressure on healthcare systems. Untargeted metabolomics can expand our understanding of the pathogenesis of obesity and elucidate mechanisms related to its symptoms. However, the metabolic signatures of obesity in children have not been thoroughly investigated. Herein, we explored metabolites associated with obesity development in childhood. Untargeted metabolomic profiling was performed on fasting serum samples from 27 obese Caucasian children and adolescents and 15 sex- and age-matched normal-weight children. Three metabolomic assays were combined and yielded 726 unique identified metabolites: gas chromatography–mass spectrometry (GC–MS), hydrophilic interaction liquid chromatography coupled to mass spectrometry (HILIC LC–MS/MS), and lipidomics. Univariate and multivariate analyses showed clear discrimination between the untargeted metabolomes of obese and normal-weight children, with 162 significantly differentially expressed metabolites between groups. Children with obesity had higher concentrations of branch-chained amino acids and various lipid metabolites, including phosphatidylcholines, cholesteryl esters, triglycerides. Thus, an early manifestation of obesity pathogenesis and its metabolic consequences in the serum metabolome are correlated with altered lipid metabolism. Obesity metabolite patterns in the adult population were very similar to the metabolic signature of childhood obesity. Identified metabolites could be potential biomarkers and used to study obesity pathomechanisms.
Collapse
|
13
|
Abstract
Background Cardiorespiratory fitness (CRF) is a potent health marker, the improvement of which is associated with a reduced incidence of non-communicable diseases and all-cause mortality. Identifying metabolic signatures associated with CRF could reveal how CRF fosters human health and lead to the development of novel health-monitoring strategies. Objective This article systematically reviewed reported associations between CRF and metabolites measured in human tissues and body fluids. Methods PubMed, EMBASE, and Web of Science were searched from database inception to 3 June, 2021. Metabolomics studies reporting metabolites associated with CRF, measured by means of cardiopulmonary exercise test, were deemed eligible. Backward and forward citation tracking on eligible records were used to complement the results of database searching. Risk of bias at the study level was assessed using QUADOMICS. Results Twenty-two studies were included and 667 metabolites, measured in plasma (n = 619), serum (n = 18), skeletal muscle (n = 16), urine (n = 11), or sweat (n = 3), were identified. Lipids were the metabolites most commonly positively (n = 174) and negatively (n = 274) associated with CRF. Specific circulating glycerophospholipids (n = 85) and cholesterol esters (n = 17) were positively associated with CRF, while circulating glycerolipids (n = 152), glycerophospholipids (n = 42), acylcarnitines (n = 14), and ceramides (n = 12) were negatively associated with CRF. Interestingly, muscle acylcarnitines were positively correlated with CRF (n = 15). Conclusions Cardiorespiratory fitness was associated with circulating and muscle lipidome composition. Causality of the revealed associations at the molecular species level remains to be investigated further. Finally, included studies were heterogeneous in terms of participants’ characteristics and analytical and statistical approaches. PROSPERO Registration Number CRD42020214375. Supplementary Information The online version contains supplementary material available at 10.1007/s40279-021-01590-y.
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: 3.7] [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
|
The effect of physical activity level and exercise training on the association between plasma branched-chain amino acids and intrahepatic lipid content in participants with obesity. Int J Obes (Lond) 2021; 45:1510-1520. [PMID: 33935282 PMCID: PMC8236400 DOI: 10.1038/s41366-021-00815-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/01/2021] [Accepted: 04/09/2021] [Indexed: 01/05/2023]
Abstract
Aims To evaluate whether the association between plasma branched-chain amino acids (BCAA) and intrahepatic lipid (IHL) was affected by physical activity level. Furthermore, to investigate if a conventional exercise training program, a subcategory of physical activity, could lower plasma BCAA along with alterations in IHL content in patients with type 2 diabetes (T2DM) and people with nonalcoholic fatty liver (NAFL). Methods To investigate the effect of physical activity on the association between plasma BCAA and IHL content, linear regression analyses were performed in 1983 individuals from the Netherlands Epidemiology of Obesity (NEO) stratified by physical activity frequency. Furthermore, the effect of a 12-week supervised combined aerobic resistance-exercise program on plasma BCAA, insulin sensitivity (hyperinsulinemic–euglycemic clamp), and IHL (proton-magnetic resonance spectroscopy (1H-MRS)) was investigated in seven patients with T2DM, seven individuals with NAFL and seven BMI-matched control participants (CON). Results We observed positive associations between plasma valine, isoleucine and leucine level, and IHL content (1.29 (95% CI: 1.21, 1.38), 1.52 (95% CI: 1.43, 1.61), and 1.54 (95% CI: 1.44, 1.64) times IHL, respectively, per standard deviation of plasma amino acid level). Similar associations were observed in less active versus more active individuals. Exercise training did not change plasma BCAA levels among groups, but reduced IHL content in NAFL (from 11.6 ± 3.0% pre-exercise to 8.1 ± 2.0% post exercise, p < 0.05) and CON (from 2.4 ± 0.6% pre-exercise to 1.6 ± 1.4% post exercise, p < 0.05), and improved peripheral insulin sensitivity in NAFL as well by ~23% (p < 0.05). Conclusions The association between plasma BCAA levels and IHL is not affected by physical activity level. Exercise training reduced IHL without affecting plasma BCAA levels in individuals with NAFL and CON. We conclude that exercise training-induced reduction in IHL content is not related to changes in plasma BCAA levels. Trial registration Trial registry number: NCT01317576.
Collapse
|
16
|
Ding C, Egli L, Bosco N, Sun L, Goh HJ, Yeo KK, Yap JJL, Actis-Goretta L, Leow MKS, Magkos F. Plasma Branched-Chain Amino Acids Are Associated With Greater Fasting and Postprandial Insulin Secretion in Non-diabetic Chinese Adults. Front Nutr 2021; 8:664939. [PMID: 33996878 PMCID: PMC8113402 DOI: 10.3389/fnut.2021.664939] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 03/30/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Plasma branched-chain amino acids (BCAA) are consistently elevated in subjects with obesity and type 2 diabetes (T2DM) and correlate with insulin resistance. The association of BCAA with insulin secretion and clearance rates has not been adequately described. Objective: To evaluate the relationships between fasting and postprandial plasma BCAA, insulin secretion and insulin clearance. Design: Ninety-five non-diabetic Chinese subjects (43 females) underwent a mixed-meal tolerance test; blood biomarkers including BCAAs (leucine, isoleucine, valine) were measured for 6 h. Fasting and postprandial insulin secretion rates (ISR) and insulin clearance were determined by oral minimal modeling of glucose and C-peptide. Results: Fasting and postprandial plasma BCAA correlated strongly with each other (ρ = 0.796, P < 0.001), and both were positively associated with basal ISR (ρ = 0.45/0.36, P < 0.001), total postprandial ISR AUC (ρ = 0.37/0.45, P < 0.001), and negatively with insulin clearance (ρ = -0.29/-0.29, P < 0.01), after adjusting for sex and body mass index. These relationships largely persisted after adjusting further for insulin resistance and postprandial glucose. Compared with subjects in the middle and lowest tertiles for fasting or postprandial plasma BCAA, subjects in the highest tertile had significantly greater postprandial glucose (by 7-10%) and insulin (by 74-98%) concentrations, basal ISRs (by 34-53%), postprandial ISR AUCs (by 41-49%), and lower insulin clearance rates (by 17-22%) (all P < 0.05). Conclusions: Fasting and postprandial plasma BCAA levels are associated with greater fasting and postprandial insulin secretion and reduced insulin clearance in healthy Chinese subjects. These observations potentially highlight an additional layer of involvement of BCAA in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
| | - Leonie Egli
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Singapore, Singapore
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Lijuan Sun
- Singapore Institute for Clinical Sciences, Singapore, Singapore
| | - Hui Jen Goh
- Singapore Institute for Clinical Sciences, Singapore, Singapore
| | - Khung Keong Yeo
- Duke-NUS Medical School, Singapore, Singapore
- National Heart Centre Singapore, Singapore, Singapore
| | - Jonathan Jiunn Liang Yap
- National Heart Centre Singapore, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - Melvin Khee-Shing Leow
- Singapore Institute for Clinical Sciences, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Faidon Magkos
- Department of Nutrition, Exercise & Sports, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
17
|
de Mello VD, Sehgal R, Männistö V, Klåvus A, Nilsson E, Perfilyev A, Kaminska D, Miao Z, Pajukanta P, Ling C, Hanhineva K, Pihlajamäki J. Serum aromatic and branched-chain amino acids associated with NASH demonstrate divergent associations with serum lipids. Liver Int 2021; 41:754-763. [PMID: 33219609 PMCID: PMC8048463 DOI: 10.1111/liv.14743] [Citation(s) in RCA: 19] [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] [Received: 06/17/2020] [Revised: 11/04/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) has been associated with multiple metabolic abnormalities. By applying a non-targeted metabolomics approach, we aimed at investigating whether serum metabolite profile that associates with NAFLD would differ in its association with NAFLD-related metabolic risk factors. METHODS & RESULTS A total of 233 subjects (mean ± SD: 48.3 ± 9.3 years old; BMI: 43.1 ± 5.4 kg/m2 ; 64 male) undergoing bariatric surgery were studied. Of these participants, 164 with liver histology could be classified as normal liver (n = 79), simple steatosis (SS, n = 40) or non-alcoholic steatohepatitis (NASH, n = 45). Among the identified fasting serum metabolites with higher levels in those with NASH when compared to those with normal phenotype were the aromatic amino acids (AAAs: tryptophan, tyrosine and phenylalanine), the branched-chain amino acids (BCAAs: leucine and isoleucine), a phosphatidylcholine (PC(16:0/16:1)) and uridine (all FDRp < 0.05). Only tryptophan was significantly higher in those with NASH compared to those with SS (FDRp < 0.05). Only the AAAs tryptophan and tyrosine correlated positively with serum total and LDL cholesterol (FDRp < 0.1), and accordingly, with liver LDLR at mRNA expression level. In addition, tryptophan was the single AA associated with liver DNA methylation of CpG sites known to be differentially methylated in those with NASH. CONCLUSIONS We found that serum levels of the NASH-related AAAs and BCAAs demonstrate divergent associations with serum lipids. The specific correlation of tryptophan with LDL-c may result from the molecular events affecting LDLR mRNA expression and NASH-associated methylation of genes in the liver.
Collapse
Affiliation(s)
- Vanessa D. de Mello
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Ratika Sehgal
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Ville Männistö
- Department of MedicineUniversity of Eastern Finland and Kuopio University HospitalKuopioFinland
| | - Anton Klåvus
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Emma Nilsson
- Epigenetics and Diabetes UnitDepartment of Clinical SciencesLund University Diabetes CentreMalmöSweden
| | - Alexander Perfilyev
- Epigenetics and Diabetes UnitDepartment of Clinical SciencesLund University Diabetes CentreMalmöSweden
| | - Dorota Kaminska
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Zong Miao
- Department of Human GeneticsDavid Geffen School of Medicine at University of California Los Angeles (UCLA)Los AngelesCAUSA
| | - Päivi Pajukanta
- Department of Human GeneticsDavid Geffen School of Medicine at University of California Los Angeles (UCLA)Los AngelesCAUSA,Institute for Precision HealthSchool of MedicineUCLALos AngelesCAUSA
| | - Charlotte Ling
- Epigenetics and Diabetes UnitDepartment of Clinical SciencesLund University Diabetes CentreMalmöSweden
| | - Kati Hanhineva
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland,Department of BiochemistryFood Chemistry and Food Development UnitUniversity of TurkuTurkuFinland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland,Department of Medicine, Endocrinology and Clinical NutritionKuopio University HospitalKuopioFinland
| |
Collapse
|
18
|
de Almeida-Pititto B, Dualib PM, Jordão MC, Izar Helfenstein Fonseca M, Jones SR, Blaha MJ, Toth PP, Santos RD, Bensenor IM, Ferreira SRG, Lotufo PA. Branched-chain amino acids predict incident diabetes in the Brazilian Longitudinal Study of Adult Health - ELSA-Brasil. Diabetes Res Clin Pract 2021; 174:108747. [PMID: 33713721 DOI: 10.1016/j.diabres.2021.108747] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/14/2021] [Accepted: 03/01/2021] [Indexed: 11/16/2022]
Abstract
AIMS To evaluate the role of branch chain amino acid (BCAA) concentrations as a predictor for incident type 2 diabetes (DM). METHODS Participants from ELSA-Brasil without diabetes at baseline and followed for 3.9 ± 0.6 years were included in the analysis. The determinations of BCAA (valine, leucine, isoleucine) were performed by proton nuclear magnetic resonance spectroscopy. Cardiometabolic profile and incidence of DM were evaluated according to quartiles of BCAA at baseline, stratified by sex. RESULTS From 3,828 participants (56% female, 50.5 ± 8.7 years) 299 (8.5%) were diagnosed with DM. For both sexes, a worsening of cardiometabolic profile was observed across increasing BCAA quartiles. In survival analysis, incidence rates of DM for the entire period were highest in participants in the third and fourth quartile of BCAA (log Rank analysis < 0.001 for both sexes). In Cox regression analysis, for men, the HR (95%CI) for risk of DM was 2.24 (1.24-4.03) for those from the fourth quartile of BCAA, while in women it was 1.94 (1.07-3.50), comparing to first quartile of BCAA after adjustments for age, BMI, physical activity, family history of DM, pre-diabetes, blood pressure, total cholesterol and HOMA-IR. CONCLUSIONS Higher levels of BCAA were independently predictors of DM.
Collapse
Affiliation(s)
- Bianca de Almeida-Pititto
- Departamento de Medicina Preventiva, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 740, CEP 04023900 São Paulo, Brazil; Programa de Pós-Graduação em Endocrinologia e Metabologia, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Estado de Israel 639, CEP 04023900, São Paulo, Brazil.
| | - Patrícia M Dualib
- Programa de Pós-Graduação em Endocrinologia e Metabologia, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Estado de Israel 639, CEP 04023900, São Paulo, Brazil.
| | - Martha C Jordão
- Programa de Pós-Graduação em Endocrinologia e Metabologia, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Estado de Israel 639, CEP 04023900, São Paulo, Brazil.
| | | | - Steven R Jones
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Blalock 524 D1, 600 N. Wolfe St, Baltimore, MD, USA.
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Blalock 524 D1, 600 N. Wolfe St, Baltimore, MD, USA.
| | - Peter P Toth
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Blalock 524 D1, 600 N. Wolfe St, Baltimore, MD, USA; Department of Preventive Cardiology, CGH Medical Center, 100 E. Le Fevre Road 61081 Sterling, IL, USA.
| | - Raul D Santos
- Heart Institute (InCor), University of São Paulo Medical School Hospital, Av. Dr. Enéas de Carvalho Aguiar, 44, CEP 01246-000 São Paulo, Brazil.
| | - Isabela M Bensenor
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade de São Paulo, Av. Lineu Prestes 2565 - 4° floor, CEP: 05508-000 São Paulo, SP, Brazil; Centro de Pesquisa do Hospital Universitário, Universidade de São Paulo, Av. Lineu Prestes 2565, 3rd floor, CEP 05508-000 São Paulo, SP, Brazil
| | - Sandra Roberta G Ferreira
- Departamento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo, Brazil.
| | - Paulo A Lotufo
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade de São Paulo, Av. Lineu Prestes 2565 - 4° floor, CEP: 05508-000 São Paulo, SP, Brazil; Centro de Pesquisa do Hospital Universitário, Universidade de São Paulo, Av. Lineu Prestes 2565, 3rd floor, CEP 05508-000 São Paulo, SP, Brazil.
| |
Collapse
|
19
|
Long J, Yang Z, Wang L, Han Y, Peng C, Yan C, Yan D. Metabolite biomarkers of type 2 diabetes mellitus and pre-diabetes: a systematic review and meta-analysis. BMC Endocr Disord 2020; 20:174. [PMID: 33228610 PMCID: PMC7685632 DOI: 10.1186/s12902-020-00653-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND We aimed to explore metabolite biomarkers that could be used to identify pre-diabetes and type 2 diabetes mellitus (T2DM) using systematic review and meta-analysis. METHODS Four databases, the Cochrane Library, EMBASE, PubMed and Scopus were selected. A random effect model and a fixed effect model were applied to the results of forest plot analyses to determine the standardized mean difference (SMD) and 95% confidence interval (95% CI) for each metabolite. The SMD for every metabolite was then converted into an odds ratio to create an metabolite biomarker profile. RESULTS Twenty-four independent studies reported data from 14,131 healthy individuals and 3499 patients with T2DM, and 14 included studies reported 4844 healthy controls and a total of 2139 pre-diabetes patients. In the serum and plasma of patients with T2DM, compared with the healthy participants, the concentrations of valine, leucine, isoleucine, proline, tyrosine, lysine and glutamate were higher and that of glycine was lower. The concentrations of isoleucine, alanine, proline, glutamate, palmitic acid, 2-aminoadipic acid and lysine were higher and those of glycine, serine, and citrulline were lower in prediabetic patients. Metabolite biomarkers of T2DM and pre-diabetes revealed that the levels of alanine, glutamate and palmitic acid (C16:0) were significantly different in T2DM and pre-diabetes. CONCLUSIONS Quantified multiple metabolite biomarkers may reflect the different status of pre-diabetes and T2DM, and could provide an important reference for clinical diagnosis and treatment of pre-diabetes and T2DM.
Collapse
Affiliation(s)
- Jianglan Long
- Beijing Key Laboratory and Joint Laboratory for International Cooperation of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Zhirui Yang
- Beijing Key Laboratory and Joint Laboratory for International Cooperation of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
| | - Long Wang
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yumei Han
- Beijing Physical Examination Center, Beijing, 100077, China
| | - Cheng Peng
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Can Yan
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Dan Yan
- Beijing Key Laboratory and Joint Laboratory for International Cooperation of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China.
| |
Collapse
|
20
|
High Fat-High Fructose Diet-Induced Changes in the Gut Microbiota Associated with Dyslipidemia in Syrian Hamsters. Nutrients 2020; 12:nu12113557. [PMID: 33233570 PMCID: PMC7699731 DOI: 10.3390/nu12113557] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Aim: The objective of this study was to characterize the early effects of high fructose diets (with and without high fat) on both the composition of the gut microbiota and lipid metabolism in Syrian hamsters, a reproducible preclinical model of diet-induced dyslipidemia. Methods: Eight-week-old male hamsters were fed diets consisting of high-fat/high-fructose, low-fat/high-fructose or a standard chow diet for 14 days. Stool was collected at baseline (day 0), day 7 and day 14. Fasting levels of plasma triglycerides and cholesterol were monitored on day 0, day 7 and day 14, and nonfasting levels were also assayed on day 15. Then, 16S rRNA sequencing of stool samples was used to determine gut microbial composition, and predictive metagenomics was performed to evaluate dietary-induced shifts in deduced microbial functions. Results: Both high-fructose diets resulted in divergent gut microbiota composition. A high-fat/high-fructose diet induced the largest shift in overall gut microbial composition, with dramatic shifts in the Firmicute/Bacteroidetes ratio, and changes in beta diversity after just seven days of dietary intervention. Significant associations between genus level taxa and dietary intervention were identified, including an association with Ruminococceace NK4A214 group in high-fat/high-fructose fed animals and an association with Butryimonas with the low-fat/high-fructose diet. High-fat/high-fructose feeding induced dyslipidemia with increases in plasma triglycerides and cholesterol, and hepatomegaly. Dietary-induced changes in several genus level taxa significantly correlated with lipid levels over the two-week period. Differences in microbial metabolic pathways between high-fat/high-fructose and low-fat/high-fructose diet fed hamsters were identified, and several of these pathways also correlated with lipid profiles in hamsters. Conclusions: The high-fat/high-fructose diet caused shifts in the host gut microbiota. These dietary-induced alterations in gut microbial composition were linked to changes in the production of secondary metabolites, which contributed to the development of metabolic syndrome in the host.
Collapse
|
21
|
Muyyarikkandy MS, McLeod M, Maguire M, Mahar R, Kattapuram N, Zhang C, Surugihalli C, Muralidaran V, Vavilikolanu K, Mathews CE, Merritt ME, Sunny NE. Branched chain amino acids and carbohydrate restriction exacerbate ketogenesis and hepatic mitochondrial oxidative dysfunction during NAFLD. FASEB J 2020; 34:14832-14849. [PMID: 32918763 DOI: 10.1096/fj.202001495r] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/10/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022]
Abstract
Mitochondrial adaptation during non-alcoholic fatty liver disease (NAFLD) include remodeling of ketogenic flux and sustained tricarboxylic acid (TCA) cycle activity, which are concurrent to onset of oxidative stress. Over 70% of obese humans have NAFLD and ketogenic diets are common weight loss strategies. However, the effectiveness of ketogenic diets toward alleviating NAFLD remains unclear. We hypothesized that chronic ketogenesis will worsen metabolic dysfunction and oxidative stress during NAFLD. Mice (C57BL/6) were kept (for 16-wks) on either a low-fat, high-fat, or high-fat diet supplemented with 1.5X branched chain amino acids (BCAAs) by replacing carbohydrate calories (ketogenic). The ketogenic diet induced hepatic lipid oxidation and ketogenesis, and produced multifaceted changes in flux through the individual steps of the TCA cycle. Higher rates of hepatic oxidative fluxes fueled by the ketogenic diet paralleled lower rates of de novo lipogenesis. Interestingly, this metabolic remodeling did not improve insulin resistance, but induced fibrogenic genes and inflammation in the liver. Under a chronic "ketogenic environment," the hepatocyte diverted more acetyl-CoA away from lipogenesis toward ketogenesis and TCA cycle, a milieu which can hasten oxidative stress and inflammation. In summary, chronic exposure to ketogenic environment during obesity and NAFLD has the potential to aggravate hepatic mitochondrial dysfunction.
Collapse
Affiliation(s)
| | - Marc McLeod
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Meghan Maguire
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Rohit Mahar
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Nathan Kattapuram
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Christine Zhang
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Chaitra Surugihalli
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Vaishna Muralidaran
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Kruthi Vavilikolanu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Clayton E Mathews
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Matthew E Merritt
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Nishanth E Sunny
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| |
Collapse
|
22
|
Panjwani AA, Ji Y, Fahey JW, Palmer A, Wang G, Hong X, Zuckerman B, Wang X. Maternal Dyslipidemia, Plasma Branched-Chain Amino Acids, and the Risk of Child Autism Spectrum Disorder: Evidence of Sex Difference. J Autism Dev Disord 2019; 50:540-550. [PMID: 31686281 DOI: 10.1007/s10803-019-04264-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In contrast to the well-observed associations between obesity, diabetes, and autism spectrum disorder (ASD), the roles of maternal dyslipidemia and sex disparity in ASD have not been well-studied. We examined the joint associations of maternal plasma cholesterols, branched-chain amino acids (BCAAs) and child sex on child ASD risk. We analyzed data from 756 mother-infant pairs (86 ASD) from the Boston Birth Cohort. Maternal plasma cholesterols and BCAAs were measured in samples collected 24-72 h postpartum. We found that in this urban, low-income prospective birth cohort, low maternal high-density lipoprotein cholesterol (HDL-C), above-median maternal plasma BCAA concentrations, and male sex additively or synergistically increased risk of ASD. Additional studies are necessary to confirm our findings.
Collapse
Affiliation(s)
- Anita A Panjwani
- Department of International Health, Center for Human Nutrition, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yuelong Ji
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, E4132, Baltimore, MD, 21205-2179, USA
| | - Jed W Fahey
- Department of International Health, Center for Human Nutrition, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.,Department of Pharmacology and Molecular Sciences, Cullman Chemoprotection Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amanda Palmer
- Department of International Health, Center for Human Nutrition, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Guoying Wang
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, E4132, Baltimore, MD, 21205-2179, USA
| | - Xiumei Hong
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, E4132, Baltimore, MD, 21205-2179, USA
| | - Barry Zuckerman
- Department of Pediatrics, School of Medicine and Boston Medical Center, Boston University, Boston, MA, USA
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Center on the Early Life Origins of Disease, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, E4132, Baltimore, MD, 21205-2179, USA. .,Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
Abstract
Metabolomics uses advanced analytical chemistry techniques to enable the high-throughput characterization of metabolites from cells, organs, tissues, or biofluids. The rapid growth in metabolomics is leading to a renewed interest in metabolism and the role that small molecule metabolites play in many biological processes. As a result, traditional views of metabolites as being simply the "bricks and mortar" of cells or just the fuel for cellular energetics are being upended. Indeed, metabolites appear to have much more varied and far more important roles as signaling molecules, immune modulators, endogenous toxins, and environmental sensors. This review explores how metabolomics is yielding important new insights into a number of important biological and physiological processes. In particular, a major focus is on illustrating how metabolomics and discoveries made through metabolomics are improving our understanding of both normal physiology and the pathophysiology of many diseases. These discoveries are yielding new insights into how metabolites influence organ function, immune function, nutrient sensing, and gut physiology. Collectively, this work is leading to a much more unified and system-wide perspective of biology wherein metabolites, proteins, and genes are understood to interact synergistically to modify the actions and functions of organelles, organs, and organisms.
Collapse
Affiliation(s)
- David S Wishart
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
24
|
Beneficial effects of running and milk protein supplements on Sirtuins and risk factors of metabolic disorders in rats with low aerobic capacity. Metabol Open 2019; 4:100019. [PMID: 32812928 PMCID: PMC7424841 DOI: 10.1016/j.metop.2019.100019] [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: 08/19/2019] [Revised: 10/06/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022] Open
Abstract
Background Physical activity and dietary intake of dairy products are associated with improved metabolic health. Dairy products are rich with branched chain amino acids that are essential for energy production. To gain insight into the mechanisms underlying the benefit of the sub-chronic effects of running and intake of milk protein supplements, we studied Low Capacity Runner rats (LCR), a rodent exercise model with risk for metabolic disorders. We especially focused on the role of Sirtuins, energy level dependent proteins that affect many cellular metabolic processes. Methods Forty-seven adult LCR female rats sedentary or running voluntarily in wheels were fed normal chow and given supplements of either whey or milk protein drink (PD)-supplemented water, or water only for 21 weeks. Physiological responses were measured in vivo. Blood lipids were determined from serum. Mitochondrial markers and Sirtuins (Sirt1-7) including downstream targets were measured in plantaris muscle by western blotting. Results For the first 10 weeks whey-drinking rats ran about 50% less compared to other groups; still, in all runners glucose tolerance improved and triglycerides decreased. Generally, running induced a ∼six-fold increase in running capacity and a ∼8% decrease in % body fat. Together with running, protein supplements increased the relative lean mass of the total body weight by ∼11%. In comparison with sedentary controls, running and whey increased HDL (21%) and whey, with or without running, lowered LDL (−34%). Running increased mitochondrial biogenesis and Sirtuins 3 and 4. When combined with exercise, both whey and milk protein drink induced about a 4-fold increase in Sirt3, compared to runners drinking water only, and about a 2-fold increase compared to the respective sedentary group. Protein supplements, with or without running, enhanced the phosphorylation level of the acetyl-coA-carboxylase, suggesting increased fat oxidation. Both supplemented diets increased Sirt5 and Sirt7 without an additional effect from exercise. Running diminished and PD supplement increased Sirt6. Conclusion We demonstrate in rats new sub-chronic effects of milk proteins on metabolism that involve Sirtuins and their downstream targets in skeletal muscle. The results show that running and milk proteins act on reducing the risk factors of metabolic disorders and suggest that the underlying mechanisms may involve Sirtuins. Notably, we found that milk protein supplements have some favorable effects on metabolism even without running. Interactive effects of running and/or milk protein supplements were studied. Milk protein drink enhanced and whey diminished the amount of voluntary running. Despite less running whey-supplementation improved metabolic health. Almost all Sirtuins in muscle adapted to milk protein and running interventions.
Collapse
|
25
|
Altered branched chain amino acid metabolism: toward a unifying cardiometabolic hypothesis. Curr Opin Cardiol 2019; 33:558-564. [PMID: 29994805 DOI: 10.1097/hco.0000000000000552] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Atherosclerotic cardiovascular disease (CVD) and type II diabetes (T2D) share common etiologic pathways that may long precede the development of clinically evident disease. Early identification of risk markers could support efforts to individualize risk prediction and improve the efficacy of primary prevention, as well as uncover novel therapeutic targets. RECENT FINDINGS Altered metabolism of branched-chain amino acids (BCAAs), and their subsequent accumulation in circulation, may precede the development of insulin resistance and clinically manifest cardiometabolic diseases. BCAAs - the essential amino acids leucine, isoleucine and valine - likely promote insulin resistance through activation of mammalian target of rapamycin complex 1. Epidemiologic studies demonstrate robust associations between BCAAs and incident T2D, and Mendelian randomization supports a potentially causal relationship. More recently, there is emerging evidence that BCAAs are also associated with incident atherosclerotic CVD, possibly mediated by the development of T2D. SUMMARY In this article, we review the biochemistry of BCAAs, their potential contribution to cardiometabolic risk, the available evidence from molecular epidemiologic studies to date, and, finally, consider future research and clinical directions. Overall, BCAAs represent a promising emerging target for risk stratification and possible intervention, to support efforts to mitigate the burden of cardiometabolic disease in the population.
Collapse
|
26
|
Liang Y, Kong F, Torres-Romero I, Burlacot A, Cuine S, Légeret B, Billon E, Brotman Y, Alseekh S, Fernie AR, Beisson F, Peltier G, Li-Beisson Y. Branched-Chain Amino Acid Catabolism Impacts Triacylglycerol Homeostasis in Chlamydomonas reinhardtii. PLANT PHYSIOLOGY 2019; 179:1502-1514. [PMID: 30728273 PMCID: PMC6446750 DOI: 10.1104/pp.18.01584] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/30/2019] [Indexed: 05/05/2023]
Abstract
Nitrogen (N) starvation-induced triacylglycerol (TAG) synthesis, and its complex relationship with starch metabolism in algal cells, has been intensively studied; however, few studies have examined the interaction between amino acid metabolism and TAG biosynthesis. Here, via a forward genetic screen for TAG homeostasis, we isolated a Chlamydomonas (Chlamydomonas reinhardtii) mutant (bkdE1α) that is deficient in the E1α subunit of the branched-chain ketoacid dehydrogenase (BCKDH) complex. Metabolomics analysis revealed a defect in the catabolism of branched-chain amino acids in bkdE1α Furthermore, this mutant accumulated 30% less TAG than the parental strain during N starvation and was compromised in TAG remobilization upon N resupply. Intriguingly, the rate of mitochondrial respiration was 20% to 35% lower in bkdE1α compared with the parental strains. Three additional knockout mutants of the other components of the BCKDH complex exhibited phenotypes similar to that of bkdE1α Transcriptional responses of BCKDH to different N status were consistent with its role in TAG homeostasis. Collectively, these results indicate that branched-chain amino acid catabolism contributes to TAG metabolism by providing carbon precursors and ATP, thus highlighting the complex interplay between distinct subcellular metabolisms for oil storage in green microalgae.
Collapse
Affiliation(s)
- Yuanxue Liang
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Fantao Kong
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Ismael Torres-Romero
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Adrien Burlacot
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Stéphan Cuine
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Bertrand Légeret
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Emmanuelle Billon
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Yariv Brotman
- Max Planck Institute of Molecular Plant Physiology, 14476 Potsdam-Golm, Germany
| | - Saleh Alseekh
- Max Planck Institute of Molecular Plant Physiology, 14476 Potsdam-Golm, Germany
| | - Alisdair R Fernie
- Max Planck Institute of Molecular Plant Physiology, 14476 Potsdam-Golm, Germany
| | - Fred Beisson
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Gilles Peltier
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| | - Yonghua Li-Beisson
- Aix-Marseille University, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Commissariat à l'Energie Atomique Cadarache, Saint-Paul-lez Durance F-13108, France
| |
Collapse
|
27
|
Gu Q, Spinelli JJ, Dummer TBJ, McDonald TE, Moore SC, Murphy RA. Metabolic profiling of adherence to diet, physical activity and body size recommendations for cancer prevention. Sci Rep 2018; 8:16293. [PMID: 30390014 PMCID: PMC6214951 DOI: 10.1038/s41598-018-34662-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022] Open
Abstract
Maintaining a healthy body weight, eating well and being physically active lowers cancer risk by 30%. However, the biology underlying these relationships is not well understood. We examined cross-sectional associations between metabolites and cancer preventive behaviors as well as the relevance to cancer-related pathways among 120 participants (50% men, mean BMI 26.6 kg/m2, mean age 54 years) with no history of smoking or cancer. Participants completed questionnaires, physical measurements and provided blood samples. Non-targeted nuclear magnetic resonance captured 223 metabolite measures. Factor analysis was performed separately for amino acid, fatty acid and lipoprotein groups. Multivariable-adjusted linear regression was used to evaluate associations between cancer preventive recommendations and metabolite-containing factors (p-value < 0.05, false discovery rate <0.20). An inflammation-related metabolite (glycoprotein acetylation) loaded strongly on a factor that was associated with excess adiposity (body fat ≥25% (men) or ≥30% (women) ß (SE) = 0.74 (0.18)) and not meeting physical activity recommendations (ß (SE) = 0.40 (0.20)). Insulin sensitivity-related metabolites including monounsaturated and polyunsaturated fats were lower among participants not meeting recommendations for adiposity, fruits and vegetables and physical activity while branched chain amino acids were higher. Cancer preventive behaviors were associated with complex metabolic signatures, including alterations in pathways known to be involved in cancer pathogenesis.
Collapse
Affiliation(s)
- Qianqian Gu
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - John J Spinelli
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada.,Cancer Control Research, BC Cancer Agency, Vancouver, BC, Canada
| | - Trevor B J Dummer
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | | | - Steven C Moore
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Rachel A Murphy
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
28
|
Kalavalapalli S, Bril F, Koelmel JP, Abdo K, Guingab J, Andrews P, Li WY, Jose D, Yost RA, Frye RF, Garrett TJ, Cusi K, Sunny NE. Pioglitazone improves hepatic mitochondrial function in a mouse model of nonalcoholic steatohepatitis. Am J Physiol Endocrinol Metab 2018; 315:E163-E173. [PMID: 29634314 PMCID: PMC6139494 DOI: 10.1152/ajpendo.00023.2018] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pioglitazone is effective in improving insulin resistance and liver histology in patients with nonalcoholic steatohepatitis (NASH). Because dysfunctional mitochondrial metabolism is a central feature of NASH, we hypothesized that an important target of pioglitazone would be alleviating mitochondrial oxidative dysfunction. To this end, we studied hepatic mitochondrial metabolism in mice fed high-fructose high-transfat diet (TFD) supplemented with pioglitazone for 20 wk, using nuclear magnetic resonance-based 13C isotopomer analysis. Pioglitazone improved whole body and adipose insulin sensitivity in TFD-fed mice. Furthermore, pioglitazone reduced intrahepatic triglyceride content and fed plasma ketones and hepatic TCA cycle flux, anaplerosis, and pyruvate cycling in mice with NASH. This was associated with a marked reduction in most intrahepatic diacylglycerol classes and, to a lesser extent, some ceramide species (C22:1, C23:0). Considering the cross-talk between mitochondrial function and branched-chain amino acid (BCAA) metabolism, pioglitazone's impact on plasma BCAA profile was determined in a cohort of human subjects. Pioglitazone improved the plasma BCAA concentration profile in patients with NASH. This appeared to be related to an improvement in BCAA degradation in multiple tissues. These results provide evidence that pioglitazone-induced changes in NASH are related to improvements in hepatic mitochondrial oxidative dysfunction and changes in whole body BCAA metabolism.
Collapse
Affiliation(s)
- Srilaxmi Kalavalapalli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida , Gainesville, Florida
| | - Fernando Bril
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida , Gainesville, Florida
| | - Jeremy P Koelmel
- Department of Chemistry, University of Florida , Gainesville, Florida
| | - Kaitlyn Abdo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida , Gainesville, Florida
| | - Joy Guingab
- Department of Pathology, University of Florida , Gainesville, Florida
| | - Paige Andrews
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida , Gainesville, Florida
| | - Wen-Yi Li
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida , Gainesville, Florida
| | - Dhanya Jose
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida , Gainesville, Florida
| | - Richard A Yost
- Department of Chemistry, University of Florida , Gainesville, Florida
- Department of Pathology, University of Florida , Gainesville, Florida
| | - Reginald F Frye
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida , Gainesville, Florida
| | - Timothy J Garrett
- Department of Pathology, University of Florida , Gainesville, Florida
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida , Gainesville, Florida
- Division of Endocrinology, Diabetes and Metabolism, Malcom Randall Veterans Administration Medical Center , Gainesville, Florida
| | - Nishanth E Sunny
- Department of Animal and Avian Sciences, University of Maryland , College Park, Maryland
| |
Collapse
|
29
|
Ruiz-Canela M, Guasch-Ferré M, Toledo E, Clish CB, Razquin C, Liang L, Wang DD, Corella D, Estruch R, Hernáez Á, Yu E, Gómez-Gracia E, Zheng Y, Arós F, Romaguera D, Dennis C, Ros E, Lapetra J, Serra-Majem L, Papandreou C, Portoles O, Fitó M, Salas-Salvadó J, Hu FB, Martínez-González MA. Plasma branched chain/aromatic amino acids, enriched Mediterranean diet and risk of type 2 diabetes: case-cohort study within the PREDIMED Trial. Diabetologia 2018; 61:1560-1571. [PMID: 29663011 PMCID: PMC5988977 DOI: 10.1007/s00125-018-4611-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/12/2018] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs) are associated with type 2 diabetes. However, repeated measurements of BCAA/AAA and their interactions with dietary interventions have not been evaluated. We investigated the associations between baseline and changes at 1 year in BCAA/AAA with type 2 diabetes in the context of a Mediterranean diet (MedDiet) trial. METHODS We included 251 participants with incident type 2 diabetes and a random sample of 694 participants (641 participants without type 2 diabetes and 53 overlapping cases) in a case-cohort study nested within the PREvención con DIeta MEDiterránea (PREDIMED) trial. Participants were randomised to a MedDiet+extra-virgin olive oil (n = 273), a MedDiet+nuts (n = 324) or a control diet (n = 295). We used LC-MS/MS to measure plasma levels of amino acids. Type 2 diabetes was a pre-specified secondary outcome of the PREDIMED trial. RESULTS Elevated plasma levels of individual BCAAs/AAAs were associated with higher type 2 diabetes risk after a median follow-up of 3.8 years: multivariable HR for the highest vs lowest quartile ranged from 1.32 for phenylalanine ([95% CI 0.90, 1.92], p for trend = 0.015) to 3.29 for leucine ([95% CI 2.03, 5.34], p for trend<0.001). Increases in BCAA score at 1 year were associated with higher type 2 diabetes risk in the control group with HR per SD = 1.61 (95% CI 1.02, 2.54), but not in the MedDiet groups (p for interaction <0.001). The MedDiet+extra-virgin olive oil significantly reduced BCAA levels after 1 year of intervention (p = 0.005 vs the control group). CONCLUSIONS/INTERPRETATION Our results support that higher baseline BCAAs and their increases at 1 year were associated with higher type 2 diabetes risk. A Mediterranean diet rich in extra-virgin olive oil significantly reduced the levels of BCAA and attenuated the positive association between plasma BCAA levels and type 2 diabetes incidence. Clinical trial number: SRCTN35739639 ( www.controlled-trials.com ).
Collapse
Affiliation(s)
- Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, Facultad de Medicina, Universidad de Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain.
| | - Marta Guasch-Ferré
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Estefanía Toledo
- Department of Preventive Medicine and Public Health, Facultad de Medicina, Universidad de Navarra, Irunlarrea 1, 31008, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Clary B Clish
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cristina Razquin
- Department of Preventive Medicine and Public Health, Facultad de Medicina, Universidad de Navarra, Irunlarrea 1, 31008, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Liming Liang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dolores Corella
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Ramón Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Biomedical Research Institute August Pi Sunyer (IDI- BAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Álvaro Hernáez
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular and Nutrition Research Group (Regicor Study Group), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Edward Yu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Yan Zheng
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Fernando Arós
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cardiology, University Hospital, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Dora Romaguera
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases, Mallorca, Spain
| | - Courtney Dennis
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Emilio Ros
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition Biomedical Research Institute August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - José Lapetra
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Family Medicine, Research Unit, Primary Care Division of Sevilla, Sevilla, Spain
| | - Lluis Serra-Majem
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Research Institute of Biomedical and Health Sciences and Medical School University of Las Palmas de Gran Canarias, Las Palmas de Gran Canaria, Spain
| | - Christopher Papandreou
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
| | - Olga Portoles
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Montserrat Fitó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular and Nutrition Research Group (Regicor Study Group), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Jordi Salas-Salvadó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Miguel A Martínez-González
- Department of Preventive Medicine and Public Health, Facultad de Medicina, Universidad de Navarra, Irunlarrea 1, 31008, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
30
|
Abstract
Branched-chain amino acids (BCAA) have increasingly been studied as playing a role in diabetes, with the PubMed search string "diabetes" AND "branched chain amino acids" showing particular growth in studies of the topic over the past decade (Fig. ). In the Young Finn's Study, BCAA and, to a lesser extent, the aromatic amino acids phenylalanine and tyrosine were associated with insulin resistance (IR) in men but not in women, whereas the gluconeogenic amino acids alanine, glutamine, or glycine, and several other amino acids (i.e. histidine, arginine, and tryptophan) did not show an association with IR. Obesity may track more strongly than metabolic syndrome and diabetes with elevated BCAA. In a study of 1302 people aged 40-79; higher levels of BCAA tracked with older age, male sex, and metabolic syndrome, as well as with obesity, cardiovascular risk, dyslipidemia, hypertension, and uric acid. Medium- and long-chain acylcarnitines, by-products of mitochondrial catabolism of BCAAs, as well as branched-chain keto acids and the BCAA themselves distinguished obese people having versus not having features of IR, and in a study of 898 patients with essential hypertension, the BCAA and tyrosine and phenylalanine were associated with metabolic syndrome and impaired fasting glucose. In a meta-analysis of three genome-wide association studies, elevations in BCAA and, to a lesser extent, in alanine tracked with IR, whereas higher levels of glutamine and glycine were associated with lesser likelihood of IR. Given these associations with IR, it is not surprising that a number of studies have shown higher BCAA levels in people with and prior to development of type 2 diabetes (T2D), although this has particularly been shown in Caucasian and Asian ethnic groups while not appearing to occur in African Americans. Similarly, higher BCAA levels track with cardiovascular disease. [Figure: see text] The metabolism of BCAA involves two processes: (i) a reversible process catalysed by a branched-chain aminotransferase (BCAT), either cytosolic or mitochondrial, requiring pyridoxal to function as an amino group carrier, by which the BCAA with 2-ketoglutarate produce a branched-chain keto acid plus glutamate; and (ii) the irreversible mitochondrial process catalysed by branched-chain keto acid dehydrogenase (BCKDH) leading to formation of acetyl-coenzyme A (CoA), propionyl-CoA, and 2-methylbutyryl-CoA from leucine, valine, and isoleucine, respectively, which enter the tricarboxylic acid (Krebs) cycle as acetyl-CoA, propionyl-CoA, and 2-methylbutyryl-CoA, respectively, leading to ATP formation. The BCAA stimulate secretion of both insulin and glucagon and, when given orally, of both glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), with oral administration leading to greater and more prolonged insulin and glucagon secretion. Insulin may particularly reduce BCAA turnover to a greater extent than that of other amino acids, and decreases the appearance and increases the uptake of amino acids. However, older studies of the effect of glucose or insulin on BCAA concentrations and rates of leucine appearance and oxidation showed no reduction in T2D, although the higher baseline levels of BCAA in obesity have long been recognized. Impaired function of BCAT and BCKDH has been posited, either as a primary genetic abnormality or due to effects of elevated fatty acids, proinflammatory cytokines, or insulin levels with consequent accumulation of branched-chain keto acids and metabolites such as diacylglycerol and ceramide, potentially contributing to the development of further insulin resistance, and decreased skeletal muscle BCAT and BCKDH expression has been shown in people with diabetes, supporting this concept. A Mendelian randomization study used measured variation in genes involved in BCAA metabolism to test the hypothesis of a causal effect of modifiable exposure on IR, showing that variants in protein phosphatase, Mg2+ /Mn2+ dependent 1K (PPM1K), a gene encoding the mitochondrial phosphatase activating the BCKDH complex, are associated with T2D, but another such study suggested that genetic variations associated with IR are causally related to higher BCAA levels. Another hypothesis involves the mammalian target of rapamycin complex 1 (mTORC1), which is activated by BCAA, as well as by insulin and glucose via cellular ATP availability. If this is the relevant pathway, BCAA overload may cause insulin resistance by activation of mammalian target of rapamycin (mTOR), as well as by leading to increases in acylcarnitines, with mTOR seen in this scenario as a central signal of cross-talk between the BCAA and insulin. At this point, whether whole-body or tissue-specific BCAA metabolism is increased or decreased in states of insulin-resistant obesity and T2D is uncertain. Insulin action in the hypothalamus induces but overfeeding decreases hepatic BCKDH, leading to the concept that hypothalamic insulin resistance impairs BCAA metabolism in obesity and diabetes, so that plasma BCAAs may be markers of hypothalamic insulin action rather than direct mediators of changes in IR. A way to address this may be to understand the effects of changes in diet and other interventions on BCAA, as well as on IR and T2D. In an animal model, lowering dietary BCAA increased energy expenditure and improved insulin sensitivity. Two large human population studies showed an association of estimated dietary BCAA intake with T2D risk, although another population study showed higher dietary BCAA to be associated with lower T2D risk. Ethnic differences, reflecting underlying differences in genetic variants, may be responsible for such differences. In the study of Asghari et al. in the current issue of the Journal of Diabetes, BCAA intake was associated with the development of subsequent IR. Studies of bariatric surgery suggest lower basal and post-insulin infusion BCAA levels are associated with greater insulin sensitivity, with reductions in BCAA not seen with weight loss per se with gastric band procedures, but occurring after Roux-en-Y gastric bypass, an intervention that may have metabolic benefits over and above those from reduction in body weight. The gut microbiota may be important for the supply of the BCAA to mammalian hosts, either by de novo biosynthesis or by modifying nutrient absorption. A final fascinating preliminary set of observations is that of the effects of empagliflozin on metabolomics; evidence of increased Krebs cycle activation and of higher levels of BCAA metabolites, such as acylcarnitines, suggests that sodium-glucose cotransporter 2 (SGLT2) inhibition may, to some extent, involve BCAA metabolism. Certainly, we do not yet have a full understanding of these complex associations. However, the suggestion of multiple roles of BCAA in the development of IR promises to be important and to lead to the development of novel effective T2D therapies.
Collapse
|