1
|
Korvyakova Y, Azarova I, Klyosova E, Postnikova M, Makarenko V, Bushueva O, Solodilova M, Polonikov A. The link between the ANPEP gene and type 2 diabetes mellitus may be mediated by the disruption of glutathione metabolism and redox homeostasis. Gene 2025; 935:149050. [PMID: 39489227 DOI: 10.1016/j.gene.2024.149050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Aminopeptidase N (ANPEP), a membrane-associated ectoenzyme, has been identified as a susceptibility gene for type 2 diabetes (T2D) by genome-wide association and transcriptome studies; however, the mechanisms by which this gene contributes to disease pathogenesis remain unclear. The aim of this study was to determine the comprehensive contribution of ANPEP polymorphisms to T2D risk and annotate the underlying mechanisms. A total of 3206 unrelated individuals including 1579 T2D patients and 1627 controls were recruited for the study. Twenty-three common functional single nucleotide polymorphisms (SNP) of ANPEP were genotyped by the MassArray-4 system. Six polymorphisms, rs11073891, rs12898828, rs12148357, rs9920421, rs7111, and rs25653, were found to be associated with type 2 diabetes (Pperm ≤ 0.05). Common haplotype rs9920421G-rs4932143G-rs7111T was strongly associated with increased risk of T2D (Pperm = 5.9 × 10-12), whereas two rare haplotypes such as rs9920421G-rs4932143C-rs7111T (Pperm = 6.5 × 10-40) and rs12442778A-rs12898828A-rs6496608T-rs11073891C (Pperm = 1.0 × 10-7) possessed strong protection against disease. We identified 38 and 109 diplotypes associated with T2D risk in males and females, respectively (FDR ≤ 0.05). ANPEP polymorphisms showed associations with plasma levels of fasting blood glucose, aspartate aminotransferase, total protein and glutathione (P < 0.05), and several haplotypes were strongly associated with the levels of reactive oxygen species and uric acid (P < 0.0001). A deep literature analysis has facilitated the formulation of a hypothesis proposing that increased plasma levels of ANPEP as well as liver enzymes such as aspartate aminotransferase, alanine aminotransferase and gammaglutamyltransferase serve as an adaptive response directed towards the restoration of glutathione deficiency in diabetics by stimulating the production of amino acid precursors for glutathione biosynthesis.
Collapse
Affiliation(s)
- Yaroslava Korvyakova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Research Centre for Medical Genetics, 1 Moskvorechie St., Moscow 115522, Russian Federation.
| | - Iuliia Azarova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Maria Postnikova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| | - Victor Makarenko
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation
| | - Olga Bushueva
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation; Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| | - Maria Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation; Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| |
Collapse
|
2
|
Xiao QA, Chen L, Li XL, Han Q, Li HT, Zhang XL. Effect of different types of milk consumption on type 2 diabetes and the mediating effect of amino acids: a Mendelian randomization study of East Asian population. J Dairy Sci 2025:S0022-0302(25)00016-5. [PMID: 39824484 DOI: 10.3168/jds.2024-26017] [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: 11/15/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
There is currently a lack of research examining the association between the consumption of different dairy products and type 2 diabetes (T2D) in East Asian populations. To address this gap, the present study employs Mendelian randomization (MR) to investigate the potential impact of 3 different types of milk consumption (including whole milk, semi-skim milk, and skim milk) on the risk of developing T2D. The results indicate that both whole milk and skim milk are associated with an increased risk of T2D (whole milk: OR = 1.022, 95% confidence interval [CI]: 1.001-1.044, P = 0.042, skim milk: OR = 1.023, 95% CI: 1.007-1.039, P = 0.005). Mediation analysis reveals that asparagine acts as a mediator between skim milk consumption and T2D, with a mediation effect of 0.003 (95% CI: 0.000 to 0.008), accounting for 14.269% of the total effect.
Collapse
Affiliation(s)
- Qing-Ao Xiao
- Department of Interventional Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, Hubei Province, China; Department of Interventional Radiology, Yichang Central People's Hospital, Yichang 443003, Hubei Province, China
| | - Lin Chen
- Department of Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, Hubei Province, China; Department of Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, Hubei Province, China
| | - Xiao-Long Li
- Department of Interventional Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, Hubei Province, China; Department of Interventional Radiology, Yichang Central People's Hospital, Yichang 443003, Hubei Province, China
| | - Qiang Han
- Department of Interventional Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, Hubei Province, China; Department of Interventional Radiology, Yichang Central People's Hospital, Yichang 443003, Hubei Province, China
| | - Hai-Tao Li
- Department of Interventional Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, Hubei Province, China; Department of Interventional Radiology, Yichang Central People's Hospital, Yichang 443003, Hubei Province, China
| | - Xiao-Ling Zhang
- Department of Interventional Radiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang 443003, Hubei Province, China; Department of Interventional Radiology, Yichang Central People's Hospital, Yichang 443003, Hubei Province, China.
| |
Collapse
|
3
|
Peng M, Fu Y, Qin C, Shi L, Zhang M, Zhou S. A stratified study of human blood metabolites and coronary artery diseases-A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2025; 35:103754. [PMID: 39448312 DOI: 10.1016/j.numecd.2024.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/07/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysregulation is closely associated with coronary artery diseases (CAD). Exploring the relationship between metabolites and CAD is helpful in identifying changes in energy metabolism during disease progression. METHODS AND RESULTS We use Mendelian Randomization (MR) analysis to assess the relationships between 275 serum metabolites and CAD such as angina pectoris, post-myocardial infarction complications, coronary atherosclerosis, myocardial infarction (MI), and unstable angina pectoris (UA). The inverse variance-weighted method (IVW) served as the primary approach for causal analysis, with MR-Egger and weighted median (WM) as supplementary methods. Sensitivity analyses were conducted to assess heterogeneity and multiple effects. We also analyzed potentially related metabolic pathways.We identified causal relationships between 42 known metabolites and CAD. Among them, the genetic susceptibility to elevated levels of amino acid Isobutyrylcarnitine is associated with an increased risk of coronary artery atherosclerosis; but it provides protection against the development of MI. Genetic susceptibility to elevated levels of fatty acids Stearate, Caprylate is associated with higher risk of angina pectoris, while Threonate has a protective effect in the development of angina; Stearate is associated with an increased risk of UA, whereas higher levels of the lipids Choline, 1-arachidonoylglycerophosphoinositol∗, Hexadecanedioate, Tetradecanedioate play a protective role in UA.Metabolic pathway analysis identified 6 pathways that may be associated with CAD. CONCLUSION We identified causal relationships between 42 serum metabolites and CAD. Specifically, changes in metabolites such as Isobutyrylcarnitine, Caprylate, and Stearate were associated with risks of CAD. These findings provide new insights into the metabolic mechanisms of CAD.
Collapse
Affiliation(s)
- Mengling Peng
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Yu Fu
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Cong Qin
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Lei Shi
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Meiwei Zhang
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China
| | - Shanshan Zhou
- The Center of Cardiovascular Diseases, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, China.
| |
Collapse
|
4
|
Jiang Z, Qian M, Zhen Z, Yang X, Xu C, Zuo L, Jiang J, Zhang W, Hu N. Gut microbiota and metabolomic profile changes play critical roles in tacrolimus-induced diabetes in rats. Front Cell Infect Microbiol 2024; 14:1436477. [PMID: 39355267 PMCID: PMC11442430 DOI: 10.3389/fcimb.2024.1436477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/14/2024] [Indexed: 10/03/2024] Open
Abstract
Aims Hyperglycemia is one of the adverse effects of tacrolimus (TAC), but the underlying mechanism is not fully identified. We used multi-omics analysis to evaluate the changes in the gut microbiota and metabolic profile of rats with TAC-induced diabetes. Methods To establish a diabetic animal model, Sprague Dawley rats were divided randomly into two groups. Those in the TAC group received intraperitoneal injections of TAC (3 mg/kg) for 8 weeks, and those in the CON group served as the control. 16S rRNA sequencing was used to analyze fecal microbiota. The metabolites of the two groups were detected and analyzed by nontargeted and targeted metabolomics, including amino acids (AAs), bile acids (BAs), and short-chain fatty acids (SCFAs). Results The rats treated with TAC exhibited hyperglycemia as well as changes in the gut microbiota and metabolites. Specifically, their gut microbiota had significantly higher abundances of Escherichia-Shigella, Enterococcus, and Allobaculum, and significantly lower abundances of Ruminococcus, Akkermansia, and Roseburia. In addition, they had significantly reduced serum levels of AAs including asparagine, aspartic acid, glutamic acid, and methionine. With respect to BAs, they had significantly higher serum levels of taurocholic acid (TCA), and glycochenodeoxycholic acid (GCDCA), but significantly lower levels of taurodeoxycholic acid (TDCA) and tauroursodeoxycholic acid (TUDCA). There were no differences in the levels of SCFAs between the two groups. Correlations existed among glucose metabolism indexes (fasting blood glucose and fasting insulin), gut microbiota (Ruminococcus and Akkermansia), and metabolites (glutamic acid, hydroxyproline, GCDCA, TDCA, and TUDCA). Conclusions Both AAs and BAs may play crucial roles as signaling molecules in the regulation of TAC-induced diabetes.
Collapse
Affiliation(s)
- Zhenwei Jiang
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Minyan Qian
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Zeng Zhen
- Changzhou Key Laboratory of Human Use Experience Research & Transformation of Menghe Medical School, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Xuping Yang
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Caomei Xu
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Li'an Zuo
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wenting Zhang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Pediatric Central Laboratory, Affiliated Changzhou Children's Hospital of Nantong University, Changzhou, China
| | - Nan Hu
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| |
Collapse
|
5
|
Wu J, Palasantzas V, Andreu-Sánchez S, Plösch T, Leonard S, Li S, Bonder MJ, Westra HJ, van Meurs J, Ghanbari M, Franke L, Zhernakova A, Fu J, Hoogerland JA, Zhernakova DV. Epigenome-wide association study on the plasma metabolome suggests self-regulation of the glycine and serine pathway through DNA methylation. Clin Epigenetics 2024; 16:104. [PMID: 39138531 PMCID: PMC11323446 DOI: 10.1186/s13148-024-01718-7] [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: 12/14/2023] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND The plasma metabolome reflects the physiological state of various biological processes and can serve as a proxy for disease risk. Plasma metabolite variation, influenced by genetic and epigenetic mechanisms, can also affect the cellular microenvironment and blood cell epigenetics. The interplay between the plasma metabolome and the blood cell epigenome remains elusive. In this study, we performed an epigenome-wide association study (EWAS) of 1183 plasma metabolites in 693 participants from the LifeLines-DEEP cohort and investigated the causal relationships in DNA methylation-metabolite associations using bidirectional Mendelian randomization and mediation analysis. RESULTS After rigorously adjusting for potential confounders, including genetics, we identified five robust associations between two plasma metabolites (L-serine and glycine) and three CpG sites located in two independent genomic regions (cg14476101 and cg16246545 in PHGDH and cg02711608 in SLC1A5) at a false discovery rate of less than 0.05. Further analysis revealed a complex bidirectional relationship between plasma glycine/serine levels and DNA methylation. Moreover, we observed a strong mediating role of DNA methylation in the effect of glycine/serine on the expression of their metabolism/transport genes, with the proportion of the mediated effect ranging from 11.8 to 54.3%. This result was also replicated in an independent population-based cohort, the Rotterdam Study. To validate our findings, we conducted in vitro cell studies which confirmed the mediating role of DNA methylation in the regulation of PHGDH gene expression. CONCLUSIONS Our findings reveal a potential feedback mechanism in which glycine and serine regulate gene expression through DNA methylation.
Collapse
Affiliation(s)
- Jiafei Wu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Victoria Palasantzas
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Perinatal Neurobiology Research Group, Department of Pediatrics, School of Medicine and Health Sciences, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Sam Leonard
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Shuang Li
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Marc Jan Bonder
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Harm-Jan Westra
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Joyce van Meurs
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lude Franke
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Joanne A Hoogerland
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands.
- Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands.
| | - Daria V Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
6
|
Shen CY, Lu CH, Cheng CF, Li KJ, Kuo YM, Wu CH, Liu CH, Hsieh SC, Tsai CY, Yu CL. Advanced Glycation End-Products Acting as Immunomodulators for Chronic Inflammation, Inflammaging and Carcinogenesis in Patients with Diabetes and Immune-Related Diseases. Biomedicines 2024; 12:1699. [PMID: 39200164 PMCID: PMC11352041 DOI: 10.3390/biomedicines12081699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/22/2024] [Accepted: 07/27/2024] [Indexed: 09/02/2024] Open
Abstract
Increased production of advanced glycation end products (AGEs) among reducing sugars (glucose, fructose, galactose, or ribose) and amino acids/proteins via non-enzymatic Maillard reaction can be found in lifestyle-related disease (LSRD), metabolic syndrome (MetS), and obesity and immune-related diseases. Increased serum levels of AGEs may induce aging, diabetic complications, cardiovascular diseases (CVD), neurodegenerative diseases (NDD), cancer, and inflamm-aging (inflammation with immunosenescence). The Maillard reaction can also occur among reducing sugars and lipoproteins or DNAs to alter their structure and induce immunogenicity/genotoxicity for carcinogenesis. AGEs, as danger-associated molecular pattern molecules (DAMPs), operate via binding to receptor for AGE (RAGE) or other scavenger receptors on cell surface to activate PI3K-Akt-, P38-MAPK-, ERK1/2-JNK-, and MyD88-induced NF-κB signaling pathways to mediate various pathological effects. Recently, the concept of "inflamm-aging" became more defined, and we have unveiled some interesting findings in relation to it. The purpose of the present review is to dissect the potential molecular basis of inflamm-aging in patients with diabetes and immune-mediated diseases caused by different AGEs.
Collapse
Affiliation(s)
- Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| | - Cheng-Hsun Lu
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Chiao-Feng Cheng
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital-Hsinchu Branch, # 2, Section 1, Shengyi Road, Hsinchu County 302058, Taiwan;
| | - Chin-Hsiu Liu
- Department of Internal Medicine, National Taiwan University Hospital-Yunlin Branch, # 579, Section 2, Yunlin Road, Yunlin County 640203, Taiwan;
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| | - Chang-Youh Tsai
- Department of Internal Medicine, Fu-Jen Catholic University Hospital, College of Medicine, Fu-Jen Catholic University, # 69 Guizi Road, New Taipei City 24352, Taiwan
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, # 7 Chung-Shan South Road, Taipei 10002, Taiwan; (C.-Y.S.); (C.-H.L.); (C.-F.C.); (K.-J.L.)
| |
Collapse
|
7
|
Liu N, Yin Z, Wang M, Kui H, Yuan Z, Tian Y, Liu C, Huang J. Pharmacodynamic and targeted amino acid metabolomics researches on the improvement of diabetic retinopathy with Fufang Xueshuantong component compatibility. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1242:124194. [PMID: 38924945 DOI: 10.1016/j.jchromb.2024.124194] [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: 12/04/2023] [Revised: 03/28/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
The Fufang Xueshuantong capsule (FXT) has significant preventive and therapeutic effects on diabetic retinopathy(DR), but the compatibility of its active components remains to be thoroughly explored. In this study, a zebrafish diabetic retinopathy model was established using high-mixed sugars, and the optimal ratios of notoginseng total saponins, total salvianolic acid, astragaloside, and harpagide were selected through orthogonal experiments. Furthermore, we used UPLC-QqQ/MS to detect the changes in amino acid content of DR zebrafish tissues after administration of FXT and its compatible formula to analyze the effects of FXT and its compatible formula on amino acid metabolites. The results showed that the final compatibility ratios of the components were 8: 5: 1: 6.6 by comprehensive evaluation of the indicators. FXT and its compatibility formula had beneficial effects on retinal vasodilatation, lipid accumulation in the liver, total glucose, and VEGF levels in DR zebrafish, and all of them could call back some amino acid levels in DR zebrafish. In this research, we determined the compatible formulation of the active ingredients in the FXT and investigated their efficacy in DR zebrafish for further clinical applications.
Collapse
Affiliation(s)
- Ning Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Lab for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziqiang Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingshuang Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Hongqian Kui
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhenshuang Yuan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yue Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chuanxin Liu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003.
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
8
|
Tanase DM, Valasciuc E, Costea CF, Scripcariu DV, Ouatu A, Hurjui LL, Tarniceriu CC, Floria DE, Ciocoiu M, Baroi LG, Floria M. Duality of Branched-Chain Amino Acids in Chronic Cardiovascular Disease: Potential Biomarkers versus Active Pathophysiological Promoters. Nutrients 2024; 16:1972. [PMID: 38931325 PMCID: PMC11206939 DOI: 10.3390/nu16121972] [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: 05/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Branched-chain amino acids (BCAAs), comprising leucine (Leu), isoleucine (Ile), and valine (Val), are essential nutrients vital for protein synthesis and metabolic regulation via specialized signaling networks. Their association with cardiovascular diseases (CVDs) has become a focal point of scientific debate, with emerging evidence suggesting both beneficial and detrimental roles. This review aims to dissect the multifaceted relationship between BCAAs and cardiovascular health, exploring the molecular mechanisms and clinical implications. Elevated BCAA levels have also been linked to insulin resistance (IR), type 2 diabetes mellitus (T2DM), inflammation, and dyslipidemia, which are well-established risk factors for CVD. Central to these processes are key pathways such as mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-light-chain-enhancer of activate B cells (NF-κB)-mediated inflammation, and oxidative stress. Additionally, the interplay between BCAA metabolism and gut microbiota, particularly the production of metabolites like trimethylamine-N-oxide (TMAO), adds another layer of complexity. Contrarily, some studies propose that BCAAs may have cardioprotective effects under certain conditions, contributing to muscle maintenance and metabolic health. This review critically evaluates the evidence, addressing the biological basis and signal transduction mechanism, and also discusses the potential for BCAAs to act as biomarkers versus active mediators of cardiovascular pathology. By presenting a balanced analysis, this review seeks to clarify the contentious roles of BCAAs in CVD, providing a foundation for future research and therapeutic strategies required because of the rising prevalence, incidence, and total burden of CVDs.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Claudia Florida Costea
- Department of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iași, Romania
| | - Dragos Viorel Scripcariu
- Department of General Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Regional Institute of Oncology, 700483 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| | - Loredana Liliana Hurjui
- Department of Morpho-Functional Sciences II, Physiology Discipline, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Laboratory, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Claudia Cristina Tarniceriu
- Department of Morpho-Functional Sciences I, Discipline of Anatomy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Clinic, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Diana Elena Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Institute of Gastroenterology and Hepatology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Livia Genoveva Baroi
- Department of Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Vascular Surgery, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (A.O.); (D.E.F.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, Iasi 700111, Romania
| |
Collapse
|
9
|
Miguel-Albarreal AD, Rivero-Pino F, Marquez-Paradas E, Grao-Cruces E, Gonzalez-de la Rosa T, Montserrat-de la Paz S. Mediterranean Diet Combined with Regular Aerobic Exercise and Hemp Protein Supplementation Modulates Plasma Circulating Amino Acids and Improves the Health Status of Overweight Individuals. Nutrients 2024; 16:1594. [PMID: 38892526 PMCID: PMC11174559 DOI: 10.3390/nu16111594] [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: 04/30/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Plant protein is considered a sustainable health-promoting strategy to prevent metabolic syndrome. Lifestyle changes (including dietary patterns and exercise) have been demonstrated to exert an effect on human health by modulating the biochemical status in humans. The objective of this study was to assess whether supplementation with hemp protein within a Mediterranean diet context together with exercise could help to ameliorate the metabolic statuses of patients prone to developing metabolic syndrome. For this study, 23 patients followed with Mediterranean diet and engaged in aerobic exercise according to the WHO's recommendations, while also being supplemented with hemp protein, for 12 weeks. A comparison of anthropometric, biochemical, and mineral data as well as amino acid values was made between the start and the end of the study, with the subjects acting as their own control group. Statistical analyses included a paired t-test, Wilcoxon paired test, Pearson correlation coefficient, and Sparse Partial Least Squares Discriminant Analysis to evaluate significant differences and correlations among parameters. There were statistically significant changes in total cholesterol, HDL-C (+52.3%), LDL-C (-54.0%), and TAG levels (-49.8%), but not in glucose plasma levels. Following the intervention, plasma concentrations of some amino acids, including α-aminoadipic acid, phosphoethanolamine, and 1-metylhistidine, increased, whereas those of asparagine and alanine declined. Different correlations between amino acids and the other parameters evaluated were reported and discussed. A Mediterranean diet combined with regular aerobic exercise, together with protein supplementation, can highly improve the metabolic parameters and anthropometric parameters of subjects with obesity and impaired glucose levels, ameliorating their health status and likely delaying the development of metabolic syndrome.
Collapse
Affiliation(s)
- Antonio D. Miguel-Albarreal
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.D.M.-A.); (E.M.-P.); (E.G.-C.); (T.G.-d.l.R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain
| | - Fernando Rivero-Pino
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.D.M.-A.); (E.M.-P.); (E.G.-C.); (T.G.-d.l.R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain
| | - Elvira Marquez-Paradas
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.D.M.-A.); (E.M.-P.); (E.G.-C.); (T.G.-d.l.R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain
| | - Elena Grao-Cruces
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.D.M.-A.); (E.M.-P.); (E.G.-C.); (T.G.-d.l.R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain
| | - Teresa Gonzalez-de la Rosa
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.D.M.-A.); (E.M.-P.); (E.G.-C.); (T.G.-d.l.R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.D.M.-A.); (E.M.-P.); (E.G.-C.); (T.G.-d.l.R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain
| |
Collapse
|
10
|
Figueiredo JC, Bhowmick NA, Karlstaedt A. Metabolic basis of cardiac dysfunction in cancer patients. Curr Opin Cardiol 2024; 39:138-147. [PMID: 38386340 PMCID: PMC11185275 DOI: 10.1097/hco.0000000000001118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
PURPOSE OF REVIEW The relationship between metabolism and cardiovascular diseases is complex and bidirectional. Cardiac cells must adapt metabolic pathways to meet biosynthetic demands and energy requirements to maintain contractile function. During cancer, this homeostasis is challenged by the increased metabolic demands of proliferating cancer cells. RECENT FINDINGS Tumors have a systemic metabolic impact that extends beyond the tumor microenvironment. Lipid metabolism is critical to cancer cell proliferation, metabolic adaptation, and increased cardiovascular risk. Metabolites serve as signals which provide insights for diagnosis and prognosis in cardio-oncology patients. SUMMARY Metabolic processes demonstrate a complex relationship between cancer cell states and cardiovascular remodeling with potential for therapeutic interventions.
Collapse
Affiliation(s)
- Jane C. Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Neil Adri Bhowmick
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Division of Hematology and Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anja Karlstaedt
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
11
|
John T, Saffoon N, Walsby-Tickle J, Hester SS, Dingler FA, Millington CL, McCullagh JSO, Patel KJ, Hopkinson RJ, Schofield CJ. Aldehyde-mediated inhibition of asparagine biosynthesis has implications for diabetes and alcoholism. Chem Sci 2024; 15:2509-2517. [PMID: 38362406 PMCID: PMC10866355 DOI: 10.1039/d3sc06551k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/01/2024] [Indexed: 02/17/2024] Open
Abstract
Patients with alcoholism and type 2 diabetes manifest altered metabolism, including elevated aldehyde levels and unusually low asparagine levels. We show that asparagine synthetase B (ASNS), the only human asparagine-forming enzyme, is inhibited by disease-relevant reactive aldehydes, including formaldehyde and acetaldehyde. Cellular studies show non-cytotoxic amounts of reactive aldehydes induce a decrease in asparagine levels. Biochemical analyses reveal inhibition results from reaction of the aldehydes with the catalytically important N-terminal cysteine of ASNS. The combined cellular and biochemical results suggest a possible mechanism underlying the low asparagine levels in alcoholism and diabetes. The results will stimulate research on the biological consequences of the reactions of aldehydes with nucleophilic residues.
Collapse
Affiliation(s)
- Tobias John
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Nadia Saffoon
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - John Walsby-Tickle
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Svenja S Hester
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford Oxford UK
| | - Felix A Dingler
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - Christopher L Millington
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - James S O McCullagh
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Ketan J Patel
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - Richard J Hopkinson
- Leicester Institute for Structural and Chemical Biology and School of Chemistry, University of Leicester, Henry Wellcome Building Lancaster Road Leicester LE1 7RH UK
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| |
Collapse
|
12
|
Markin SS, Ponomarenko EA, Romashova YA, Pleshakova TO, Ivanov SV, Bedretdinov FN, Konstantinov SL, Nizov AA, Koledinskii AG, Girivenko AI, Shestakova KM, Markin PA, Moskaleva NE, Kozhevnikova MV, Chefranova ZY, Appolonova SA. A novel preliminary metabolomic panel for IHD diagnostics and pathogenesis. Sci Rep 2024; 14:2651. [PMID: 38302683 PMCID: PMC10834974 DOI: 10.1038/s41598-024-53215-9] [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: 09/28/2023] [Accepted: 01/30/2024] [Indexed: 02/03/2024] Open
Abstract
Cardiovascular disease (CVD) represents one of the main causes of mortality worldwide and nearly a half of it is related to ischemic heart disease (IHD). The article represents a comprehensive study on the diagnostics of IHD through the targeted metabolomic profiling and machine learning techniques. A total of 112 subjects were enrolled in the study, consisting of 76 IHD patients and 36 non-CVD subjects. Metabolomic profiling was conducted, involving the quantitative analysis of 87 endogenous metabolites in plasma. A novel regression method of age-adjustment correction of metabolomics data was developed. We identified 36 significantly changed metabolites which included increased cystathionine and dimethylglycine and the decreased ADMA and arginine. Tryptophan catabolism pathways showed significant alterations with increased levels of serotonin, intermediates of the kynurenine pathway and decreased intermediates of indole pathway. Amino acid profiles indicated elevated branched-chain amino acids and increased amino acid ratios. Short-chain acylcarnitines were reduced, while long-chain acylcarnitines were elevated. Based on these metabolites data, machine learning algorithms: logistic regression, support vector machine, decision trees, random forest, and gradient boosting, were used for IHD diagnostic models. Random forest demonstrated the highest accuracy with an AUC of 0.98. The metabolites Norepinephrine; Xanthurenic acid; Anthranilic acid; Serotonin; C6-DC; C14-OH; C16; C16-OH; GSG; Phenylalanine and Methionine were found to be significant and may serve as a novel preliminary panel for IHD diagnostics. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- S S Markin
- Institute of Biomedical Chemistry, Moscow, Russia, 119121.
| | | | - Yu A Romashova
- Institute of Biomedical Chemistry, Moscow, Russia, 119121
| | - T O Pleshakova
- Institute of Biomedical Chemistry, Moscow, Russia, 119121
| | - S V Ivanov
- Institute of Biomedical Chemistry, Moscow, Russia, 119121
| | | | - S L Konstantinov
- Belgorod Regional Clinical Hospital of St. Joseph, Belgorod, Russia, 308007
| | - A A Nizov
- I.P. Pavlov Ryazan State Medical University, Ryazan, Russia, 390026
| | - A G Koledinskii
- Peoples' Friendship University of Russia, Moscow, Russia, 117198
| | - A I Girivenko
- I.P. Pavlov Ryazan State Medical University, Ryazan, Russia, 390026
| | - K M Shestakova
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow Medical University (Sechenov University), Moscow, Russia, 119435
| | - P A Markin
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow Medical University (Sechenov University), Moscow, Russia, 119435
| | - N E Moskaleva
- World-Class Research Center Digital Biodesign and Personalized Healthcare, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| | - M V Kozhevnikova
- Hospital Therapy No1, Department of the N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow Medical University (Sechenov University), Moscow, Russia, 119435
| | - Zh Yu Chefranova
- Belgorod Regional Clinical Hospital of St. Joseph, Belgorod, Russia, 308007
| | - S A Appolonova
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow Medical University (Sechenov University), Moscow, Russia, 119435
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119435
| |
Collapse
|
13
|
Wang M, Ou Y, Yuan XL, Zhu XF, Niu B, Kang Z, Zhang B, Ahmed A, Xing GQ, Su H. Heterogeneously elevated branched-chain/aromatic amino acids among new-onset type-2 diabetes mellitus patients are potentially skewed diabetes predictors. World J Diabetes 2024; 15:53-71. [PMID: 38313852 PMCID: PMC10835491 DOI: 10.4239/wjd.v15.i1.53] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/03/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND The lack of specific predictors for type-2 diabetes mellitus (T2DM) severely impacts early intervention/prevention efforts. Elevated branched-chain amino acids (BCAAs: Isoleucine, leucine, valine) and aromatic amino acids (AAAs: Tyrosine, tryptophan, phenylalanine)) show high sensitivity and specificity in predicting diabetes in animals and predict T2DM 10-19 years before T2DM onset in clinical studies. However, improvement is needed to support its clinical utility. AIM To evaluate the effects of body mass index (BMI) and sex on BCAAs/AAAs in new-onset T2DM individuals with varying body weight. METHODS Ninety-seven new-onset T2DM patients (< 12 mo) differing in BMI [normal weight (NW), n = 33, BMI = 22.23 ± 1.60; overweight, n = 42, BMI = 25.9 ± 1.07; obesity (OB), n = 22, BMI = 31.23 ± 2.31] from the First People's Hospital of Yunnan Province, Kunming, China, were studied. One-way and 2-way ANOVAs were conducted to determine the effects of BMI and sex on BCAAs/AAAs. RESULTS Fasting serum AAAs, BCAAs, glutamate, and alanine were greater and high-density lipoprotein (HDL) was lower (P < 0.05, each) in OB-T2DM patients than in NW-T2DM patients, especially in male OB-T2DM patients. Arginine, histidine, leucine, methionine, and lysine were greater in male patients than in female patients. Moreover, histidine, alanine, glutamate, lysine, valine, methionine, leucine, isoleucine, tyrosine, phenylalanine, and tryptophan were significantly correlated with abdominal adiposity, body weight and BMI, whereas isoleucine, leucine and phenylalanine were negatively correlated with HDL. CONCLUSION Heterogeneously elevated amino acids, especially BCAAs/AAAs, across new-onset T2DM patients in differing BMI categories revealed a potentially skewed prediction of T2DM development. The higher BCAA/AAA levels in obese T2DM patients would support T2DM prediction in obese individuals, whereas the lower levels of BCAAs/AAAs in NW-T2DM individuals may underestimate T2DM risk in NW individuals. This potentially skewed T2DM prediction should be considered when BCAAs/AAAs are to be used as the T2DM predictor.
Collapse
Affiliation(s)
- Min Wang
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan Province, China
| | - Yang Ou
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Xiang-Lian Yuan
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Xiu-Fang Zhu
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan Province, China
| | - Ben Niu
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Zhuang Kang
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Bing Zhang
- Clinical Laboratory, Nanchong Central Hospital & The Second Clinical Medical College of North Sichuan Medical University, Nanchong 637000, Sichuan Province, China
| | - Anwar Ahmed
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Guo-Qiang Xing
- The Affiliated Hospital and Second Clinical Medical College, North Sichuan Medical University, Nanchong 637000, Sichuan Province, China
- Department of Research and Development, Lotus Biotech.com LLC, Gaithersburg, MD 20878, United States
| | - Heng Su
- Department of Endocrinology, The First People’s Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| |
Collapse
|
14
|
Guevara-Ramírez P, Paz-Cruz E, Cadena-Ullauri S, Ruiz-Pozo VA, Tamayo-Trujillo R, Felix ML, Simancas-Racines D, Zambrano AK. Molecular pathways and nutrigenomic review of insulin resistance development in gestational diabetes mellitus. Front Nutr 2023; 10:1228703. [PMID: 37799768 PMCID: PMC10548225 DOI: 10.3389/fnut.2023.1228703] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/01/2023] [Indexed: 10/07/2023] Open
Abstract
Gestational diabetes mellitus is a condition marked by raised blood sugar levels and insulin resistance that usually occurs during the second or third trimester of pregnancy. According to the World Health Organization, hyperglycemia affects 16.9% of pregnancies worldwide. Dietary changes are the primarily alternative treatment for gestational diabetes mellitus. This paper aims to perform an exhaustive overview of the interaction between diet, gene expression, and the metabolic pathways related to insulin resistance. The intake of foods rich in carbohydrates can influence the gene expression of glycolysis, as well as foods rich in fat, can disrupt the beta-oxidation and ketogenesis pathways. Furthermore, vitamins and minerals are related to inflammatory processes regulated by the TLR4/NF-κB and one carbon metabolic pathways. We indicate that diet regulated gene expression of PPARα, NOS, CREB3L3, IRS, and CPT I, altering cellular physiological mechanisms and thus increasing or decreasing the risk of gestational diabetes. The alteration of gene expression can cause inflammation, inhibition of fatty acid transport, or on the contrary help in the modulation of ketogenesis, improve insulin sensitivity, attenuate the effects of glucotoxicity, and others. Therefore, it is critical to comprehend the metabolic changes of pregnant women with gestational diabetes mellitus, to determine nutrients that help in the prevention and treatment of insulin resistance and its long-term consequences.
Collapse
Affiliation(s)
- Patricia Guevara-Ramírez
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación Genética y Genómica, Universidad UTE, Quito, Ecuador
| | - Elius Paz-Cruz
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación Genética y Genómica, Universidad UTE, Quito, Ecuador
| | - Santiago Cadena-Ullauri
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación Genética y Genómica, Universidad UTE, Quito, Ecuador
| | - Viviana A. Ruiz-Pozo
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación Genética y Genómica, Universidad UTE, Quito, Ecuador
| | - Rafael Tamayo-Trujillo
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación Genética y Genómica, Universidad UTE, Quito, Ecuador
| | - Maria L. Felix
- Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Daniel Simancas-Racines
- Centro de Investigación de Salud Pública y Epidemiología Clínica (CISPEC), Universidad UTE, Quito, Ecuador
| | - Ana Karina Zambrano
- Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación Genética y Genómica, Universidad UTE, Quito, Ecuador
| |
Collapse
|
15
|
Ding Y, Wang S, Lu J. Unlocking the Potential: Amino Acids' Role in Predicting and Exploring Therapeutic Avenues for Type 2 Diabetes Mellitus. Metabolites 2023; 13:1017. [PMID: 37755297 PMCID: PMC10535527 DOI: 10.3390/metabo13091017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Diabetes mellitus, particularly type 2 diabetes mellitus (T2DM), imposes a significant global burden with adverse clinical outcomes and escalating healthcare expenditures. Early identification of biomarkers can facilitate better screening, earlier diagnosis, and the prevention of diabetes. However, current clinical predictors often fail to detect abnormalities during the prediabetic state. Emerging studies have identified specific amino acids as potential biomarkers for predicting the onset and progression of diabetes. Understanding the underlying pathophysiological mechanisms can offer valuable insights into disease prevention and therapeutic interventions. This review provides a comprehensive summary of evidence supporting the use of amino acids and metabolites as clinical biomarkers for insulin resistance and diabetes. We discuss promising combinations of amino acids, including branched-chain amino acids, aromatic amino acids, glycine, asparagine and aspartate, in the prediction of T2DM. Furthermore, we delve into the mechanisms involving various signaling pathways and the metabolism underlying the role of amino acids in disease development. Finally, we highlight the potential of targeting predictive amino acids for preventive and therapeutic interventions, aiming to inspire further clinical investigations and mitigate the progression of T2DM, particularly in the prediabetic stage.
Collapse
Affiliation(s)
- Yilan Ding
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.D.); (S.W.)
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.D.); (S.W.)
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.D.); (S.W.)
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
16
|
Safari-Alighiarloo N, Mani-Varnosfaderani A, Madani NH, Tabatabaei SM, Babaei MR, Khamseh ME. Potential metabolic biomarkers of critical limb ischemia in people with type 2 diabetes mellitus. Metabolomics 2023; 19:66. [PMID: 37452163 DOI: 10.1007/s11306-023-02029-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Type 2 diabetes mellitus (T2DM) is a significant risk factor for the development of critical limb ischemia (CLI), the most advanced stage of peripheral arterial disease. The concurrent existence of T2DM and CLI often leads to adverse outcomes, namely limb amputation. OBJECTIVE To identify biomarkers for improving the screening of CLI in high-risk people with T2DM. METHODS We investigated metabolome profiles in serum samples of 113 T2DM people with CLI (n = 23, G2) and without CLI (n = 45, G0: no lower limb stenosis (LLS) and n = 45, G1: LLS < 50%), using hydrogen nuclear magnetic resonance (1H NMR) approach. Principle component analysis (PCA) and partial least squares-discriminant analysis (PLS-DA) were used to analyze 1H NMR data. RESULTS Twenty potential metabolites that could discriminate people with T2DM and CLI (G2) from non-CLI patients without LLS (G0) were determined in serum samples. The correct percent of classification for the PLS-DA model for the test set samples was 85% (n = 20) and 100% (n = 5) for G0 and G2 groups, respectively. Non-CLI patients with LLS < 50% (G1) were projected on the PCA abstract space built using 20 discriminatory metabolites. Eleven people with T2DM and LLS < 50% were prospectively followed, and their ankle-brachial index (ABI) was measured after 4 years. A promising agreement existed between the PCA model's predictions and those obtained by ABI values. CONCLUSION The findings suggest that confirmation of blood potential metabolic biomarkers as a complement to ABI for screening of CLI in a large group of high-risk people with T2DM is needed.
Collapse
Affiliation(s)
- Nahid Safari-Alighiarloo
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran.
- Endocrine and Metabolism Research Institute, Firoozeh Alley, Valiasr Square, Tehran, Iran.
| | - Ahmad Mani-Varnosfaderani
- Chemometrics and Cheminformatics Laboratory, Department of Chemistry, Tarbiat Modares University, Tehran, Iran.
| | - Nahid Hashemi Madani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Seyyed Mohammad Tabatabaei
- Medical Informatics Department, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Babaei
- Department of Interventional Radiology, Firouzgar Hospital, Iran University of Medical Science, Tehran, Iran
| | - Mohammad E Khamseh
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Rafiq T, Stearns JC, Shanmuganathan M, Azab SM, Anand SS, Thabane L, Beyene J, Williams NC, Morrison KM, Teo KK, Britz-McKibbin P, de Souza RJ. Integrative multiomics analysis of infant gut microbiome and serum metabolome reveals key molecular biomarkers of early onset childhood obesity. Heliyon 2023; 9:e16651. [PMID: 37332914 PMCID: PMC10272340 DOI: 10.1016/j.heliyon.2023.e16651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023] Open
Abstract
Evidence supports a complex interplay of gut microbiome and host metabolism as regulators of obesity. The metabolic phenotype and microbial metabolism of host diet may also contribute to greater obesity risk in children early in life. This study aimed to identify features that discriminated overweight/obese from normal weight infants by integrating gut microbiome and serum metabolome profiles. This prospective analysis included 50 South Asian children living in Canada, selected from the SouTh Asian biRth cohorT (START). Serum metabolites were measured by multisegment injection-capillary electrophoresis-mass spectrometry and the relative abundance of bacterial 16S rRNA gene amplicon sequence variant was evaluated at 1 year. Cumulative body mass index (BMIAUC) and skinfold thickness (SSFAUC) scores were calculated from birth to 3 years as the total area under the growth curve (AUC). BMIAUC and/or SSFAUC >85th percentile was used to define overweight/obesity. Data Integration Analysis for Biomarker discovery using Latent cOmponent (DIABLO) was used to identify discriminant features associated with childhood overweight/obesity. The associations between identified features and anthropometric measures were examined using logistic regression. Circulating metabolites including glutamic acid, acetylcarnitine, carnitine, and threonine were positively, whereas γ-aminobutyric acid (GABA), symmetric dimethylarginine (SDMA), and asymmetric dimethylarginine (ADMA) were negatively associated with childhood overweight/obesity. The abundance of the Pseudobutyrivibrio and Lactobacillus genera were positively, and Clostridium sensu stricto 1 and Akkermansia were negatively associated with childhood overweight/obesity. Integrative analysis revealed that Akkermansia was positively whereas Lactobacillus was inversely correlated with GABA and SDMA, and Pseudobutyrivibrio was inversely correlated with GABA. This study provides insights into metabolic and microbial signatures which may regulate satiety, energy metabolism, inflammatory processes, and/or gut barrier function, and therefore, obesity trajectories in childhood. Understanding the functional capacity of these molecular features and potentially modifiable risk factors such as dietary exposures early in life may offer a novel approach for preventing childhood obesity.
Collapse
Affiliation(s)
- Talha Rafiq
- Medical Sciences Graduate Program, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
| | - Jennifer C. Stearns
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Meera Shanmuganathan
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Sandi M. Azab
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Pharmacognosy, Alexandria University, Alexandria 21521, Egypt
| | - Sonia S. Anand
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Lehana Thabane
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Biostatistics Unit, Father Sean O’Sullivan Research Centre, The Research Institute, St Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
- Faculty of Health Sciences, University of Johannesburg, Johannesburg 524, South Africa
| | - Joseph Beyene
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | | - Katherine M. Morrison
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Koon K. Teo
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Russell J. de Souza
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
18
|
Li M, Cheng D, Peng C, Huang Y, Geng J, Huang G, Wang T, Xu A. Therapeutic mechanisms of the medicine and food homology formula Xiao-Ke-Yin on glucolipid metabolic dysfunction revealed by transcriptomics, metabolomics and microbiomics in mice. Chin Med 2023; 18:57. [PMID: 37202792 DOI: 10.1186/s13020-023-00752-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/13/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND In recent decades, the prevalence of metabolic diseases, particularly diabetes, hyperlipidemia, obesity, and non-alcoholic fatty liver disease (NAFLD), has increased dramatically, causing great public health and economic burdens worldwide. Traditional Chinese medicine (TCM) serves as an effective therapeutic choice. Xiao-Ke-Yin (XKY) is a medicine and food homology TCM formula consisting of nine "medicine and food homology" herbs and is used to ameliorate metabolic diseases, such as insulin resistance, diabetes, hyperlipidemia and NAFLD. However, despite its therapeutic potential in metabolic disorders, the underlying mechanisms of this TCM remain unclear. This study aimed to evaluate the therapeutic effectiveness of XKY on glucolipid metabolism dysfunction and explore the potential mechanisms in db/db mice. METHODS To verify the effects of XKY, db/db mice were treated with different concentrations of XKY (5.2, 2.6 and 1.3 g/kg/d) and metformin (0.2 g/kg/d, a hypoglycemic positive control) for 6 weeks, respectively. During this study, we detected the body weight (BW) and fasting blood glucose (FBG), oral glucose tolerance test (OGTT), insulin tolerance test (ITT), daily food intake and water intake. At the end of the animal experiment, blood samples, feces, liver and intestinal tissue of mice in all groups were collected. The potential mechanisms were investigated by using hepatic RNA sequencing, 16 S rRNA sequencing of the gut microbiota and metabolomics analysis. RESULTS XKY efficiently mitigated hyperglycemia, IR, hyperlipidemia, inflammation and hepatic pathological injury in a dose dependent manner. Mechanistically, hepatic transcriptomic analysis showed that XKY treatment significantly reversed the upregulated cholesterol biosynthesis which was further confirmed by RT-qPCR. Additionally, XKY administration maintained intestinal epithelial homeostasis, modulated gut microbiota dysbiosis, and regulated its metabolites. In particular, XKY decreased secondary bile acid producing bacteria (Clostridia and Lachnospircaeae) and lowered fecal secondary bile acid (lithocholic acid (LCA) and deoxycholic acid (DCA)) levels to promote hepatic bile acid synthesis by inhibiting the LCA/DCA-FXR-FGF15 signalling pathway. Furthermore, XKY regulated amino acid metabolism including arginine biosynthesis, alanine, aspartate and glutamate metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, and tryptophan metabolism likely by increasing Bacilli, Lactobacillaceae and Lactobacillus, and decreasing Clostridia, Lachnospircaeae, Tannerellaceae and Parabacteroides abundances. CONCLUSION Taken together, our findings demonstrate that XKY is a promising "medicine food homology" formula for ameliorating glucolipid metabolism and reveal that the therapeutic effects of XKY may due to its downregulation of hepatic cholesterol biosynthesis and modulation of the dysbiosis of the gut microbiota and metabolites.
Collapse
Affiliation(s)
- Mei Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ding Cheng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chuan Peng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yujiao Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Geng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Anlong Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
19
|
Smith ML, Bull CJ, Holmes MV, Davey Smith G, Sanderson E, Anderson EL, Bell JA. Distinct metabolic features of genetic liability to type 2 diabetes and coronary artery disease: a reverse Mendelian randomization study. EBioMedicine 2023; 90:104503. [PMID: 36870196 PMCID: PMC10009453 DOI: 10.1016/j.ebiom.2023.104503] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) and coronary artery disease (CAD) both have known genetic determinants, but the mechanisms through which their associated genetic variants lead to disease onset remain poorly understood. METHODS We used large-scale metabolomics data in a two-sample reverse Mendelian randomization (MR) framework to estimate effects of genetic liability to T2D and CAD on 249 circulating metabolites in the UK Biobank (N = 118,466). We examined the potential for medication use to distort effect estimates by conducting age-stratified metabolite analyses. FINDINGS Using inverse variance weighted (IVW) models, higher genetic liability to T2D was estimated to decrease high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C) (e.g. , HDL-C -0.05 SD; 95% CI -0.07 to -0.03, per doubling of liability), whilst increasing all triglyceride groups and branched chain amino acids (BCAAs). IVW estimates for CAD liability suggested an effect on reducing HDL-C as well as raising very-low density lipoprotein cholesterol (VLDL-C) and LDL-C. In pleiotropy-robust models, T2D liability was still estimated to increase BCAAs, but several estimates for higher CAD liability reversed and supported decreased LDL-C and apolipoprotein-B. Estimated effects of CAD liability differed substantially by age for non-HDL-C traits, with higher CAD liability lowering LDL-C only at older ages when statin use was common. INTERPRETATION Overall, our results support largely distinct metabolic features of genetic liability to T2D and CAD, illustrating both challenges and opportunities for preventing these commonly co-occurring diseases. FUNDING Wellcome Trust [218495/Z/19/Z], UK MRC [MC_UU_00011/1; MC_UU_00011/4], the University of Bristol, Diabetes UK [17/0005587], World Cancer Research Fund [IIG_2019_2009].
Collapse
Affiliation(s)
- Madeleine L Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Caroline J Bull
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; School of Translational Health Sciences, University of Bristol, Bristol, UK
| | - Michael V Holmes
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Eleanor Sanderson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Emma L Anderson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Joshua A Bell
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
20
|
Shah RV, Steffen LM, Nayor M, Reis JP, Jacobs DR, Allen NB, Lloyd-Jones D, Meyer K, Cole J, Piaggi P, Vasan RS, Clish CB, Murthy VL. Dietary metabolic signatures and cardiometabolic risk. Eur Heart J 2023; 44:557-569. [PMID: 36424694 PMCID: PMC10169425 DOI: 10.1093/eurheartj/ehac446] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/23/2022] [Accepted: 07/28/2022] [Indexed: 11/27/2022] Open
Abstract
AIMS Observational studies of diet in cardiometabolic-cardiovascular disease (CM-CVD) focus on self-reported consumption of food or dietary pattern, with limited information on individual metabolic responses to dietary intake linked to CM-CVD. Here, machine learning approaches were used to identify individual metabolic patterns related to diet and relation to long-term CM-CVD in early adulthood. METHODS AND RESULTS In 2259 White and Black adults (age 32.1 ± 3.6 years, 45% women, 44% Black) in the Coronary Artery Risk Development in Young Adults (CARDIA) study, multivariate models were employed to identify metabolite signatures of food group and composite dietary intake across 17 food groups, 2 nutrient groups, and healthy eating index-2015 (HEI2015) diet quality score. A broad array of metabolites associated with diet were uncovered, reflecting food-related components/catabolites (e.g. fish and long-chain unsaturated triacylglycerols), interactions with host features (microbiome), or pathways broadly implicated in CM-CVD (e.g. ceramide/sphingomyelin lipid metabolism). To integrate diet with metabolism, penalized machine learning models were used to define a metabolite signature linked to a putative CM-CVD-adverse diet (e.g. high in red/processed meat, refined grains), which was subsequently associated with long-term diabetes and CVD risk numerically more strongly than HEI2015 in CARDIA [e.g. diabetes: standardized hazard ratio (HR): 1.62, 95% confidence interval (CI): 1.32-1.97, P < 0.0001; CVD: HR: 1.55, 95% CI: 1.12-2.14, P = 0.008], with associations replicated for diabetes (P < 0.0001) in the Framingham Heart Study. CONCLUSION Metabolic signatures of diet are associated with long-term CM-CVD independent of lifestyle and traditional risk factors. Metabolomics improves precision to identify adverse consequences and pathways of diet-related CM-CVD.
Collapse
Affiliation(s)
- Ravi V Shah
- Vanderbilt University Medical Center, Vanderbilt Clinical and Translational Research Center (VTRACC), Nashville, TN, USA
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Matthew Nayor
- Cardiology Division, Boston University School of Medicine, Boston, MA, USA
| | - Jared P Reis
- Epidemiology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Norrina B Allen
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Donald Lloyd-Jones
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Katie Meyer
- Nutrition Department, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Joanne Cole
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Paolo Piaggi
- Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Ramachandran S Vasan
- Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine, Department of Medicine, and Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Venkatesh L Murthy
- Department of Medicine and Radiology, University of Michigan, 1338 Cardiovascular Center, Ann Arbor, MI 48109-5873, USA
| |
Collapse
|
21
|
Metabolomic profiling in kidney cells treated with a sodium glucose-cotransporter 2 inhibitor. Sci Rep 2023; 13:2026. [PMID: 36739309 PMCID: PMC9899225 DOI: 10.1038/s41598-023-28850-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/25/2023] [Indexed: 02/06/2023] Open
Abstract
We aimed to determine the metabolomic profile of kidney cells under high glucose conditions and following sodium-glucose cotransporter 2 (SGLT2) inhibitor treatment. Targeted metabolomics using the Absolute IDQ-p180 kit was applied to quantify metabolites in kidney cells stimulated with high glucose (25 and 50 mM) and treated with SGLT2 inhibitor, dapagliflozin (2 µM). Primary cultured human tubular epithelial cells and podocytes were used to identify the metabolomic profile in high glucose conditions following dapagliflozin treatment. The levels of asparagine, PC ae C34:1, and PC ae C36:2 were elevated in tubular epithelial cells stimulated with 50 mM glucose and were significantly decreased after 2 µM dapagliflozin treatment. The level of PC aa C32:0 was significantly decreased after 50 mM glucose treatment compared with the control, and its level was significantly increased after dapagliflozin treatment in podocytes. The metabolism of glutathione, asparagine and proline was significantly changed in tubular epithelial cells under high-glucose stimulation. And the pathway analysis showed that aminoacyl-tRNA biosynthesis, arginine and proline metabolism, glutathione metabolism, valine, leucine and isoleucine biosynthesis, phenylalanine, tyrosine, and tryptophan biosynthesis, beta-alanine metabolism, phenylalanine metabolism, arginine biosynthesis, alanine, aspartate and glutamate metabolism, glycine, serine and threonine metabolism were altered in tubular epithelial cells after dapagliflozin treatment following 50 mM glucose compared to those treated with 50 mM glucose.
Collapse
|
22
|
Luo S, Zhao Y, Zhu S, Liu L, Cheng K, Ye B, Han Y, Fan J, Xia M. Flavonifractor plautii Protects Against Elevated Arterial Stiffness. Circ Res 2023; 132:167-181. [PMID: 36575982 DOI: 10.1161/circresaha.122.321975] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Dysbiosis of gut microbiota plays a pivotal role in vascular dysfunction and microbial diversity was reported to be inversely correlated with arterial stiffness. However, the causal role of gut microbiota in the progression of arterial stiffness and the specific species along with the molecular mechanisms underlying this change remain largely unknown. METHODS Participants with elevated arterial stiffness and normal controls free of medication were matched for age and sex. The microbial composition and metabolic capacities between the 2 groups were compared with the integration of metagenomics and metabolomics. Subsequently, Ang II (angiotensin II)-induced and humanized mouse model were employed to evaluate the protective effect of Flavonifractor plautii (F plautii) and its main effector cis-aconitic acid. RESULTS Human fecal metagenomic sequencing revealed a significantly high abundance and centrality of F plautii in normal controls, which was absent in the microbial community of subjects with elevated arterial stiffness. Moreover, blood pressure only mediated part of the effect of F plautii on lower arterial stiffness. The microbiome of normal controls exhibited an enhanced capacity for glycolysis and polysaccharide degradation, whereas, those of subjects with increased arterial stiffness were characterized by increased biosynthesis of fatty acids and aromatic amino acids. Integrative analysis with metabolomics profiling further suggested that increased cis-aconitic acid served as the main effector for the protective effect of F plautii against arterial stiffness. Replenishment with F plautii and cis-aconitic acid improved elastic fiber network and reversed increased pulse wave velocity through the suppression of MMP-2 (matrix metalloproteinase-2) and inhibition of MCP-1 (monocyte chemoattractant protein-1) and NF-κB (nuclear factor kappa-B) activation in both Ang II-induced and humanized model of arterial stiffness. CONCLUSIONS Our translational study identifies a novel link between F plautii and arterial function and raises the possibility of sustaining vascular health by targeting gut microbiota.
Collapse
Affiliation(s)
- Shiyun Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition (S.L., Y.Z., S.Z., L.L., B.Y., J.F., M.X.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China
| | - Yawen Zhao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition (S.L., Y.Z., S.Z., L.L., B.Y., J.F., M.X.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China
| | - Shanshan Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition (S.L., Y.Z., S.Z., L.L., B.Y., J.F., M.X.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China
| | - Ludi Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition (S.L., Y.Z., S.Z., L.L., B.Y., J.F., M.X.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China.,Department of Statistics and Epidemiology (L.L., B.Y.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China
| | - Ken Cheng
- XJTLU Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China (K.C., Y.H.)
| | - Bingqi Ye
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition (S.L., Y.Z., S.Z., L.L., B.Y., J.F., M.X.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China.,Department of Statistics and Epidemiology (L.L., B.Y.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China
| | - Yueyuan Han
- XJTLU Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China (K.C., Y.H.)
| | - Jiahua Fan
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition (S.L., Y.Z., S.Z., L.L., B.Y., J.F., M.X.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition (S.L., Y.Z., S.Z., L.L., B.Y., J.F., M.X.), School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, China
| |
Collapse
|
23
|
Zhong J, Wu D, Zeng Y, Wu G, Zheng N, Huang W, Li Y, Tao X, Zhu W, Sheng L, Shen X, Zhang W, Zhu R, Li H. The Microbial and Metabolic Signatures of Patients with Stable Coronary Artery Disease. Microbiol Spectr 2022; 10:e0246722. [PMID: 36354350 PMCID: PMC9769616 DOI: 10.1128/spectrum.02467-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Growing evidence indicates an association between gut dysbiosis and coronary artery disease (CAD). However, the underlying mechanisms relevant to stable CAD (SCAD) pathogenesis, based on microbe-host metabolism interactions, are poorly explored. Here, we constructed a quasi-paired cohort based on the metabolic background of metagenomic samples by the propensity score matching (PSM) principle. Compared to healthy controls (HCs), gut microbiome disturbances were observed in SCAD patients, accompanied by differences in serum metabolome, mainly including elevated acylcarnitine and decreased unsaturated fatty acids in SCAD patients, which implicated the reduced cardiac fatty acid oxidation. Moreover, we identified Ralstonia pickettii as the core strain responsible for impaired microbial homeostasis in SCAD patientsm and may be partly responsible for the decrease of host unsaturated fatty acid levels. These findings highlight the importance of unsaturated fatty acids, R. pickettii, and their interaction in the pathogenesis of SCAD. IMPORTANCE Stable coronary artery disease (SCAD) is an early stage of CAD development. It is important to understand the pathogenesis of SCAD and find out the possible prevention and control targets for delaying the progression of CAD. We observed reduced levels of unsaturated fatty acids (USFAs) in SCAD patients. However, the reduced USFAs may be related to Ralstonia Pickettii, which was the core strain responsible for the impaired gut microbial function in SCAD patients, and further affected the host's cardiovascular health by altering amino acids, vitamin B metabolism, and LPS biosynthesis. These findings not only emphasized the importance of USFAs for cardiovascular health, but also R. Pickettii for maintaining microbial function homeostasis. More importantly, our study revealed, for the first time, that enriched R. Pickettii might be responsible for the reduced USFAs in SCAD patients, which adds new evidence on the role of altered gut microbiota for SCAD formation.
Collapse
Affiliation(s)
- Jing Zhong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Dingfeng Wu
- National Clinical Research Center for Child Health, the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Yuanyuan Zeng
- Cardiology Department of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Gaosong Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ningning Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjin Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Tao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weize Zhu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Sheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoxu Shen
- Cardiology Department of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Ruixin Zhu
- The Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People’s Republic of China
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
24
|
Chen Z, Sun Y, Chen L, Zhang Y, Wang J, Li H, Yan X, Xia L, Yao G. Differences in meat quality between Angus cattle and Xinjiang brown cattle in association with gut microbiota and its lipid metabolism. Front Microbiol 2022; 13:988984. [PMID: 36560955 PMCID: PMC9763702 DOI: 10.3389/fmicb.2022.988984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota plays important roles in mediating fat metabolic events in humans and animals. However, the differences of meat quality traits related to the lipid metabolism (MQT-LM) in association with gut microbiota involving in lipid metabolism have not been well explored between Angus cattle (AG) and Xinjiang brown cattle (BC). Ten heads of 18-month-old uncastrated male AG and BC (5 in each group) raised under the identical conditions were selected to test MQT-LM, i.e., the backfat thickness (BFT), the intramuscular fat (IMF) content, the intramuscular adipocyte areas (IAA), the eye muscle area (EMA), the muscle fiber sectional area (MFSA) and the muscle shear force after sacrifice. The gut microbiota composition and structure with its metabolic function were analyzed by means of metagenomics and metabolomics with rectal feces. The correlation of MQT-LM with the gut microbiota and its metabolites was analyzed. In comparison with AG, BC had significant lower EMA, IMF content and IAA but higher BFT and MFSA. Chao1 and ACE indexes of α-diversity were lower. β-diversity between AG and BC were significantly different. The relative abundance of Bacteroidetes, Prevotella and Blautia and Prevotella copri, Blautia wexlerae, and Ruminococcus gnavus was lower. The lipid metabolism related metabolites, i.e., succinate, oxoglutaric acid, L-aspartic acid and L-glutamic acid were lower, while GABA, L-asparagine and fumaric acid were higher. IMF was positively correlated with Prevotella copri, Blautia wexlerae and Ruminococcus gnavus, and the metabolites succinate, oxoglutaric acid, L-aspartic acid and L-glutamic acid, while negatively with GABA, L-asparagine and fumaric acid. BFT was negatively correlated with Blautia wexlerae and the metabolites succinate, L-aspartic acid and L-glutamic acid, while positively with GABA, L-asparagine and fumaric acid. Prevotella Copri, Blautia wexlerae, and Ruminococcus gnavus was all positively correlated with succinate, oxoglutaric acid, while negatively with L-asparagine and fumaric acid. In conclusion, Prevotella copri, Prevotella intermedia, Blautia wexlerae, and Ruminococcus gnavus may serve as the potential differentiated bacterial species in association with MQT-LM via their metabolites of oxoglutaric acid, succinate, fumaric acid, L-aspartic acid, L-asparagine, L-glutamic acid and GABA between BC and AG.
Collapse
Affiliation(s)
- Zhuo Chen
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yawei Sun
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Lijing Chen
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yang Zhang
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Jinquan Wang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Hongbo Li
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Xiangming Yan
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Lining Xia
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China,Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJKLNDSCHA), Xinjiang Agricultural University, Urumqi, China,*Correspondence: Lining Xia,
| | - Gang Yao
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China,Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJKLNDSCHA), Xinjiang Agricultural University, Urumqi, China,Gang Yao,
| |
Collapse
|
25
|
A Study of the Metabolic Pathways Affected by Gestational Diabetes Mellitus: Comparison with Type 2 Diabetes. Diagnostics (Basel) 2022; 12:diagnostics12112881. [PMID: 36428943 PMCID: PMC9689375 DOI: 10.3390/diagnostics12112881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) remains incompletely understood and increases the risk of developing Diabetes mellitus type 2 (DM2). Metabolomics provides insights etiology and pathogenesis of disease and discovery biomarkers for accurate detection. Nuclear magnetic resonance (NMR) spectroscopy is a key platform defining metabolic signatures in intact serum/plasma. In the present study, we used NMR-based analysis of macromolecules free-serum to accurately characterize the altered metabolic pathways of GDM and assessing their similarities to DM2. Our findings could contribute to the understanding of the pathophysiology of GDM and help in the identification of metabolomic markers of the disease. METHODS Sixty-two women with GDM matched with seventy-seven women without GDM (control group). 1H NMR serum spectra were acquired on an 11.7 T Bruker Avance DRX NMR spectrometer. RESULTS We identified 55 metabolites in both groups, 25 of which were significantly altered in the GDM group. GDM group showed elevated levels of ketone bodies, 2-hydroxybutyrate and of some metabolic intermediates of branched-chain amino acids (BCAAs) and significantly lower levels of metabolites of one-carbon metabolism, energy production, purine metabolism, certain amino acids, 3-methyl-2-oxovalerate, ornithine, 2-aminobutyrate, taurine and trimethylamine N-oxide. CONCLUSION Metabolic pathways affected in GDM were beta-oxidation, ketone bodies metabolism, one-carbon metabolism, arginine and ornithine metabolism likewise in DM2, whereas BCAAs catabolism and aromatic amino acids metabolism were affected, but otherwise than in DM2.
Collapse
|
26
|
Yan Y, Smith E, Melander O, Ottosson F. The association between plasma metabolites and future risk of all-cause mortality. J Intern Med 2022; 292:804-815. [PMID: 35796403 PMCID: PMC9796397 DOI: 10.1111/joim.13540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Metabolite profiles provide snapshots of the overall effect of numerous exposures accumulated over life courses, which may lead to health outcomes in the future. OBJECTIVE We hypothesized that the risk of all-cause mortality is linked to alterations in metabolism earlier in life, which are reflected in plasma metabolite profiles. We aimed to identify plasma metabolites associated with future risk of all-cause mortality. METHODS Through metabolomics, 110 metabolites were measured in 3833 individuals from the Malmö Diet and Cancer-Cardiovascular Cohort (MDC-CC). A total of 1574 deaths occurred within an average follow-up time of 22.2 years. Metabolites that were significantly associated with all-cause mortality in MDC-CC were replicated in 1500 individuals from Malmö Preventive Project re-examination (MPP), among whom 715 deaths occurred within an average follow-up time of 11.3 years. RESULTS Twenty two metabolites were significantly associated with all-cause mortality in MDC-CC, of which 13 were replicated in MPP. Levels of trigonelline, glutamate, dimethylglycine, C18-1-carnitine, C16-1-carnitine, C14-1-carnitine, and 1-methyladenosine were associated with an increased risk, while levels of valine, tryptophan, lysine, leucine, histidine, and 2-aminoisobutyrate were associated with a decreased risk of all-cause mortality. CONCLUSION We used metabolomics in two Swedish prospective cohorts and identified replicable associations between 13 metabolites and future risk of all-cause mortality. Novel associations between five metabolites-C18-1-carnitine, C16-1-carnitine, C14-1-carnitine, trigonelline, and 2-aminoisobutyrate-and all-cause mortality were discovered. These findings suggest potential new biomarkers for the prediction of mortality and provide insights for understanding the biochemical pathways that lead to mortality.
Collapse
Affiliation(s)
- Yingxiao Yan
- Department of Clinical Science, Lund University, Malmö, Sweden.,Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Einar Smith
- Department of Clinical Science, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Science, Lund University, Malmö, Sweden
| | - Filip Ottosson
- Department of Clinical Science, Lund University, Malmö, Sweden.,Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
27
|
Ramzan I, Ardavani A, Vanweert F, Mellett A, Atherton PJ, Idris I. The Association between Circulating Branched Chain Amino Acids and the Temporal Risk of Developing Type 2 Diabetes Mellitus: A Systematic Review & Meta-Analysis. Nutrients 2022; 14:4411. [PMID: 36297095 PMCID: PMC9610746 DOI: 10.3390/nu14204411] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: Recent studies have concluded that elevated circulating branched chain amino acids (BCAA) are associated with the pathogenesis of type 2 diabetes mellitus (T2DM) and obesity. However, the development of this association over time and the quantification of the strength of this association for individual BCAAs prior to T2DM diagnosis remains unexplored. Methods: A systematic search was conducted using the Healthcare Databases Advance Search (HDAS) via the National Institute for Health and Care Excellence (NICE) website. The data sources included EMBASE, MEDLINE and PubMed for all papers from inception until November 2021. Nine studies were identified in this systematic review and meta-analysis. Stratification was based on follow-up times (0−6, 6−12 and 12 or more years) and controlling of body mass index (BMI) through the specific assessment of overweight cohorts was also undertaken. Results: The meta-analysis revealed a statistically significant positive association between BCAA concentrations and the development of T2DM, with valine OR = 2.08 (95% CI = 2.04−2.12, p < 0.00001), leucine OR = 2.25 (95% CI = 1.76−2.87, p < 0.00001) and isoleucine OR = 2.12, 95% CI = 2.00−2.25, p < 0.00001. In addition, we demonstrated a positive consistent temporal association between circulating BCAA levels and the risk of developing T2DM with differentials in the respective follow-up times of 0−6 years, 6−12 years and ≥12 years follow-up for valine (OR = 2.08, 1.86 and 2.14, p < 0.05 each), leucine (OR = 2.10, 2.25 and 2.16, p < 0.05 each) and isoleucine (OR = 2.12, 1.90 and 2.16, p < 0.05 each) demonstrated. Conclusion: Plasma BCAA concentrations are associated with T2DM incidence across all temporal subgroups. We suggest the potential utility of BCAAs as an early biomarker for T2DM irrespective of follow-up time.
Collapse
Affiliation(s)
- Imran Ramzan
- Clinical, Metabolic and Molecular Physiology Research Group, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 6DT, UK
- National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre, Nottingham NG7 2UH, UK
| | - Arash Ardavani
- Clinical, Metabolic and Molecular Physiology Research Group, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 6DT, UK
- National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre, Nottingham NG7 2UH, UK
| | - Froukje Vanweert
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aisling Mellett
- Clinical, Metabolic and Molecular Physiology Research Group, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 6DT, UK
- School of Agriculture and Food Science, Agriculture and Food Science Centre, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Philip J. Atherton
- Clinical, Metabolic and Molecular Physiology Research Group, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 6DT, UK
- National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre, Nottingham NG7 2UH, UK
| | - Iskandar Idris
- Clinical, Metabolic and Molecular Physiology Research Group, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 6DT, UK
- National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
28
|
Hosseinkhani S, Emamgholipour S, Salari P, Khalagi K, Shirani S, Najjar N, Larijani B, Pasalar P, Razi F. Evaluating the association between amino acid and acylcarnitine profiles and different levels of coronary artery disease risk in postmenopausal women using targeted metabolomics technique. Menopause 2022; 29:1062-1070. [PMID: 35969879 DOI: 10.1097/gme.0000000000002016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Postmenopausal women are at increased risk of developing coronary artery disease (CAD). Metabolomic approaches aim at discovering more helpful biomarkers of CAD to reduce the disease burden in the future. Here, we intend to find potential blood biomarkers, amino acids, and acylcarnitines in postmenopausal women with different severity of CAD by using high-throughput methods. METHOD This cross-sectional study was performed on postmenopausal women ( n = 183) who underwent coronary CT scans. Coronary artery calcium scoring (CACS) was assessed to detect plaque burden and degree of coronary artery obstruction. The participants were divided into three groups based on the score as follows (i) "low CACS" ( n = 96); a score of 0 to 10, (ii) "medium CACS" ( n = 35); a score between 11 and 100 and (iii) "high CACS" ( n = 52); a score greater than 100. Metabolites, including amino acids and acylcarnitines, were quantified using a targeted mass spectrometry method in serum samples. The association between metabolites and disease status was evaluated using univariate and multivariate regression analyses with adjustment for confounding factors. Factor analysis was used to deal with multiple comparisons. RESULTS Metabolites, including proline, glutamic acid, and phenylalanine, were significantly lower in the high CACS group than the low CACS one. Also, a lower level of lysine and phenylalanine in high CACS compared with medium one was observed. Concerning acylcarnitines, it was found that C4 and C8:1 significantly were higher in women with high CACS. The logistic regression analysis revealed that the circulating levels of these metabolites (except C4) were associated with the presence of coronary artery calcification independently of age, body mass index, and time of menopause. Also, the amino acids were associated independently of medication and diabetes. CONCLUSIONS The present study indicated that circulating levels of amino acids and acylcarnitines profile in postmenopausal women are partly associated with the severity of CAD in these participants.
Collapse
Affiliation(s)
- Shaghayegh Hosseinkhani
- From the Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solaleh Emamgholipour
- From the Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pooneh Salari
- Medical Ethics and History of Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shapour Shirani
- Imaging Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Farideh Razi
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Ottosson F, Smith E, Ericson U, Brunkwall L, Orho-Melander M, Di Somma S, Antonini P, Nilsson PM, Fernandez C, Melander O. Metabolome-Defined Obesity and the Risk of Future Type 2 Diabetes and Mortality. Diabetes Care 2022; 45:1260-1267. [PMID: 35287165 PMCID: PMC9174969 DOI: 10.2337/dc21-2402] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/14/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Obesity is a key risk factor for type 2 diabetes; however, up to 20% of patients are normal weight. Our aim was to identify metabolite patterns reproducibly predictive of BMI and subsequently to test whether lean individuals who carry an obese metabolome are at hidden high risk of obesity-related diseases, such as type 2 diabetes. RESEARCH DESIGN AND METHODS Levels of 108 metabolites were measured in plasma samples of 7,663 individuals from two Swedish and one Italian population-based cohort. Ridge regression was used to predict BMI using the metabolites. Individuals with a predicted BMI either >5 kg/m2 higher (overestimated) or lower (underestimated) than their actual BMI were characterized as outliers and further investigated for obesity-related risk factors and future risk of type 2 diabetes and mortality. RESULTS The metabolome could predict BMI in all cohorts (r2 = 0.48, 0.26, and 0.19). The overestimated group had a BMI similar to individuals correctly predicted as normal weight, had a similar waist circumference, were not more likely to change weight over time, but had a two times higher risk of future type 2 diabetes and an 80% increased risk of all-cause mortality. These associations remained after adjustments for obesity-related risk factors and lifestyle parameters. CONCLUSIONS We found that lean individuals with an obesity-related metabolome have an increased risk for type 2 diabetes and all-cause mortality compared with lean individuals with a healthy metabolome. Metabolomics may be used to identify hidden high-risk individuals to initiate lifestyle and pharmacological interventions.
Collapse
Affiliation(s)
- Filip Ottosson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Einar Smith
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ulrika Ericson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | | | - Salvatore Di Somma
- Department of Medical-Surgery Sciences and Translational Medicine, University of Rome Sapienza, Rome, Italy
- GREAT Health Sciences, Rome, Italy
| | | | | | | | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
30
|
Perng W, Hivert MF, Michelotti G, Oken E, Dabelea D. Metabolomic Predictors of Dysglycemia in Two U.S. Youth Cohorts. Metabolites 2022; 12:404. [PMID: 35629908 PMCID: PMC9147862 DOI: 10.3390/metabo12050404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 01/27/2023] Open
Abstract
Here, we seek to identify metabolite predictors of dysglycemia in youth. In the discovery analysis among 391 youth in the Exploring Perinatal Outcomes among CHildren (EPOCH) cohort, we used reduced rank regression (RRR) to identify sex-specific metabolite predictors of impaired fasting glucose (IFG) and elevated fasting glucose (EFG: Q4 vs. Q1 fasting glucose) 6 years later and compared the predictive capacity of four models: Model 1: ethnicity, parental diabetes, in utero exposure to diabetes, and body mass index (BMI); Model 2: Model 1 covariates + baseline waist circumference, insulin, lipids, and Tanner stage; Model 3: Model 2 + baseline fasting glucose; Model 4: Model 3 + baseline metabolite concentrations. RRR identified 19 metabolite predictors of fasting glucose in boys and 14 metabolite predictors in girls. Most compounds were on lipid, amino acid, and carbohydrate metabolism pathways. In boys, no improvement in aurea under the receiver operating characteristics curve AUC occurred until the inclusion of metabolites in Model 4, which increased the AUC for prediction of IFG (7.1%) from 0.81 to 0.97 (p = 0.002). In girls, %IFG was too low for regression analysis (3.1%), but we found similar results for EFG. We replicated the results among 265 youth in the Project Viva cohort, focusing on EFG due to low %IFG, suggesting that the metabolite profiles identified herein have the potential to improve the prediction of glycemia in youth.
Collapse
Affiliation(s)
- Wei Perng
- Lifcourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA; (M.-F.H.); (E.O.)
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA; (M.-F.H.); (E.O.)
- Department of Nutrition, T. H. Chan Harvard School of Public Health, Boston, MA 02115, USA
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
31
|
Smith E, Ericson U, Hellstrand S, Orho-Melander M, Nilsson PM, Fernandez C, Melander O, Ottosson F. A healthy dietary metabolic signature is associated with a lower risk for type 2 diabetes and coronary artery disease. BMC Med 2022; 20:122. [PMID: 35443726 PMCID: PMC9022292 DOI: 10.1186/s12916-022-02326-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The global burden of cardiovascular disease and type 2 diabetes could be decreased by improving dietary factors, but identification of groups suitable for interventional approaches can be difficult. Reporting of dietary intake is prone to errors, and measuring of metabolites has shown promise in determining habitual dietary intake. Our aim is to create a metabolic signature that is associated with healthy eating and test if it associates with type 2 diabetes and coronary artery disease risk. METHODS Using plasma metabolite data consisting of 111 metabolites, partial least square (PLS) regression was used to identify a metabolic signature associated with a health conscious food pattern in the Malmö Offspring Study (MOS, n = 1538). The metabolic signature's association with dietary intake was validated in the Malmö Diet and Cancer study (MDC, n = 2521). The associations between the diet-associated metabolic signature and incident type 2 diabetes and coronary artery disease (CAD) were tested using Cox regression in MDC and logistic regression in Malmö Preventive Project (MPP, n = 1083). Modelling was conducted unadjusted (model 1), adjusted for potential confounders (model 2) and additionally for potential mediators (model 3). RESULTS The metabolic signature was associated with lower risk for type 2 diabetes in both MDC (hazard ratio: 0.58, 95% CI 0.52-0.66, per 1 SD increment of the metabolic signature) and MPP (odds ratio: 0.54, 95% CI 0.44-0.65 per 1 SD increment of the metabolic signature) in model 2. The results were attenuated but remained significant in model 3 in both MDC (hazard ratio 0.73, 95% CI 0.63-0.83) and MPP (odds ratio 0.70, 95% CI 0.55-0.88). The diet-associated metabolic signature was also inversely associated with lower risk of CAD in both MDC and MPP in model 1, but the association was non-significant in model 3. CONCLUSIONS In this proof-of-concept study, we identified a healthy diet-associated metabolic signature, which was inversely associated with future risk for type 2 diabetes and coronary artery disease in two different cohorts. The association with diabetes was independent of traditional risk factors and might illustrate an effect of health conscious dietary intake on cardiometabolic health.
Collapse
Affiliation(s)
- Einar Smith
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden.
| | - Ulrika Ericson
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden
| | - Sophie Hellstrand
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden.,Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Céline Fernandez
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden.,Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Filip Ottosson
- Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, 91-12-027, SE-214 28, Malmö, Sweden.,Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
32
|
Morze J, Wittenbecher C, Schwingshackl L, Danielewicz A, Rynkiewicz A, Hu FB, Guasch-Ferré M. Metabolomics and Type 2 Diabetes Risk: An Updated Systematic Review and Meta-analysis of Prospective Cohort Studies. Diabetes Care 2022; 45:1013-1024. [PMID: 35349649 PMCID: PMC9016744 DOI: 10.2337/dc21-1705] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/20/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Due to the rapidly increasing availability of metabolomics data in prospective studies, an update of the meta evidence on metabolomics and type 2 diabetes risk is warranted. PURPOSE To conduct an updated systematic review and meta-analysis of plasma, serum, and urine metabolite markers and incident type 2 diabetes. DATA SOURCES We searched PubMed and Embase until 6 March 2021. STUDY SELECTION We selected prospective observational studies where investigators used high-throughput techniques to investigate the relationship between plasma, serum, or urine metabolites and incident type 2 diabetes. DATA EXTRACTION Baseline metabolites per-SD risk estimates and 95% CIs for incident type 2 diabetes were extracted from all eligible studies. DATA SYNTHESIS A total of 61 reports with 71,196 participants and 11,771 type 2 diabetes cases/events were included in the updated review. Meta-analysis was performed for 412 metabolites, of which 123 were statistically significantly associated (false discovery rate-corrected P < 0.05) with type 2 diabetes risk. Higher plasma and serum levels of certain amino acids (branched-chain, aromatic, alanine, glutamate, lysine, and methionine), carbohydrates and energy-related metabolites (mannose, trehalose, and pyruvate), acylcarnitines (C4-DC, C4-OH, C5, C5-OH, and C8:1), the majority of glycerolipids (di- and triacylglycerols), (lyso)phosphatidylethanolamines, and ceramides included in meta-analysis were associated with higher risk of type 2 diabetes (hazard ratio 1.07-2.58). Higher levels of glycine, glutamine, betaine, indolepropionate, and (lyso)phosphatidylcholines were associated with lower type 2 diabetes risk (hazard ratio 0.69-0.90). LIMITATIONS Substantial heterogeneity (I2 > 50%, τ2 > 0.1) was observed for some of the metabolites. CONCLUSIONS Several plasma and serum metabolites, including amino acids, lipids, and carbohydrates, are associated with type 2 diabetes risk.
Collapse
Affiliation(s)
- Jakub Morze
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cardiology and Internal Medicine, School of Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
- Department of Human Nutrition, Faculty of Food Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Clemens Wittenbecher
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Lukas Schwingshackl
- Institute for Evidence in Medicine, Medical Centre—University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna Danielewicz
- Department of Human Nutrition, Faculty of Food Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Andrzej Rynkiewicz
- Department of Cardiology and Internal Medicine, School of Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Frank B. Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
33
|
Murthy VL, Nayor M, Carnethon M, Reis JP, Lloyd-Jones D, Allen NB, Kitchen R, Piaggi P, Steffen LM, Vasan RS, Freedman JE, Clish CB, Shah RV. Circulating metabolite profile in young adulthood identifies long-term diabetes susceptibility: the Coronary Artery Risk Development in Young Adults (CARDIA) study. Diabetologia 2022; 65:657-674. [PMID: 35041022 PMCID: PMC8969893 DOI: 10.1007/s00125-021-05641-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS The aim of this work was to define metabolic correlates and pathways of diabetes pathogenesis in young adults during a subclinical latent phase of diabetes development. METHODS We studied 2083 young adults of Black and White ethnicity in the prospective observational cohort Coronary Artery Risk Development in Young Adults (CARDIA) study (mean ± SD age 32.1 ± 3.6 years; 43.9% women; 42.7% Black; mean ± SD BMI 25.6 ± 4.9 kg/m2) and 1797 Framingham Heart Study (FHS) participants (mean ± SD age 54.7 ± 9.7 years; 52.1% women; mean ± SD BMI 27.4 ± 4.8 kg/m2), examining the association of comprehensive metabolite profiles with endophenotypes of diabetes susceptibility (adipose and muscle tissue phenotypes and systemic inflammation). Statistical learning techniques and Cox regression were used to identify metabolite signatures of incident diabetes over a median of nearly two decades of follow-up across both cohorts. RESULTS We identified known and novel metabolites associated with endophenotypes that delineate the complex pathophysiological architecture of diabetes, spanning mechanisms of muscle insulin resistance, inflammatory lipid signalling and beta cell metabolism (e.g. bioactive lipids, amino acids and microbe- and diet-derived metabolites). Integrating endophenotypes of diabetes susceptibility with the metabolome generated two multi-parametric metabolite scores, one of which (a proinflammatory adiposity score) was associated with incident diabetes across the life course in participants from both the CARDIA study (young adults; HR in a fully adjusted model 2.10 [95% CI 1.72, 2.55], p<0.0001) and FHS (middle-aged and older adults; HR 1.33 [95% CI 1.14, 1.56], p=0.0004). A metabolite score based on the outcome of diabetes was strongly related to diabetes in CARDIA study participants (fully adjusted HR 3.41 [95% CI 2.85, 4.07], p<0.0001) but not in the older FHS population (HR 1.15 [95% CI 0.99, 1.33], p=0.07). CONCLUSIONS/INTERPRETATION Selected metabolic abnormalities in young adulthood identify individuals with heightened diabetes risk independent of race, sex and traditional diabetes risk factors. These signatures replicate across the life course.
Collapse
Affiliation(s)
- Venkatesh L Murthy
- Department of Medicine and Radiology, University of Michigan, Ann Arbor, MI, USA.
| | - Matthew Nayor
- Section of Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | - Jared P Reis
- National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | | | | | - Robert Kitchen
- Simches Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Paolo Piaggi
- Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Lyn M Steffen
- University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Jane E Freedman
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
34
|
Izundegui DG, Nayor M. Metabolomics of Type 1 and Type 2 Diabetes: Insights into Risk Prediction and Mechanisms. Curr Diab Rep 2022; 22:65-76. [PMID: 35113332 PMCID: PMC8934149 DOI: 10.1007/s11892-022-01449-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Metabolomics enables rapid interrogation of widespread metabolic processes making it well suited for studying diabetes. Here, we review the current status of metabolomic investigation in diabetes, highlighting its applications for improving risk prediction and mechanistic understanding. RECENT FINDINGS Findings of metabolite associations with type 2 diabetes risk have confirmed experimental observations (e.g., branched-chain amino acids) and also pinpointed novel pathways of diabetes risk (e.g., dimethylguanidino valeric acid). In type 1 diabetes, abnormal metabolite patterns are observed prior to the development of autoantibodies and hyperglycemia. Diabetes complications display specific metabolite signatures that are distinct from the metabolic derangements of diabetes and differ across vascular beds. Lastly, metabolites respond acutely to pharmacologic treatment, providing opportunities to understand inter-individual treatment responses. Metabolomic studies have elucidated biological mechanisms underlying diabetes development, complications, and therapeutic response. While not yet ready for clinical translation, metabolomics is a powerful and promising precision medicine tool.
Collapse
Affiliation(s)
| | - Matthew Nayor
- Sections of Cardiology and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, 72 E Concord Street, Suite L-516, Boston, MA, 02118, USA.
| |
Collapse
|
35
|
Roberts I, Wright Muelas M, Taylor JM, Davison AS, Xu Y, Grixti JM, Gotts N, Sorokin A, Goodacre R, Kell DB. Untargeted metabolomics of COVID-19 patient serum reveals potential prognostic markers of both severity and outcome. Metabolomics 2021; 18:6. [PMID: 34928464 PMCID: PMC8686810 DOI: 10.1007/s11306-021-01859-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The diagnosis of COVID-19 is normally based on the qualitative detection of viral nucleic acid sequences. Properties of the host response are not measured but are key in determining outcome. Although metabolic profiles are well suited to capture host state, most metabolomics studies are either underpowered, measure only a restricted subset of metabolites, compare infected individuals against uninfected control cohorts that are not suitably matched, or do not provide a compact predictive model. OBJECTIVES Here we provide a well-powered, untargeted metabolomics assessment of 120 COVID-19 patient samples acquired at hospital admission. The study aims to predict the patient's infection severity (i.e., mild or severe) and potential outcome (i.e., discharged or deceased). METHODS High resolution untargeted UHPLC-MS/MS analysis was performed on patient serum using both positive and negative ionization modes. A subset of 20 intermediary metabolites predictive of severity or outcome were selected based on univariate statistical significance and a multiple predictor Bayesian logistic regression model was created. RESULTS The predictors were selected for their relevant biological function and include deoxycytidine and ureidopropionate (indirectly reflecting viral load), kynurenine (reflecting host inflammatory response), and multiple short chain acylcarnitines (energy metabolism) among others. Currently, this approach predicts outcome and severity with a Monte Carlo cross validated area under the ROC curve of 0.792 (SD 0.09) and 0.793 (SD 0.08), respectively. A blind validation study on an additional 90 patients predicted outcome and severity at ROC AUC of 0.83 (CI 0.74-0.91) and 0.76 (CI 0.67-0.86). CONCLUSION Prognostic tests based on the markers discussed in this paper could allow improvement in the planning of COVID-19 patient treatment.
Collapse
Affiliation(s)
- Ivayla Roberts
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Marina Wright Muelas
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| | - Joseph M Taylor
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Royal Liverpool University Hospitals Trust, Liverpool, UK
| | - Andrew S Davison
- Department of Clinical Biochemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Royal Liverpool University Hospitals Trust, Liverpool, UK
| | - Yun Xu
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Metabolomics Research (CMR), Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Justine M Grixti
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Nigel Gotts
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Metabolomics Research (CMR), Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Anatolii Sorokin
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Royston Goodacre
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Centre for Metabolomics Research (CMR), Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Douglas B Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Chemitorvet, 2000, Kgs Lyngby, Denmark.
| |
Collapse
|
36
|
His M, Viallon V, Dossus L, Schmidt JA, Travis RC, Gunter MJ, Overvad K, Kyrø C, Tjønneland A, Lécuyer L, Rothwell JA, Severi G, Johnson T, Katzke V, Schulze MB, Masala G, Sieri S, Panico S, Tumino R, Macciotta A, Boer JMA, Monninkhof EM, Olsen KS, Nøst TH, Sandanger TM, Agudo A, Sánchez MJ, Amiano P, Colorado-Yohar SM, Ardanaz E, Vidman L, Winkvist A, Heath AK, Weiderpass E, Huybrechts I, Rinaldi S. Lifestyle correlates of eight breast cancer-related metabolites: a cross-sectional study within the EPIC cohort. BMC Med 2021; 19:312. [PMID: 34886862 PMCID: PMC8662901 DOI: 10.1186/s12916-021-02183-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/09/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Metabolomics is a promising molecular tool for identifying novel etiological pathways leading to cancer. In an earlier prospective study among pre- and postmenopausal women not using exogenous hormones, we observed a higher risk of breast cancer associated with higher blood concentrations of one metabolite (acetylcarnitine) and a lower risk associated with higher blood concentrations of seven others (arginine, asparagine, phosphatidylcholines (PCs) aa C36:3, ae C34:2, ae C36:2, ae C36:3, and ae C38:2). METHODS To identify determinants of these breast cancer-related metabolites, we conducted a cross-sectional analysis to identify their lifestyle and anthropometric correlates in 2358 women, who were previously included as controls in case-control studies nested within the European Prospective Investigation into Cancer and Nutrition cohort and not using exogenous hormones at blood collection. Associations of each metabolite concentration with 42 variables were assessed using linear regression models in a discovery set of 1572 participants. Significant associations were evaluated in a validation set (n = 786). RESULTS For the metabolites previously associated with a lower risk of breast cancer, concentrations of PCs ae C34:2, C36:2, C36:3, and C38:2 were negatively associated with adiposity and positively associated with total and saturated fat intakes. PC ae C36:2 was also negatively associated with alcohol consumption and positively associated with two scores reflecting adherence to a healthy lifestyle. Asparagine concentration was negatively associated with adiposity. Arginine and PC aa C36:3 concentrations were not associated to any of the factors examined. For the metabolite previously associated with a higher risk of breast cancer, acetylcarnitine, a positive association with age was observed. CONCLUSIONS These associations may indicate possible mechanisms underlying associations between lifestyle and anthropometric factors, and risk of breast cancer. Further research is needed to identify potential non-lifestyle correlates of the metabolites investigated.
Collapse
Affiliation(s)
- Mathilde His
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, 150 cours Albert Thomas, 69372, CEDEX 08, Lyon, France
| | - Vivian Viallon
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, 150 cours Albert Thomas, 69372, CEDEX 08, Lyon, France
| | - Laure Dossus
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, 150 cours Albert Thomas, 69372, CEDEX 08, Lyon, France
| | - Julie A Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Marc J Gunter
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, 150 cours Albert Thomas, 69372, CEDEX 08, Lyon, France
| | - Kim Overvad
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Cecilie Kyrø
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Public Health, Section of Environmental Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lucie Lécuyer
- Université Paris-Saclay, UVSQ, Inserm, CESP U1018, "Exposome and Heredity" team, Gustave Roussy, Villejuif, France
| | - Joseph A Rothwell
- Université Paris-Saclay, UVSQ, Inserm, CESP U1018, "Exposome and Heredity" team, Gustave Roussy, Villejuif, France
| | - Gianluca Severi
- Université Paris-Saclay, UVSQ, Inserm, CESP U1018, "Exposome and Heredity" team, Gustave Roussy, Villejuif, France
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Theron Johnson
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Katzke
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Giovanna Masala
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Instituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Salvatore Panico
- Dipartimento Di Medicina Clinica E Chirurgia, Federico Ii University, Naples, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, Provincial Health Authority (ASP 7) Ragusa, Ragusa, Italy
| | - Alessandra Macciotta
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Jolanda M A Boer
- Center for Nutrition, Prevention, and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, 3720, BA, the Netherlands
| | - Evelyn M Monninkhof
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Karina Standahl Olsen
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Therese H Nøst
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Torkjel M Sandanger
- Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Antonio Agudo
- Unit of Nutrition and Cancer, Catalan Institute of Oncology - ICO, L'Hospitalet de Llobregat, Spain
- Nutrition and Cancer Group; Epidemiology, Public Health, Cancer Prevention and Palliative Care Program; Bellvitge Biomedical Research Institute - IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
| | - Pilar Amiano
- Ministry of Health of the Basque Government, Sub-Directorate for Public Health and Addictions of Gipuzkoa, San Sebastián, Spain
- Biodonostia Health Research Institute, Group of Epidemiology of Chronic and Communicable Diseases, San Sebastián, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Sandra M Colorado-Yohar
- CIBER Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Research Group on Demography and Health, National Faculty of Public Health, University of Antioquia, Medellín, Colombia
| | - Eva Ardanaz
- CIBER Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Linda Vidman
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Anna Winkvist
- Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Elisabete Weiderpass
- International Agency for Research on Cancer (IARC/WHO), Office of the Director, Lyon, France
| | - Inge Huybrechts
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, 150 cours Albert Thomas, 69372, CEDEX 08, Lyon, France
| | - Sabina Rinaldi
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, 150 cours Albert Thomas, 69372, CEDEX 08, Lyon, France.
| |
Collapse
|
37
|
Sun Y, Lu YK, Gao HY, Yan YX. Effect of Metabolite Levels on Type 2 Diabetes Mellitus and Glycemic Traits: A Mendelian Randomization Study. J Clin Endocrinol Metab 2021; 106:3439-3447. [PMID: 34363473 DOI: 10.1210/clinem/dgab581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To assess the causal associations of plasma levels of metabolites with type 2 diabetes mellitus (T2DM) and glycemic traits. METHODS Two-sample mendelian randomization (MR) was conducted to assess the causal associations. Genetic variants strongly associated with metabolites at genome-wide significance level (P < 5 × 10-8) were selected from public genome-wide association studies, and single-nucleotide polymorphisms of outcomes were obtained from the Diabetes Genetics Replication and Meta-analysis consortium for T2DM and from the Meta-Analyses of Glucose and Insulin-related Traits Consortium for fasting glucose, insulin, and glycated hemoglobin (HbA1c). The Wald ratio and inverse-variance weighted methods were used for analyses, and MR-Egger was used for sensitivity analysis. RESULTS The β estimates per 1-SD increase of arachidonic acid (AA) level was 0.16 (95% CI, 0.078-0.242; P < 0.001). Genetic predisposition to higher plasma AA levels were associated with higher fasting glucose levels (β 0.10 [95% CI, 0.064-0.134], P < 0.001), higher HbA1c levels (β 0.04 [95% CI, 0.027-0.061]), and lower fasting insulin levels (β -0.025 [95% CI, -0.047 to -0.002], P = 0.033). Besides, 2-hydroxybutyric acid (2-HBA) might have a positive causal effect on glycemic traits. CONCLUSIONS Our findings suggest that AA and 2-HBA may have causal associations on T2DM and glycemic traits. This is beneficial for clarifying the pathogenesis of T2DM, which would be valuable for early identification and prevention for T2DM.
Collapse
Affiliation(s)
- Yue Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, 100069, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing, 100069, China
| | - Ya-Ke Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, 100069, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing, 100069, China
| | - Hao-Yu Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, 100069, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing, 100069, China
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, 100069, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing, 100069, China
| |
Collapse
|
38
|
Jauhiainen R, Vangipurapu J, Laakso A, Kuulasmaa T, Kuusisto J, Laakso M. The Association of 9 Amino Acids With Cardiovascular Events in Finnish Men in a 12-Year Follow-up Study. J Clin Endocrinol Metab 2021; 106:3448-3454. [PMID: 34346487 PMCID: PMC8634085 DOI: 10.1210/clinem/dgab562] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS To investigate the significance of 9 amino acids as risk factors for incident cardiovascular disease events in 9584 Finnish men. MATERIALS AND METHODS A total of 9584 men (age 57.4 ± 7.0 years, body mass index 27.2 ± 4.2 kg/m2) from the Metabolic Syndrome in Men study without cardiovascular disease and type 1 diabetes at baseline were included in this study. A total of 662 coronary artery disease (CAD) events, 394 ischemic stroke events, and 966 cardiovascular disease (CVD; CAD and stroke combined) events were recorded in a 12.3-year follow-up. Amino acids were measured using nuclear magnetic resonance platform. RESULTS In Cox regression analysis, phenylalanine and tyrosine were significantly associated with increased risk of CAD and CVD events, and phenylalanine with increased risk of ischemic stroke after the adjustment for confounding factors. Glutamine was significantly associated with decreased risk of stroke and CVD events and nominally with CAD events. Alanine was nominally associated with CAD events. CONCLUSION We identified alanine as a new amino acid associated with increased risk of CAD and glutamine as a new amino acid associated with decreased risk of ischemic stroke. We also confirmed that phenylalanine and tyrosine were associated with CAD, ischemic stroke, and CVD events.
Collapse
Affiliation(s)
- Raimo Jauhiainen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Annamaria Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Teemu Kuulasmaa
- Institute of Biomedicine, Bioinformatics Center, University of Eastern Finland, 70210 Kuopio, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, and Kuopio University Hospital, 70210 Kuopio, Finland
- Correspondence: Markku Laakso, MD, PhD, Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland.
| |
Collapse
|
39
|
Duan Y, Sun H, Yao Y, Han L, Chen L. Perturbation of serum metabolome in relation to type 2 diabetes mellitus and urinary levels of phthalate metabolites and bisphenols. ENVIRONMENT INTERNATIONAL 2021; 155:106609. [PMID: 33965767 DOI: 10.1016/j.envint.2021.106609] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/08/2021] [Accepted: 04/26/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Emerging evidence has proved the associations between exposure to phthalates (PAEs) and bisphenols and type 2 diabetes mellitus (T2DM), but the underlying mechanisms for these associations are poorly understood. Metabolomics is a powerful tool to identify differential metabolites and metabolic pathways related to diseases and chemical exposure, which may reveal underlying mechanisms. However, little is known about the roles of metabolism in the associations for PAE and bisphenol exposure with T2DM. OBJECTIVES The purpose of the study was to investigate the roles of metabolism in the associations for exposure to PAEs and bisphenols with T2DM. METHODS In this study, 60 T2DM cases and 60 controls, who were matched in age, sex, and body mass index (BMI), were selected from the total study population in our previous studies. Fasting blood and spot urine samples of the volunteers were used for non-targeted metabolomics analysis and determination of phthalate metabolites (mPAEs) and bisphenols, respectively. The associations of urinary mPAEs and bisphenols with screened metabolic biomarkers in metabolomics analysis were analyzed using multiple linear regression models. RESULTS Based on non-targeted metabolomics, 19 serum metabolic biomarkers were screened between T2DM cases and controls, mostly related to galactose metabolism, amino acid metabolism, and pyrimidine metabolism. More than half of mPAEs were mostly positively associated with up-regulated metabolic biomarkers and negatively associated with down-regulated biomarkers. Different from PAEs, no evident results suggested the roles of metabolism in the associations between bisphenol exposure and T2DM. CONCLUSIONS Combined with the positive associations between most urinary mPAEs and T2DM in our previous study, our findings indicated that PAE exposure may contribute to T2DM risk through disturbing galactose metabolism, amino acid metabolism (especially arginine biosynthesis and alanine, aspartate and glutamate metabolism), and pyrimidine metabolism.
Collapse
Affiliation(s)
- Yishuang Duan
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan, China; Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China.
| | - Hongwen Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China.
| | - Yiming Yao
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, China.
| | - Liping Han
- Tianjin Medical University Metabolic Diseases Hospital, Tianjin, China.
| | - Liming Chen
- Tianjin Medical University Metabolic Diseases Hospital, Tianjin, China.
| |
Collapse
|
40
|
The gut microbiota-related metabolite phenylacetylglutamine associates with increased risk of incident coronary artery disease. J Hypertens 2021; 38:2427-2434. [PMID: 32665522 DOI: 10.1097/hjh.0000000000002569] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The gut microbiota is increasingly being implicated in cardiovascular health. Metabolites produced by bacteria have been suggested to be mediators in the bacterial action on cardiovascular health. We aimed to identify gut microbiota-related plasma metabolites and test whether these metabolites associate with future risk of coronary artery disease (CAD). METHODS Nontargeted metabolomics was performed using liquid chromatography-mass spectrometry in order to measure 1446 metabolite features in the Malmö Offspring Study (MOS) (N = 776). The gut microbiota was characterized using 16S rRNA sequencing. Gut bacteria-related metabolites were measured in two independent prospective cohorts, the Malmö Diet and Cancer - Cardiovascular Cohort (MDC-CC) (N = 3361) and the Malmö Preventive Project (MPP) (N = 880), in order to investigate the associations between gut bacteria-related metabolites and risk of CAD. RESULTS In MOS, 33 metabolite features were significantly (P < 4.8e-7) correlated with at least one operational taxonomic unit. Phenylacetylglutamine (PAG) was associated with an increased risk of future CAD, using inverse variance weighted meta-analysis of age and sex-adjusted logistic regression models in MDC-CC and MPP. PAG remained significantly associated with CAD (OR = 1.17, 95% CI = 1.06-1.29, P = 1.9e-3) after adjustments for cardiovascular risk factors. CONCLUSION The levels of 33 plasma metabolites were correlated with the gut microbiota. Out of these, PAG was associated with an increased risk of future CAD independently of other cardiovascular risk factors. Our results highlight a link between the gut microbiota and CAD risk and should encourage further studies testing if modification of PAG levels inhibits development of CAD.
Collapse
|
41
|
Bardanzellu F, Puddu M, Peroni DG, Fanos V. The clinical impact of maternal weight on offspring health: lights and shadows in breast milk metabolome. Expert Rev Proteomics 2021; 18:571-606. [PMID: 34107825 DOI: 10.1080/14789450.2021.1940143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Pre-pregnancy overweight and obesity, depending on maternal nutrition and metabolic state, can influence fetal, neonatal and long-term offspring health, regarding cardio-metabolic, respiratory, immunological and cognitive outcomes. Thus, maternal weight can act, through mechanisms that are not full understood, on the physiology and metabolism of some fetal organs and tissues, to adapt themselves to the intrauterine environment and nutritional reserves. These effects could occur by modulating gene expression, neonatal microbiome, and through breastfeeding. AREAS COVERED In this paper, we investigated the potential effects of metabolites found altered in breast milk (BM) of overweight/obese mothers, through an extensive review of metabolomics studies, and the potential short- and long-term clinical effects in the offspring, especially regarding overweight, glucose homeostasis, insulin resistance, oxidative stress, infections, immune processes, and neurodevelopment. EXPERT OPINION Metabolomics seems the ideal tool to investigate BM variation depending on maternal or fetal/neonatal factors. In particular, BM metabolome alterations according to maternal conditions were recently pointed out in cases of gestational diabetes, preeclampsia, intrauterine growth restriction and maternal overweight/obesity. In our opinion, even if BM is the food of choice in neonatal nutrition, the deepest comprehension of its composition in overweight/obese mothers could allow targeted supplementation, to improve offspring health and metabolic homeostasis.
Collapse
Affiliation(s)
- Flaminia Bardanzellu
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari. SS 554 km 4,500, 09042 Monserrato. Italy
| | - Melania Puddu
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari. SS 554 km 4,500, 09042 Monserrato. Italy
| | - Diego Giampietro Peroni
- Clinical and Experimental Medicine Department, section of Pediatrics, University of Pisa, Italy. Via Roma, 55, 56126 Pisa PI, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, AOU and University of Cagliari. SS 554 km 4,500, 09042 Monserrato. Italy
| |
Collapse
|
42
|
Wang S, Cao YF, Sun XY, Hong M, Fang ZZ, Luo HH, Sun H, Yang P. Plasma Amino Acids and Residual Hypertriglyceridemia in Diabetic Patients Under Statins: Two Independent Cross-Sectional Hospital-Based Cohorts. Front Cardiovasc Med 2021; 8:605716. [PMID: 34136538 PMCID: PMC8200824 DOI: 10.3389/fcvm.2021.605716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Objective: The objective of the study was to investigate the relationship of amino acid metabolism with hypertriglyceridemia in diabetic patients under statins free of prior cardiovascular diseases. Methods: Two independent cross-sectional hospital based cohorts, i.e., Liaoning Medical University First Affiliated Hospital (LMUFAH, n = 146) and the Second Affiliated Hospital of Dalian Medical University (SAHDMU, n = 294) were included in the current analysis. Hypertriglyceridemia was defined as triglyceride ≥1.7 mmol/L, and well-controlled LDL-C was defined as <2.6 mmol/L. The adjusted ORs (95% CI) of circulating metabolic measures for hypertriglyceridemia were assessed using logistic regression. Pooled results of metabolites with the same direction of association in both cohorts were combined using inverse variance-weighted fixed-effect meta-analysis. Difference of identified metabolites in patients with and without hypertriglyceridemia were also obtained in the context of LDL-C. Results: Patients, 86 and 106, were with hypertriglyceridemia in LMUFAH and SAHDMU, respectively. We observed that elevated alanine, asparagine, leucine, and valine were consistently associated with increased hypertriglyceridemia in both cohorts. In fixed-effect pooled analysis, the OR (95% CI) per SD increase was 1.71 (1.32–2.20) for alanine, 1.62 (1.20–2.19) for asparagine, 1.64 (1.22–2.20) for leucine, and 1.62 (1.22–2.13) for valine (all P values ranged from 0.0018 to <0.0001); adjusting for C-peptide attenuated effect sizes of Ala, Leu, and Val for hypertriglyceridemia. The difference were robust in groups with well- or bad-controlled LDL-C. Conclusion: Among 23 amino acids, alanine, asparagine, leucine, and valine were positively associated with increased residual risk of hypertriglyceridemia in diabetic patients with statin treatment.
Collapse
Affiliation(s)
- Shuang Wang
- Cardiology Department, China-Japan Union Hospital of Jilin University, Changchun, China.,Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Changchun, China.,Jilin Provincial Cardiovascular Research Institute, Changchun, China
| | - Yun-Feng Cao
- Key Laboratory of Liaoning Tumor Clinical Metabolomics, Jinzhou, China
| | | | - Mo Hong
- RSKT Biopharma Inc, Dalian, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Hui-Huan Luo
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Huan Sun
- Cardiology Department, China-Japan Union Hospital of Jilin University, Changchun, China.,Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Changchun, China.,Jilin Provincial Cardiovascular Research Institute, Changchun, China
| | - Ping Yang
- Cardiology Department, China-Japan Union Hospital of Jilin University, Changchun, China.,Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Changchun, China.,Jilin Provincial Cardiovascular Research Institute, Changchun, China
| |
Collapse
|
43
|
Deidda M, Noto A, Cadeddu Dessalvi C, Andreini D, Andreotti F, Ferrannini E, Latini R, Maggioni AP, Magnoni M, Maseri A, Mercuro G. Metabolomic correlates of coronary atherosclerosis, cardiovascular risk, both or neither. Results of the 2 × 2 phenotypic CAPIRE study. Int J Cardiol 2021; 336:14-21. [PMID: 34022320 DOI: 10.1016/j.ijcard.2021.05.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Traditional cardiovascular risk factors (RFs) and coronary artery disease (CAD) do not always run parallel. We investigated functional-metabolic correlations of CAD, RFs, or neither in the CAPIRE (Coronary Atherosclerosis in Outlier Subjects: Protective and Novel Individual Risk Factors Evaluation) 2 × 2 phenotypic observational study. METHODS Two hundred and fortyone subjects were included based on RF burden, presence/absence of CAD (assessed by computed tomography angiography), age and sex. Participants displayed one of four phenotypes: CAD with ≥3 RFs, no-CAD with ≥3 RFs, CAD with ≤1 RF and no-CAD with ≤1 RF. Metabolites were identified by gas chromatography-mass spectrometry and pathways by metabolite set enrichment analysis. RESULTS Characteristic patterns and specific pathways emerged for each phenotypic group: amino sugars for CAD/high-RF; urea cycle for no-CAD/high-RF; glutathione for CAD/low-RF; glycine and serine for no-CAD/low-RF. Presence of CAD correlated with ammonia recycling; absence of CAD with the transfer of acetyl groups into mitochondria; high-risk profile with alanine metabolism (all p < 0.05). The comparative case-control analyses showed a statistically significant difference for the two pathways of phenylalanine, tyrosine and tryptophan biosynthesis and phenylalanine metabolism in the CAD/Low-RF vs NoCAD/Low-RF comparison. CONCLUSIONS The present 2 × 2 observational study identified specific metabolic pathways for each of the four phenotypes, providing novel functional insights, particularly on CAD with low RF profiles and on the absence of CAD despite high-risk factor profiles.
Collapse
Affiliation(s)
- Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Antonio Noto
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - Daniele Andreini
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, Cardiovascular Section, University of Milan, Italy
| | - Felicita Andreotti
- Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Roberto Latini
- Mario Negri Institute of Pharmacological Research, IRCCS, Milan, Italy
| | - Aldo P Maggioni
- ANMCO Research Center, Heart Care Foundation, Florence, Italy; Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Marco Magnoni
- IRCCS Ospedale San Raffaele and Università Vita-Salute San Raffaele, Milan, Italy
| | | | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.
| | | |
Collapse
|
44
|
Distinct Basal Metabolism in Three Mouse Models of Neurodevelopmental Disorders. eNeuro 2021; 8:ENEURO.0292-20.2021. [PMID: 33820803 PMCID: PMC8174051 DOI: 10.1523/eneuro.0292-20.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 02/27/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Prevalence of metabolic disturbances is higher among individuals with neurodevelopmental disorders (NDDs), yet this association has been largely overlooked. Investigation into human disease remains challenging, as a complete pathophysiological understanding relies on accurate modeling and highly controlled variables. Genetically engineered mouse models are widely used to gain insight into the biology of human NDDs, but research focus has been on behavioral and neurophysiological abnormalities. Such models not only allow for evaluating usefulness in reproducing human features, including similarities and discrepancies with rodent phenotypes, but they also represent a unique opportunity to observe and quantify novel anomalies. Here, we present the first characterization and comparison of basal metabolism in three mouse models of NDDs, namely, Down syndrome (DS; Dp(16)Yey/+ mice), 16p11.2 deletion syndrome (16pDel; 16p11.2df/+ mice), and fragile X syndrome [FXS; Fmr1 knock-out (KO) mice] and their wild-type (WT) counterparts. Using the Comprehensive Lab Animal Monitoring System (CLAMS) coupled to EchoMRI, as well as quantification of key plasma metabolites by liquid chromatography mass spectrometry (LC-MS), our in vivo study reveals that each mouse model expresses a unique metabolic signature that is sex-specific, independent of the amount of food consumed and minimally influenced by physical activity. In particular, we identify striking differences in body composition, respiratory exchange ratio (RER), caloric expenditure (CE), and concentrations of circulating plasma metabolites related to mitochondrial function. Providing novel insight into NDD-associated metabolic alterations is an essential prerequisite for future preclinical and clinical interventions.
Collapse
|
45
|
Gallo W, Ottosson F, Kennbäck C, Jujic A, Esguerra JLS, Eliasson L, Melander O. Replication study reveals miR-483-5p as an important target in prevention of cardiometabolic disease. BMC Cardiovasc Disord 2021; 21:162. [PMID: 33794782 PMCID: PMC8017779 DOI: 10.1186/s12872-021-01964-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/24/2021] [Indexed: 12/15/2022] Open
Abstract
Background Alterations in levels of circulating micro-RNAs might reflect within organ signaling or subclinical tissue injury that is linked to risk of diabetes and cardiovascular risk. We previously found that serum levels of miR-483-5p is correlated with cardiometabolic risk factors and incidence of cardiometabolic disease in a case–control sample from the populations-based Malmö Diet and Cancer Study Cardiovascular Cohort (MDC-CC). We here aimed at replicating these findings and to test for association with carotid atherosclerosis. Methods We measured miR-483-5p in fasting serum of 1223 healthy subjects from the baseline examination of the population-based, prospective cohort study Malmö Offspring Study (MOS) and correlated miR-483-5p to cardiometabolic risk factors and to incidence of diabetes mellitus and coronary artery disease (CAD) during 3.7 (± 1.3) years of follow-up using logistic regression. In both MOS and MDC-CC we related mir-483-5p to carotid atherosclerosis measured with ultrasound. Results In cross-sectional analysis miR-483-5p was correlated with BMI, waist circumference, HDL, and sex. After adjustment for age and sex, the association remained significant for all risk factors except for HDL. Logistic regression analysis showed significant associations between miR-483-5p and new-onset diabetes (OR = 1.94, 95% CI 1.06–3.56, p = 0.032) and cardiovascular disease (OR = 1.99, 95% CI 1.06–3.75, p = 0.033) during 3.7 (± 1.3) years of follow-up. Furthermore, miR-483-5p was significantly related with maximum intima-media thickness of the carotid bulb in MDC-CC (p = 0.001), but not in MOS, whereas it was associated with increasing number of plaques in MOS (p = 0.007). Conclusion miR-483-5p is related to an unfavorable cardiometabolic risk factor profile and predicts diabetes and CAD, possibly through an effect on atherosclerosis. Our results encourage further studies of possible underlying mechanisms and means of modifying miR-483-5p as a possible interventional target in prevention of cardiometabolic disease.
Collapse
Affiliation(s)
- Widet Gallo
- Department of Clinical Sciences-Malmö, Hypertension and Cardiovascular Disease, Lund University, 205 02, Malmö, Sweden. .,Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University Malmö, Malmö, Sweden. .,Clinical Research Centre, Skane University Hospital, Lund and Malmö, Malmö, Sweden. .,Department of Clinical Sciences-Malmö, Clinical Research Centre, CRC, Lund University, 91:12, Jan Waldenströmsgata 35, 214 28, Malmö, Sweden.
| | - Filip Ottosson
- Department of Clinical Sciences-Malmö, Hypertension and Cardiovascular Disease, Lund University, 205 02, Malmö, Sweden.,Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University Malmö, Malmö, Sweden.,Clinical Research Centre, Skane University Hospital, Lund and Malmö, Malmö, Sweden
| | - Cecilia Kennbäck
- Department of Emergency and Internal Medicine, Skane University Hospital, Malmö, Sweden
| | - Amra Jujic
- Department of Clinical Sciences-Malmö, Hypertension and Cardiovascular Disease, Lund University, 205 02, Malmö, Sweden.,Department of Cardiology, Skane University Hospital, Malmö, Sweden
| | - Jonathan Lou S Esguerra
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University Malmö, Malmö, Sweden.,Clinical Research Centre, Skane University Hospital, Lund and Malmö, Malmö, Sweden.,Department of Clinical Sciences-Malmö, Islet Cell Exocytosis, Lund University, Malmö, Sweden
| | - Lena Eliasson
- Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University Malmö, Malmö, Sweden.,Clinical Research Centre, Skane University Hospital, Lund and Malmö, Malmö, Sweden.,Department of Clinical Sciences-Malmö, Islet Cell Exocytosis, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences-Malmö, Hypertension and Cardiovascular Disease, Lund University, 205 02, Malmö, Sweden.,Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University Malmö, Malmö, Sweden.,Clinical Research Centre, Skane University Hospital, Lund and Malmö, Malmö, Sweden.,Department of Emergency and Internal Medicine, Skane University Hospital, Malmö, Sweden
| |
Collapse
|
46
|
Xuan C, Li H, Tian QW, Guo JJ, He GW, Lun LM, Wang Q. Quantitative Assessment of Serum Amino Acids and Association with Early-Onset Coronary Artery Disease. Clin Interv Aging 2021; 16:465-474. [PMID: 33758500 PMCID: PMC7979345 DOI: 10.2147/cia.s298743] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Background Amino acids play essential roles in protein construction and metabolism. Our study aims to provide a profile of amino acid changes in the serum of patients with early-onset coronary artery disease (EOCAD) and identify potential disease biomarkers. Methods Ultra-performance liquid chromatography-multiple reaction monitoring-multistage/mass spectrometry (UPLC-MRM-MS/MS) was used to determine the amino acid profile of patients with EOCAD in sample pools. In the validation stage, the serum levels of candidate amino acids of interest are determined for each sample. Results A total of 128 EOCAD patients and 64 healthy controls were included in the study. Eight serum amino acids associated with disease state were identified. Compared with the control group, serum levels of seven amino acids (L-Arginine, L-Methionine, L-Tyrosine, L-Serine, L-Aspartic acid, L-Phenylalanine, and L-Glutamic acid) increased and one (4-Hydroxyproline) decreased in the patient group. Results from the validation stage demonstrate that serum levels of 4-Hydroxyproline were significantly lower in myocardial infarction (MI) patients (9.889 ± 3.635 μg/mL) than those in the controls (16.433 ± 4.562 μmol/L, p < 0.001). Elevated serum 4-Hydroxyproline levels were shown to be an independent protective factor for MI (OR = 0.863, 95% CI: 0.822–0.901). The significant negative correlation was seen between serum 4-Hydroxyproline levels and cardiac troponin I (r = −0.667) in MI patients. Conclusion We have provided a serum amino acid profile for EOCAD patients and screened eight disease state-related amino acids, and we have also shown that 4-Hydroxyproline is a promising target for further biomarker studies in early-onset MI.
Collapse
Affiliation(s)
- Chao Xuan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Hui Li
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Qing-Wu Tian
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Jun-Jie Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Guo-Wei He
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, People's Republic of China.,Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Li-Min Lun
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Qing Wang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| |
Collapse
|
47
|
Comparing Levels of Metabolic Predictors of Coronary Heart Disease between Healthy Lean and Overweight Females. Metabolites 2021; 11:metabo11030169. [PMID: 33804097 PMCID: PMC7999722 DOI: 10.3390/metabo11030169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/07/2021] [Accepted: 03/13/2021] [Indexed: 12/17/2022] Open
Abstract
Screening for the metabolomic signature of coronary heart disease (CHD) before disease onset could help in early diagnosis and potentially disease prevention. In this study, the levels of 17 CHD metabolic biomarkers in apparently healthy overweight females were compared to lean counterparts, and their associations with conventional clinical risk factors were determined. Clinical and metabolic data from 200 apparently healthy non-obese Qatari females were collected from Qatar Biobank (discovery cohort). Logistic regression was used to assess the association between body mass index (BMI) groups and 17 CHD metabolic biomarkers, and receiver operating characteristic (ROC) analysis was used to evaluate the prognostic value of CHD metabolic biomarkers in overweight. Stepwise linear regression was performed to identify the classical risk factors associated with CHD metabolites differentiating the two BMI groups. Validation of the association of CHD metabolic biomarkers with BMI groups was performed in 107 subjects (replication cohort). Out of the tested CHD metabolic biomarkers, five were significantly different between lean and overweight females in the discovery cohort (AUC = 0.73). Among these, the association of mannose, asparagine, and linoleate with BMI groups was confirmed in the replication cohort (AUC = 0.97). Significant correlations between predictors of CHD in overweight healthy women and classical risk factors were observed, including serum levels of cholesterol, testosterone, triiodothyronine, thyroxine, creatinine, albumin, bilirubin, glucose, c-peptide, uric acid, calcium and chloride. Apparently, healthy overweight females exhibit significantly different levels of specific CHD metabolites compared to their lean counterparts, offering a prognostic potential with preventative value.
Collapse
|
48
|
Li X, Li Y, Liang Y, Hu R, Xu W, Liu Y. Plasma Targeted Metabolomics Analysis for Amino Acids and Acylcarnitines in Patients with Prediabetes, Type 2 Diabetes Mellitus, and Diabetic Vascular Complications. Diabetes Metab J 2021; 45:195-208. [PMID: 33685035 PMCID: PMC8024149 DOI: 10.4093/dmj.2019.0209] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/26/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND We hypothesized that specific amino acids or acylcarnitines would have benefits for the differential diagnosis of diabetes. Thus, a targeted metabolomics for amino acids and acylcarnitines in patients with diabetes and its complications was carried out. METHODS A cohort of 54 normal individuals and 156 patients with type 2 diabetes mellitus and/or diabetic complications enrolled from the First Affiliated Hospital of Jinzhou Medical University was studied. The subjects were divided into five main groups: normal individuals, impaired fasting glucose, overt diabetes, diabetic microvascular complications, and diabetic peripheral vascular disease. The technique of tandem mass spectrometry was applied to obtain the plasma metabolite profiles. Metabolomics multivariate statistics were applied for the metabolic data analysis and the differential metabolites determination. RESULTS A total of 10 cross-comparisons within diabetes and its complications were designed to explore the differential metabolites. The results demonstrated that eight comparisons existed and yielded significant metabolic differences. A total number of 24 differential metabolites were determined from six selected comparisons, including up-regulated amino acids, down-regulated medium-chain and long-chain acylcarnitines. Altered differential metabolites provided six panels of biomarkers, which were helpful in distinguishing diabetic patients. CONCLUSION Our results demonstrated that the biomarker panels consisted of specific amino acids and acylcarnitines which could reflect the metabolic variations among the different stages of diabetes and might be useful for the differential diagnosis of prediabetes, overt diabetes and diabetic complications.
Collapse
Affiliation(s)
- Xin Li
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Yancheng Li
- Department of Biostatistics, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuanhao Liang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Ruixue Hu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Wenli Xu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Yufeng Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
- Natural Products Pharmaceutical Engineering Technology Research Center of Liaoning Province, Shenyang, China
- Corresponding author: Yufeng Liu https://orcid.org/0000-0001-7972-8771 School of Pharmaceutical Sciences, Liaoning University, Zheli Rd, Huanggu District, Shenyang 110036, China E-mail:
| |
Collapse
|
49
|
Sun Y, Zou H, Li X, Xu S, Liu C. Plasma Metabolomics Reveals Metabolic Profiling For Diabetic Retinopathy and Disease Progression. Front Endocrinol (Lausanne) 2021; 12:757088. [PMID: 34777253 PMCID: PMC8589034 DOI: 10.3389/fendo.2021.757088] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/29/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUNDS Diabetic retinopathy (DR), the main retinal vascular complication of DM, is the leading cause of visual impairment and blindness among working-age people worldwide. The aim of this study was to investigate the difference of plasma metabolic profiles in patients with DR to better understand the mechanism of this disease and disease progression. METHODS We used ultrahigh-performance liquid Q-Exactive mass spectrometry and multivariate statistical analyses to conduct a comprehensive analysis of plasma metabolites in a population with DR and proliferative DR (PDR). A risk score based on the level of the selected metabolite was established and evaluated using the least absolute shrinkage and selection operator regularization logistic regression (LASSO-LR) based machine learning model. RESULTS 22 differentially expressed metabolites which belonged to different metabolic pathway were identified and confirmed to be associated with the occurrence of DR. A risk score based on the level of the selected metabolite pseudouridine was established and evaluated to strongly associated with the occurrence of DR. Four circulating plasma metabolites (pseudouridine, glutamate, leucylleucine and N-acetyltryptophan) were identified to be differentially expressed between patients with PDR and other patients, and a risk score formula based on these plasma metabolites was developed and assessed to be significantly related to PDR. CONCLUSIONS Our work highlights the possible use of the risk score assessment based on the plasma metabolites not only reveal in the early diagnosis of DR and PDR but also assist in enhancing current therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Yu Sun
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
- Department of Endocrinology and Metabolism, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Huiling Zou
- Department of Endocrinology and Metabolism, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Xingjia Li
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
- Treatment of Yingbing of State Administration of Traditional Chinese Medicine, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Shuhang Xu
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
- Treatment of Yingbing of State Administration of Traditional Chinese Medicine, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
- *Correspondence: Chao Liu, ; Shuhang Xu,
| | - Chao Liu
- Department of Endocrinology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
- Treatment of Yingbing of State Administration of Traditional Chinese Medicine, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
- *Correspondence: Chao Liu, ; Shuhang Xu,
| |
Collapse
|
50
|
Gu X, Al Dubayee M, Alshahrani A, Masood A, Benabdelkamel H, Zahra M, Li L, Abdel Rahman AM, Aljada A. Distinctive Metabolomics Patterns Associated With Insulin Resistance and Type 2 Diabetes Mellitus. Front Mol Biosci 2020; 7:609806. [PMID: 33381523 PMCID: PMC7768025 DOI: 10.3389/fmolb.2020.609806] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/23/2020] [Indexed: 01/17/2023] Open
Abstract
Obesity is associated with an increased risk of insulin resistance (IR) and type 2 diabetes mellitus (T2DM) which is a multi-factorial disease associated with a dysregulated metabolism and can be prevented in pre-diabetic individuals with impaired glucose tolerance. A metabolomic approach emphasizing metabolic pathways is critical to our understanding of this heterogeneous disease. This study aimed to characterize the serum metabolomic fingerprint and multi-metabolite signatures associated with IR and T2DM. Here, we have used untargeted high-performance chemical isotope labeling (CIL) liquid chromatography-mass spectrometry (LC-MS) to identify candidate biomarkers of IR and T2DM in sera from 30 adults of normal weight, 26 obese adults, and 16 adults newly diagnosed with T2DM. Among the 3633 peak pairs detected, 62% were either identified or matched. A group of 78 metabolites were up-regulated and 111 metabolites were down-regulated comparing obese to lean group while 459 metabolites were up-regulated and 166 metabolites were down-regulated comparing T2DM to obese groups. Several metabolites were identified as IR potential biomarkers, including amino acids (Asn, Gln, and His), methionine (Met) sulfoxide, 2-methyl-3-hydroxy-5-formylpyridine-4-carboxylate, serotonin, L-2-amino-3-oxobutanoic acid, and 4,6-dihydroxyquinoline. T2DM was associated with dysregulation of 42 metabolites, including amino acids, amino acids metabolites, and dipeptides. In conclusion, these pilot data have identified IR and T2DM metabolomics panels as potential novel biomarkers of IR and identified metabolites associated with T2DM, with possible diagnostic and therapeutic applications. Further studies to confirm these associations in prospective cohorts are warranted.
Collapse
Affiliation(s)
- Xinyun Gu
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Mohammed Al Dubayee
- Department of Medicine, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Awad Alshahrani
- Department of Medicine, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Afshan Masood
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hicham Benabdelkamel
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mahmoud Zahra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Anas M Abdel Rahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.,Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|