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
|
Li J, Wu B, Fan G, Huang J, Li Z, Cao F. Lc-ms-based untargeted metabolomics reveals potential mechanisms of histologic chronic inflammation promoting prostate hyperplasia. PLoS One 2024; 19:e0314599. [PMID: 39715183 DOI: 10.1371/journal.pone.0314599] [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: 05/14/2024] [Accepted: 11/14/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Chronic prostatitis may be a risk factor for developing proliferative changes in the prostate, although the underlying mechanisms are not entirely comprehended. MATERIALS AND METHODS Fifty individual prostate tissues were examined in this study, consisting of 25 patients diagnosed with prostatic hyperplasia combined with histologic chronic inflammation and 25 patients diagnosed with prostatic hyperplasia alone. We employed UPLC-Q-TOF-MS-based untargeted metabolomics using ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry to identify differential metabolites that can reveal the mechanisms that underlie the promotion of prostate hyperplasia by histologic chronic inflammation. Selected differential endogenous metabolites were analyzed using bioinformatics and subjected to metabolic pathway studies. RESULTS Nineteen differential metabolites, consisting of nine up-regulated and ten down-regulated, were identified between the two groups of patients. These groups included individuals with combined histologic chronic inflammation and those with prostatic hyperplasia alone. Glycerolipids, glycerophospholipids, and sphingolipids were primarily the components present. Metabolic pathway enrichment was conducted on the identified differentially expressed metabolites. Topological pathway analysis revealed the differential metabolites' predominant involvement in sphingolipid, ether lipid, and glycerophospholipid metabolism. The metabolites involved in sphingolipid metabolism were Sphingosine, Cer (d18:1/24:1), and Phytosphingosine. The metabolites involved in ether lipid metabolism were Glycerophosphocholine and LysoPC (O-18:0/0:0). The metabolites involved in glycerophospholipid metabolism were LysoPC (P-18:0/0:0) and Glycerophosphocholine. with Impact > 0. 1 and FDR < 0. 05, the most important metabolic pathway was sphingolipid metabolism. CONCLUSIONS In conclusion, our findings suggest that patients with prostate hyperplasia and combined histologic chronic inflammation possess distinctive metabolic profiles. These differential metabolites appear to play a significant role in the pathogenesis of histologic chronic inflammation-induced prostate hyperplasia, primarily through the regulation of sphingolipids and glycerophospholipids metabolic pathways. The mechanism by which histologic chronic inflammation promotes prostate hyperplasia was elucidated through the analysis of small molecule metabolites. These findings support the notion that chronic prostatitis may contribute to an increased risk of prostate hyperplasia.
Collapse
Affiliation(s)
- Jiale Li
- Clinical Medical College, North China University of Science and Technology, Tangshan, China
| | - Beiwen Wu
- Clinical Medical College, North China University of Science and Technology, Tangshan, China
| | - Guorui Fan
- Clinical Medical College, North China University of Science and Technology, Tangshan, China
| | - Jie Huang
- Clinical Medical College, North China University of Science and Technology, Tangshan, China
| | - Zhiguo Li
- The Hebei Key Lab for Organ Fibrosis, The Hebei Key Lab for Chronic Disease, School of Public Health, International Science and Technology Cooperation Base of Geriatric Medicine, North China University of Science and Technology, Tangshan, China
| | - Fenghong Cao
- Clinical Medical College, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
3
|
Sharma S, Dong Q, Haid M, Adam J, Bizzotto R, Fernandez-Tajes JJ, Jones AG, Tura A, Artati A, Prehn C, Kastenmüller G, Koivula RW, Franks PW, Walker M, Forgie IM, Giordano G, Pavo I, Ruetten H, Dermitzakis M, McCarthy MI, Pedersen O, Schwenk JM, Tsirigos KD, De Masi F, Brunak S, Viñuela A, Mari A, McDonald TJ, Kokkola T, Adamski J, Pearson ER, Grallert H. Role of human plasma metabolites in prediabetes and type 2 diabetes from the IMI-DIRECT study. Diabetologia 2024; 67:2804-2818. [PMID: 39349772 PMCID: PMC11604760 DOI: 10.1007/s00125-024-06282-6] [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] [Received: 02/28/2024] [Accepted: 07/29/2024] [Indexed: 11/29/2024]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is a chronic condition that is caused by hyperglycaemia. Our aim was to characterise the metabolomics to find their association with the glycaemic spectrum and find a causal relationship between metabolites and type 2 diabetes. METHODS As part of the Innovative Medicines Initiative - Diabetes Research on Patient Stratification (IMI-DIRECT) consortium, 3000 plasma samples were measured with the Biocrates AbsoluteIDQ p150 Kit and Metabolon analytics. A total of 911 metabolites (132 targeted metabolomics, 779 untargeted metabolomics) passed the quality control. Multivariable linear and logistic regression analysis estimates were calculated from the concentration/peak areas of each metabolite as an explanatory variable and the glycaemic status as a dependent variable. This analysis was adjusted for age, sex, BMI, study centre in the basic model, and additionally for alcohol, smoking, BP, fasting HDL-cholesterol and fasting triacylglycerol in the full model. Statistical significance was Bonferroni corrected throughout. Beyond associations, we investigated the mediation effect and causal effects for which causal mediation test and two-sample Mendelian randomisation (2SMR) methods were used, respectively. RESULTS In the targeted metabolomics, we observed four (15), 34 (99) and 50 (108) metabolites (number of metabolites observed in untargeted metabolomics appear in parentheses) that were significantly different when comparing normal glucose regulation vs impaired glucose regulation/prediabetes, normal glucose regulation vs type 2 diabetes, and impaired glucose regulation vs type 2 diabetes, respectively. Significant metabolites were mainly branched-chain amino acids (BCAAs), with some derivatised BCAAs, lipids, xenobiotics and a few unknowns. Metabolites such as lysophosphatidylcholine a C17:0, sum of hexoses, amino acids from BCAA metabolism (including leucine, isoleucine, valine, N-lactoylvaline, N-lactoylleucine and formiminoglutamate) and lactate, as well as an unknown metabolite (X-24295), were associated with HbA1c progression rate and were significant mediators of type 2 diabetes from baseline to 18 and 48 months of follow-up. 2SMR was used to estimate the causal effect of an exposure on an outcome using summary statistics from UK Biobank genome-wide association studies. We found that type 2 diabetes had a causal effect on the levels of three metabolites (hexose, glutamate and caproate [fatty acid (FA) 6:0]), whereas lipids such as specific phosphatidylcholines (PCs) (namely PC aa C36:2, PC aa C36:5, PC ae C36:3 and PC ae C34:3) as well as the two n-3 fatty acids stearidonate (18:4n3) and docosapentaenoate (22:5n3) potentially had a causal role in the development of type 2 diabetes. CONCLUSIONS/INTERPRETATION Our findings identify known BCAAs and lipids, along with novel N-lactoyl-amino acid metabolites, significantly associated with prediabetes and diabetes, that mediate the effect of diabetes from baseline to follow-up (18 and 48 months). Causal inference using genetic variants shows the role of lipid metabolism and n-3 fatty acids as being causal for metabolite-to-type 2 diabetes whereas the sum of hexoses is causal for type 2 diabetes-to-metabolite. Identified metabolite markers are useful for stratifying individuals based on their risk progression and should enable targeted interventions.
Collapse
Affiliation(s)
- Sapna Sharma
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Qiuling Dong
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians-University München, Munich, Germany
| | - Mark Haid
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jonathan Adam
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| | - Roberto Bizzotto
- Institute of Neuroscience, National Research Council, Padova, Italy
| | | | - Angus G Jones
- Department of Clinical and Biomedical Sciences, University of Exeter College of Medicine & Health, Exeter, UK
| | - Andrea Tura
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Anna Artati
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Robert W Koivula
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Paul W Franks
- Department of Clinical Science, Genetic and Molecular Epidemiology, Lund University Diabetes Centre, Malmö, Sweden
| | - Mark Walker
- Translational and Clinical Research Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, UK
| | - Ian M Forgie
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Giuseppe Giordano
- Department of Clinical Science, Genetic and Molecular Epidemiology, Lund University Diabetes Centre, Malmö, Sweden
| | - Imre Pavo
- Eli Lilly Regional Operations GmbH, Vienna, Austria
| | - Hartmut Ruetten
- Sanofi Partnering, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - Manolis Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Oluf Pedersen
- Center for Clinical Metabolic Research, Herlev and Gentofte University Hospital, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jochen M Schwenk
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | | | - Federico De Masi
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Soren Brunak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ana Viñuela
- Biosciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, UK
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padova, Italy
| | | | - Tarja Kokkola
- Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jerzy Adamski
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Experimental Genetics, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ewan R Pearson
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), München Neuherberg, Germany.
| |
Collapse
|
4
|
Zhang X, Liu H, Li C, Wei Y, Kan X, Liu X, Han X, Zhao Z, An T, Fang ZZ, Ma S, Zheng R, Li J. Abdominal obesity in youth: the associations of plasma Lysophophatidylcholine concentrations with insulin resistance. Pediatr Res 2024:10.1038/s41390-024-03652-z. [PMID: 39427100 DOI: 10.1038/s41390-024-03652-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUD This study aimed to explore the associations of lysophosphatidylcholines (LPCs) with insulin resistance (IR) and abdominal obesity among children and adolescents. METHODS A cross-sectional study was conducted on 612 young individuals, aged 7 to 18 years in Tianjin City, China. LC-MS metabolomic analysis was used to measure LPCs levels. The Homeostasis Model Assessment was used to estimate IR. Waist circumference measurements were used to assess abdominal obesity. Logistic regression models were employed to explore the relationships between LPCs and IR and abdominal obesity. Mediation analyses were performed to analyze whether LPCs affected IR through abdominal obesity. RESULTS Compared to their counterparts, five specific LPCs were significantly different in youth with IR. The levels of LPC 24:0 and 26:0 were significantly associated with IR after adjustment. Both decreased levels of LPC 24:0 and 26:0 associated with the increased risks of IR (OR: 0.64, 95%CI: 0.38-0.95; OR: 0.66, 95%CI: 0.40-1.00), and the ORs for abdominal obesity were 0.68 (95%CI: 0.38-1.00) and 0.51 (95%CI: 0.28-0.90), respectively. Mediation analysis indicated that abdominal obesity mediated the association between LPC 26:0 and IR, with a total effect (c) of -0.109 (P < 0.05), a direct effect (c') of -0.055 (P > 0.05), and an indirect effect through obesity (a × b) path with "a" of -0.125 (P < 0.05) and "b" of 0.426 (P < 0.05). CONCLUSION Overall findings suggest that decreased levels of LPC 24:0 and 26:0 were associated with increased risks of IR and abdominal obesity. Importantly, addressing abdominal obesity may mediate the impact of IR driven by LPC 26:0.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Huiying Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Chenyu Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ying Wei
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuan Kan
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoxiao Liu
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyi Han
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenghao Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Tianfeng An
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Shifeng Ma
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin, China.
| | - Rongxiu Zheng
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin, China.
| | - Jing Li
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China.
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
5
|
Chen L, Goh XP, Bendt AK, Tan KML, Leow MKS, Tan KH, Chan JKY, Chan SY, Chong YS, Gluckman PD, Eriksson JG, Wenk MR, Mir SA. Association of Acylcarnitines With Maternal Cardiometabolic Risk Factors Is Defined by Chain Length: The S-PRESTO Study. J Clin Endocrinol Metab 2024; 109:2831-2846. [PMID: 38625914 DOI: 10.1210/clinem/dgae255] [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] [Received: 11/15/2023] [Revised: 03/02/2024] [Accepted: 04/10/2024] [Indexed: 04/18/2024]
Abstract
CONTEXT Due to the essential role of carnitine as an intermediary in amino acid, carbohydrate, and lipid metabolism, a detailed characterization of circulating and urinary carnitine concentrations will aid in elucidating the molecular basis of impaired maternal metabolic flexibility and facilitating timely intervention for expectant mothers. OBJECTIVE To investigate the association of maternal plasma and urinary free carnitine and acylcarnitines with cardiometabolic risk factors. METHODS Liquid chromatography tandem mass spectrometry-based quantification of free carnitine and acylcarnitines (C2-C18) was performed on 765 plasma and 702 urine samples collected at preconception, 26 to 28 weeks' pregnancy, and 3 months postpartum in the Singapore PREconception Study of long-Term maternal and child Outcomes (S-PRESTO) cohort study. RESULTS Plasma concentrations of free carnitine and acylcarnitines decreased coupled with increased renal clearance in pregnancy compared with preconception and postpartum. Renal clearance of carnitine increased with an increase in prepregnancy body mass index (ppBMI) and gestational weight gain. Plasma short-chain acylcarnitines were positively associated with ppBMI, irrespective of the physiological state, while medium- and long-chain acylcarnitines were negatively associated with ppBMI at preconception and postpartum but showed a positive association in pregnancy. Similarly, plasma short-chain acylcarnitines were positively associated with Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) whereas medium- and long-chain acylcarnitines were negatively associated with HOMA-IR at preconception and in pregnancy. Mothers who developed gestational diabetes mellitus during pregnancy had ∼10% higher plasma propionylcarnitine concentration and ∼18% higher urine tiglylcarnitine concentration than mothers with normal glucose metabolism at preconception. CONCLUSION This study provides the metabolic and physiological basis of maternal carnitine homeostasis, which can be used in assessment of maternal cardiometabolic health at preconception to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Li Chen
- Singapore Institute for Clinical Sciences, A*STAR, 117609 Singapore, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Xue Ping Goh
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Anne K Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Karen Mei-Ling Tan
- Singapore Institute for Clinical Sciences, A*STAR, 117609 Singapore, Singapore
- Department of Laboratory Medicine, National University Hospital, 119074 Singapore, Singapore
| | - Melvin Khee-Shing Leow
- Singapore Institute for Clinical Sciences, A*STAR, 117609 Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, 308433 Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921 Singapore, Singapore
- Duke-National University of Singapore (NUS) Medical School, 169857 Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research programme, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore
| | - Kok Hian Tan
- Duke-National University of Singapore (NUS) Medical School, 169857 Singapore, Singapore
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, 229899 Singapore, Singapore
| | - Jerry Kok Yen Chan
- Duke-National University of Singapore (NUS) Medical School, 169857 Singapore, Singapore
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, 229899 Singapore, Singapore
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, A*STAR, 117609 Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research programme, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, A*STAR, 117609 Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research programme, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, A*STAR, 117609 Singapore, Singapore
- Liggins Institute, University of Auckland, Auckland 1142, New Zealand
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences, A*STAR, 117609 Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research programme, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore
- Folkhalsan Research Center, 00250 Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, 00290 Helsinki, Finland
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
- Department of Biochemistry and Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117596 Singapore, Singapore
| | - Sartaj Ahmad Mir
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
- Department of Biochemistry and Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117596 Singapore, Singapore
| |
Collapse
|
6
|
Li Y, Han S. Metabolomic Applications in Gut Microbiota-Host Interactions in Human Diseases. Gastroenterol Clin North Am 2024; 53:383-397. [PMID: 39068001 DOI: 10.1016/j.gtc.2023.12.008] [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] [Indexed: 07/30/2024]
Abstract
The human gut microbiota, consisting of trillions of microorganisms, encodes diverse metabolic pathways that impact numerous aspects of host physiology. One key way in which gut bacteria interact with the host is through the production of small metabolites. Several of these microbiota-dependent metabolites, such as short-chain fatty acids, have been shown to modulate host diseases. In this review, we examine how disease-associated metabolic signatures are identified using metabolomic platforms, and where metabolomics is applied in gut microbiota-disease interactions. We further explore how integration of metagenomic and metabolomic data in human studies can facilitate biomarkers discoveries in precision medicine.
Collapse
Affiliation(s)
- Yuxin Li
- Biochemistry Graduate Program, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shuo Han
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA; Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, NC 27710, USA.
| |
Collapse
|
7
|
Bagheri M, Tanriverdi K, Iafrati MD, Mosley JD, Freedman JE, Ferguson JF. Characterization of the plasma metabolome and lipidome in response to sleeve gastrectomy and gastric bypass surgeries reveals molecular patterns of surgical weight loss. Metabolism 2024; 158:155955. [PMID: 38906372 DOI: 10.1016/j.metabol.2024.155955] [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] [Received: 01/30/2024] [Revised: 05/23/2024] [Accepted: 06/13/2024] [Indexed: 06/23/2024]
Abstract
OBJECTIVES Bariatric surgery improves metabolic health, but the underlying mechanisms are not fully understood. We analyzed the effects of two types of bariatric surgery, sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB), on the plasma metabolome and lipidome. METHODS We characterized the plasma metabolome (1268 metabolites) and lipidome (953 lipids) pre-operatively and at 3 and 12 months post-operatively in 104 obese adults who were previously recruited to a prospective cohort of bariatric surgery. The metabolomic and lipidomic responses to bariatric surgery over time were analyzed using multivariable linear mixed-effects models. RESULTS There were significant changes in multiple metabolites and lipids, including rapid early changes in amino acid and peptide metabolites, including decreases in branched-chain amino acids (BCAAs), aromatic AAs, alanine and aspartate, and increases in glycine, serine, arginine and citrulline. There were also significant decreases in many triglyceride species, with increases in phosphatidylcholines and phosphatidylethanolamines. There were significant changes in metabolites related to energy metabolism that were apparent only after 12 months. We observed differences by bariatric surgery type in the changes in a small number of primary and secondary bile acids, including glycohyocholate and glyco-beta-muricholate. CONCLUSIONS Our findings highlight the comprehensive changes in metabolites and lipids that occur over the 12 months following bariatric surgery. While both SG and RYGB caused profound changes in the metabolome and lipidome, RYGB was characterized by greater increases in bile acids following surgery.
Collapse
Affiliation(s)
- Minoo Bagheri
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Kahraman Tanriverdi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Mark D Iafrati
- Department of Vascular Surgery, Vanderbilt University Medical Center, United States of America
| | - Jonathan D Mosley
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States of America; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Jane E Freedman
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America.
| |
Collapse
|
8
|
Cuahtecontzi Delint R, Ishak MI, Tsimbouri PM, Jayawarna V, Burgess KVE, Ramage G, Nobbs AH, Damiati L, Salmeron-Sanchez M, Su B, Dalby MJ. Nanotopography Influences Host-Pathogen Quorum Sensing and Facilitates Selection of Bioactive Metabolites in Mesenchymal Stromal Cells and Pseudomonas aeruginosa Co-Cultures. ACS APPLIED MATERIALS & INTERFACES 2024; 16:43374-43386. [PMID: 39113638 PMCID: PMC11345723 DOI: 10.1021/acsami.4c09291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
Orthopedic implant-related bacterial infections and resultant antibiotic-resistant biofilms hinder implant-tissue integration and failure. Biofilm quorum sensing (QS) communication determines the pathogen colonization success. However, it remains unclear how implant modifications and host cells are influenced by, or influence, QS. High aspect ratio nanotopographies have shown to reduce biofilm formation of Pseudomonas aeruginosa, a sepsis causing pathogen with well-defined QS molecules. Producing such nanotopographies in relevant orthopedic materials (i.e., titanium) allows for probing QS using mass spectrometry-based metabolomics. However, nanotopographies can reduce host cell adhesion and regeneration. Therefore, we developed a polymer (poly(ethyl acrylate), PEA) coating that organizes extracellular matrix proteins, promoting bioactivity to host cells such as human mesenchymal stromal cells (hMSCs), maintaining biofilm reduction. This allowed us to investigate how hMSCs, after winning the race for the surface against pathogenic cells, interact with the biofilm. Our approach revealed that nanotopographies reduced major virulence pathways, such as LasR. The enhanced hMSCs support provided by the coated nanotopographies was shown to suppress virulence pathways and biofilm formation. Finally, we selected bioactive metabolites and demonstrated that these could be used as adjuncts to the nanostructured surfaces to reduce biofilm formation and enhance hMSC activity. These surfaces make excellent models to study hMSC-pathogen interactions and could be envisaged for use in novel orthopedic implants.
Collapse
Affiliation(s)
- Rosalia Cuahtecontzi Delint
- Centre
for the Cellular Microenvironment, School of Molecular Biosciences,
College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced
Research Centre, University of Glasgow, Glasgow G11 6EW, United Kingdom
| | - Mohd I. Ishak
- Bristol
Dental School Research Laboratories, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, United Kingdom
| | - Penelope M. Tsimbouri
- Centre
for the Cellular Microenvironment, School of Molecular Biosciences,
College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced
Research Centre, University of Glasgow, Glasgow G11 6EW, United Kingdom
| | - Vineetha Jayawarna
- Centre
for the Cellular Microenvironment, School of Molecular Biosciences,
College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced
Research Centre, University of Glasgow, Glasgow G11 6EW, United Kingdom
| | - Karl V. E. Burgess
- EdinOmics, University
of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, United Kingdom
| | - Gordon Ramage
- Safeguarding
Health through Infection Prevention (SHIP) Research Group, Research
Centre for Health, Glasgow Caledonian University, Glasgow G4 0BA, United Kingdom
| | - Angela H. Nobbs
- Bristol
Dental School Research Laboratories, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, United Kingdom
| | - Laila Damiati
- Department
of Biological Sciences, College of Science, University of Jeddah, Jeddah 23218, Saudi Arabia
| | - Manuel Salmeron-Sanchez
- Centre
for the Cellular Microenvironment, School of Molecular Biosciences,
College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced
Research Centre, University of Glasgow, Glasgow G11 6EW, United Kingdom
| | - Bo Su
- Bristol
Dental School Research Laboratories, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, United Kingdom
| | - Matthew J. Dalby
- Centre
for the Cellular Microenvironment, School of Molecular Biosciences,
College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced
Research Centre, University of Glasgow, Glasgow G11 6EW, United Kingdom
| |
Collapse
|
9
|
Halama A, Zaghlool S, Thareja G, Kader S, Al Muftah W, Mook-Kanamori M, Sarwath H, Mohamoud YA, Stephan N, Ameling S, Pucic Baković M, Krumsiek J, Prehn C, Adamski J, Schwenk JM, Friedrich N, Völker U, Wuhrer M, Lauc G, Najafi-Shoushtari SH, Malek JA, Graumann J, Mook-Kanamori D, Schmidt F, Suhre K. A roadmap to the molecular human linking multiomics with population traits and diabetes subtypes. Nat Commun 2024; 15:7111. [PMID: 39160153 PMCID: PMC11333501 DOI: 10.1038/s41467-024-51134-x] [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: 08/01/2023] [Accepted: 07/26/2024] [Indexed: 08/21/2024] Open
Abstract
In-depth multiomic phenotyping provides molecular insights into complex physiological processes and their pathologies. Here, we report on integrating 18 diverse deep molecular phenotyping (omics-) technologies applied to urine, blood, and saliva samples from 391 participants of the multiethnic diabetes Qatar Metabolomics Study of Diabetes (QMDiab). Using 6,304 quantitative molecular traits with 1,221,345 genetic variants, methylation at 470,837 DNA CpG sites, and gene expression of 57,000 transcripts, we determine (1) within-platform partial correlations, (2) between-platform mutual best correlations, and (3) genome-, epigenome-, transcriptome-, and phenome-wide associations. Combined into a molecular network of > 34,000 statistically significant trait-trait links in biofluids, our study portrays "The Molecular Human". We describe the variances explained by each omics in the phenotypes (age, sex, BMI, and diabetes state), platform complementarity, and the inherent correlation structures of multiomics data. Further, we construct multi-molecular network of diabetes subtypes. Finally, we generated an open-access web interface to "The Molecular Human" ( http://comics.metabolomix.com ), providing interactive data exploration and hypotheses generation possibilities.
Collapse
Affiliation(s)
- Anna Halama
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
| | - Shaza Zaghlool
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Gaurav Thareja
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Sara Kader
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Wadha Al Muftah
- Qatar Genome Program, Qatar Foundation, Qatar Science and Technology Park, Innovation Center, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | | | - Hina Sarwath
- Proteomics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | | | - Nisha Stephan
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Sabine Ameling
- German Centre for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | | | - Jan Krumsiek
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jochen M Schwenk
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Solna, Sweden
| | - Nele Friedrich
- German Centre for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- German Centre for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - S Hani Najafi-Shoushtari
- MicroRNA Core Laboratory, Division of Research, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Joel A Malek
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
- Genomics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Johannes Graumann
- Institute of Translational Proteomics, Department of Medicine, Philipps-Universität Marburg, Marburg, Germany
| | - Dennis Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Li T, Yin D, Shi R. Gut-muscle axis mechanism of exercise prevention of sarcopenia. Front Nutr 2024; 11:1418778. [PMID: 39221163 PMCID: PMC11362084 DOI: 10.3389/fnut.2024.1418778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Sarcopenia refers to an age-related systemic skeletal muscle disorder, which is characterized by loss of muscle mass and weakening of muscle strength. Gut microbiota can affect skeletal muscle through a variety of mechanisms. Gut microbiota present distinct features among elderly people and sarcopenia patients, including a decrease in microbial diversity, which might be associated with the quality and function of the skeletal muscle. There might be a gut-muscle axis; where gut microbiota and skeletal muscle may affect each other bi-directionally. Skeletal muscle can affect the biodiversity of the gut microbiota, and the latter can, in turn, affect the anabolism of skeletal muscle. This review examines recent studies exploring the relationship between gut microbiota and skeletal muscle, summarizes the effects of exercise on gut microbiota, and discusses the possible mechanisms of the gut-muscle axis.
Collapse
Affiliation(s)
| | | | - Rengfei Shi
- School of Health and Exercise, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
11
|
Mathews IT, Saminathan P, Henglin M, Liu M, Nadig N, Fang C, Mercader K, Chee SJ, Campbell AM, Patel AA, Tiwari S, Watrous JD, Ramesh K, Dicker M, Dao K, Meyer MA, Jousilahti P, Havulinna AS, Niiranen T, Salomaa V, Joosten LA, Netea MG, Zheng P, Kronenberg M, Patel SP, Gutkind JS, Ottensmeier C, Long T, Kaech SM, Hedrick CC, Cheng S, Jain M, Sharma S. Linoleoyl-lysophosphatidylcholine suppresses immune-related adverse events due to immune checkpoint blockade. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24310974. [PMID: 39148854 PMCID: PMC11326322 DOI: 10.1101/2024.08.07.24310974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Immune related adverse events (irAEs) after immune checkpoint blockade (ICB) therapy occur in a significant proportion of cancer patients. To date, the circulating mediators of ICB-irAEs remain poorly understood. Using non-targeted mass spectrometry, here we identify the circulating bio-active lipid linoleoyl-lysophosphatidylcholine (LPC 18:2) as a modulator of ICB-irAEs. In three independent human studies of ICB treatment for solid tumor, loss of circulating LPC 18:2 preceded the development of severe irAEs across multiple organ systems. In both healthy humans and severe ICB-irAE patients, low LPC 18:2 was found to correlate with high blood neutrophilia. Reduced LPC 18:2 biosynthesis was confirmed in preclinical ICB-irAE models, and LPC 18:2 supplementation in vivo suppressed neutrophilia and tissue inflammation without impacting ICB anti-tumor response. Results indicate that circulating LPC 18:2 suppresses human ICB-irAEs, and LPC 18:2 supplementation may improve ICB outcomes by preventing severe inflammation while maintaining anti-tumor immunity.
Collapse
Affiliation(s)
- Ian T. Mathews
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | | | - Mir Henglin
- Cedars Sinai Medical Center, Los Angeles CA 90048
| | - Mingyue Liu
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201
| | | | - Camille Fang
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Kysha Mercader
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | - Serena J. Chee
- University of Southampton, Southampton, United Kingdom
- Institute of Systems, Molecular and Integrative Biology,University of Liverpool, Liverpool, United Kingdom
| | | | | | - Saumya Tiwari
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Jeramie D. Watrous
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Karthik Ramesh
- Department of Medicine, University of California San Diego, La Jolla CA 92093
| | | | - Khoi Dao
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | | | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Aki S. Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Genomics and Immunometabolism, Life and Medical Sciences Institute, University of Bonn, Germany
| | - Pan Zheng
- Institute of Human Virology, University of Maryland, Baltimore, MD 21201
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Molecular Biology, University of California San Diego, La Jolla CA 92093
| | - Sandip Pravin Patel
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla CA 92037
| | - J. Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla CA 92037
- Department of Pharmacology, University of California San Diego, La Jolla CA 92093
| | - Christian Ottensmeier
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Institute of Systems, Molecular and Integrative Biology,University of Liverpool, Liverpool, United Kingdom
| | - Tao Long
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Susan M. Kaech
- Salk Institute for Biological Studies, La Jolla CA 92037
| | - Catherine C. Hedrick
- Immunology Center of Georgia and Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Susan Cheng
- Cedars Sinai Medical Center, Los Angeles CA 90048
| | - Mohit Jain
- Department of Medicine, University of California San Diego, La Jolla CA 92093
- Sapient Bioanalytics, San Diego CA 92121
| | - Sonia Sharma
- La Jolla Institute for Immunology, La Jolla, CA 92037
| |
Collapse
|
12
|
Zhang Y, Zhao H, Zhao J, Lv W, Jia X, Lu X, Zhao X, Xu G. Quantified Metabolomics and Lipidomics Profiles Reveal Serum Metabolic Alterations and Distinguished Metabolites of Seven Chronic Metabolic Diseases. J Proteome Res 2024; 23:3076-3087. [PMID: 38407022 DOI: 10.1021/acs.jproteome.3c00760] [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] [Indexed: 02/27/2024]
Abstract
The co-occurrence of multiple chronic metabolic diseases is highly prevalent, posing a huge health threat. Clarifying the metabolic associations between them, as well as identifying metabolites which allow discrimination between diseases, will provide new biological insights into their co-occurrence. Herein, we utilized targeted serum metabolomics and lipidomics covering over 700 metabolites to characterize metabolic alterations and associations related to seven chronic metabolic diseases (obesity, hypertension, hyperuricemia, hyperglycemia, hypercholesterolemia, hypertriglyceridemia, fatty liver) from 1626 participants. We identified 454 metabolites were shared among at least two chronic metabolic diseases, accounting for 73.3% of all 619 significant metabolite-disease associations. We found amino acids, lactic acid, 2-hydroxybutyric acid, triacylglycerols (TGs), and diacylglycerols (DGs) showed connectivity across multiple chronic metabolic diseases. Many carnitines were specifically associated with hyperuricemia. The hypercholesterolemia group showed obvious lipid metabolism disorder. Using logistic regression models, we further identified distinguished metabolites of seven chronic metabolic diseases, which exhibited satisfactory area under curve (AUC) values ranging from 0.848 to 1 in discovery and validation sets. Overall, quantitative metabolome and lipidome data sets revealed widespread and interconnected metabolic disorders among seven chronic metabolic diseases. The distinguished metabolites are useful for diagnosing chronic metabolic diseases and provide a reference value for further clinical intervention and management based on metabolomics strategy.
Collapse
Affiliation(s)
- Yuqing Zhang
- School of Chemistry, Dalian University of Technology, Dalian 116024, P. R. China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Hui Zhao
- Department of the Health Checkup Center, The Second Hospital of Dalian Medical University, Dalian 116023, P. R. China
| | - Jinhui Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Science, Beijing 100049, P. R. China
| | - Wangjie Lv
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Science, Beijing 100049, P. R. China
| | - Xueni Jia
- Department of the Health Checkup Center, The Second Hospital of Dalian Medical University, Dalian 116023, P. R. China
| | - Xin Lu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Xinjie Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Guowang Xu
- School of Chemistry, Dalian University of Technology, Dalian 116024, P. R. China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Science, Beijing 100049, P. R. China
| |
Collapse
|
13
|
Trischitta V, Antonucci A, Adamski J, Prehn C, Menzaghi C, Marucci A, Di Paola R. GALNT2 expression is associated with glucose control and serum metabolites in patients with type 2 diabetes. Acta Diabetol 2024; 61:1007-1013. [PMID: 38627282 PMCID: PMC11329529 DOI: 10.1007/s00592-024-02280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/19/2024] [Indexed: 08/09/2024]
Abstract
AIMS Aim of this study was to investigate in type 2 diabetes whether expression level of GALNT2, a positive modulator of insulin sensitivity, is associated with a metabolic signature. METHODS Five different metabolite families, including acylcarnitines, aminoacids, biogenic amines, phospholipids and sphingolipids were investigated in fasting serum of 70 patients with type 2 diabetes, by targeted metabolomics. GALNT2 expression levels were measured in peripheral white blood cells by RT-PCR. The association between GALNT2 expression and serum metabolites was assessed using false discovery rate followed by stepwise selection and, finally, multivariate model including several clinical parameters as confounders. The association between GALNT2 expression and the same clinical parameters was also investigated. RESULTS GALNT2 expression was independently correlated with HbA1c levels (P value = 0.0052), a finding that is the likely consequence of the role of GALNT2 on insulin sensitivity. GALNT2 expression was also independently associated with serum levels of the aminoacid glycine (P value = 0.014) and two biogenic amines phenylethylamine (P value = 0.0065) and taurine (P value = 0.0011). The association of GALNT2 expression with HbA1c was not mediated by these three metabolites. CONCLUSIONS Our data indicate that in type 2 diabetes the expression of GALNT2 is associated with several serum metabolites. This association needs to be further investigated to understand in depth its role in mediating the effect of GALNT2 on insulin sensitivity, glucose control and other clinical features in people with diabetes.
Collapse
Affiliation(s)
- Vincenzo Trischitta
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo Della Sofferenza, 71013, San Giovanni Rotondo, Foggia, Italy.
- Department of Experimental Medicine, Sapienza University, 00161, Rome, Italy.
| | - Alessandra Antonucci
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo Della Sofferenza, 71013, San Giovanni Rotondo, Foggia, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore, 117597, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Cornelia Prehn
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Claudia Menzaghi
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo Della Sofferenza, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Antonella Marucci
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo Della Sofferenza, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Rosa Di Paola
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo Della Sofferenza, 71013, San Giovanni Rotondo, Foggia, Italy.
| |
Collapse
|
14
|
Long J, Fang Q, Shi Z, Miao Z, Yan D. Integrated biomarker profiling for predicting the response of type 2 diabetes to metformin. Diabetes Obes Metab 2024; 26:3439-3447. [PMID: 38828802 DOI: 10.1111/dom.15689] [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] [Received: 03/26/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 06/05/2024]
Abstract
AIM To explore biomarkers that can predict the response of type 2 diabetes (T2D) patients to metformin at an early stage to provide better treatment for T2D. METHODS T2D patients with (responders) or without response (non-responders) to metformin were recruited, and their serum samples were used for metabolomic analysis to identify candidate biomarkers. Moreover, the efficacy of metformin was verified by insulin-resistant mice, and the candidate biomarkers were verified to determine the biomarkers. Five different machine learning methods were used to construct the integrated biomarker profiling (IBP) with the biomarkers to predict the response of T2D patients to metformin. RESULTS A total of 73 responders and 63 non-responders were recruited, and 88 differential metabolites were identified in the serum samples. After being verified in mice, 19 of the 88 were considered as candidate biomarkers. Next, after metformin regulation, nine candidate biomarkers were confirmed as the biomarkers. After comparing five machine learning models, the nine biomarkers were constructed into the IBP for predicting the response of T2D patients to metformin based on the Naïve Bayes classifier, which was verified with an accuracy of 89.70%. CONCLUSIONS The IBP composed of nine biomarkers can be used to predict the response of T2D patients to metformin, enabling clinicians to start a combined medication strategy as soon as possible if T2D patients do not respond to metformin.
Collapse
Affiliation(s)
- Jianglan Long
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qiushi Fang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhe Shi
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zenghui Miao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dan Yan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
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
|
16
|
Li L, Ding S, Wang W, Yang L, Wilson G, Sa Y, Zhang Y, Chen J, Ma X. Serum metabolomics reveals the metabolic profile and potential biomarkers of ankylosing spondylitis. Mol Omics 2024. [PMID: 38984672 DOI: 10.1039/d4mo00076e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Ankylosing spondylitis (AS) is a chronic systemic inflammatory disease that significantly impairs physical function in young individuals. However, the identification of radiographic changes in AS is frequently delayed, and the diagnostic efficacy of biomarkers like HLA-B27 remains moderately effective, with unsatisfactory sensitivity and specificity. In contrast to existing literature, our current experiment utilized a larger sample size and employed both untargeted and targeted UHPLC-QTOF-MS/MS based metabolomics to identify the metabolite profile and potential biomarkers of AS. The results indicated a notable divergence between the two groups, and a total of 170 different metabolites were identified, which were associated with the 6 primary metabolic pathways exhibiting a correlation with AS. Among these, 26 metabolites exhibited high sensitivity and specificity with area under curve (AUC) values greater than 0.8. Subsequent targeted quantitative analysis discovered 3 metabolites, namely 3-amino-2-piperidone, hypoxanthine and octadecylamine, exhibiting excellent distinguishing ability based on the results of the ROC curve and the Random Forest model, thus qualifying as potential biomarkers for AS. Summarily, our untargeted and targeted metabolomics investigation offers novel and precise insights into potential biomarkers for AS, potentially enhancing diagnostic capabilities and furthering the comprehension of the condition's pathophysiology.
Collapse
Affiliation(s)
- Liuyan Li
- School of Pharmacy, Ningxia Medical University, 1160 Shenli Street, Yinchuan, 750004, China.
| | - Shuqin Ding
- School of Pharmacy, Ningxia Medical University, 1160 Shenli Street, Yinchuan, 750004, China.
| | - Weibiao Wang
- School of Pharmacy, Ningxia Medical University, 1160 Shenli Street, Yinchuan, 750004, China.
| | - Lingling Yang
- School of Pharmacy, Ningxia Medical University, 1160 Shenli Street, Yinchuan, 750004, China.
| | - Gidion Wilson
- School of Pharmacy, Ningxia Medical University, 1160 Shenli Street, Yinchuan, 750004, China.
| | - Yuping Sa
- School of Pharmacy, Ningxia Medical University, 1160 Shenli Street, Yinchuan, 750004, China.
| | - Yue Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shenli Street, Yinchuan, 750004, China.
| | - Jianyu Chen
- Fujian University of Traditional Chinese Medicine, No. 1, Huatuo Road, Minhoushangjie, Fuzhou 350122, China.
| | - Xueqin Ma
- School of Pharmacy, Ningxia Medical University, 1160 Shenli Street, Yinchuan, 750004, China.
| |
Collapse
|
17
|
Abdualkader AM, Karwi QG, Lopaschuk GD, Al Batran R. The role of branched-chain amino acids and their downstream metabolites in mediating insulin resistance. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13040. [PMID: 39007094 PMCID: PMC11239365 DOI: 10.3389/jpps.2024.13040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024]
Abstract
Elevated levels of circulating branched-chain amino acids (BCAAs) and their associated metabolites have been strongly linked to insulin resistance and type 2 diabetes. Despite extensive research, the precise mechanisms linking increased BCAA levels with these conditions remain elusive. In this review, we highlight the key organs involved in maintaining BCAA homeostasis and discuss how obesity and insulin resistance disrupt the intricate interplay among these organs, thus affecting BCAA balance. Additionally, we outline recent research shedding light on the impact of tissue-specific or systemic modulation of BCAA metabolism on circulating BCAA levels, their metabolites, and insulin sensitivity, while also identifying specific knowledge gaps and areas requiring further investigation. Finally, we summarize the effects of BCAA supplementation or restriction on obesity and insulin sensitivity.
Collapse
Affiliation(s)
- Abdualrahman Mohammed Abdualkader
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
- Montreal Diabetes Research Center, Montréal, QC, Canada
- Cardiometabolic Health, Diabetes and Obesity Research Network, Montréal, QC, Canada
| | - Qutuba G. Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Gary D. Lopaschuk
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Rami Al Batran
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
- Montreal Diabetes Research Center, Montréal, QC, Canada
- Cardiometabolic Health, Diabetes and Obesity Research Network, Montréal, QC, Canada
| |
Collapse
|
18
|
Rojo-López MI, Barranco-Altirriba M, Rossell J, Antentas M, Castelblanco E, Yanes O, Weber RJM, Lloyd GR, Winder C, Dunn WB, Julve J, Granado-Casas M, Mauricio D. The Lipidomic Profile Is Associated with the Dietary Pattern in Subjects with and without Diabetes Mellitus from a Mediterranean Area. Nutrients 2024; 16:1805. [PMID: 38931159 PMCID: PMC11206394 DOI: 10.3390/nu16121805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Lipid functions can be influenced by genetics, age, disease states, and lifestyle factors, particularly dietary patterns, which are crucial in diabetes management. Lipidomics is an expanding field involving the comprehensive exploration of lipids from biological samples. In this cross-sectional study, 396 participants from a Mediterranean region, including individuals with type 1 diabetes (T1D), type 2 diabetes (T2D), and non-diabetic individuals, underwent lipidomic profiling and dietary assessment. Participants completed validated food frequency questionnaires, and lipid analysis was conducted using ultra-high-performance liquid chromatography coupled with mass spectrometry (UHPLC/MS). Multiple linear regression models were used to determine the association between lipid features and dietary patterns. Across all subjects, acylcarnitines (AcCa) and triglycerides (TG) displayed negative associations with the alternate Healthy Eating Index (aHEI), indicating a link between lipidomic profiles and dietary habits. Various lipid species (LS) showed positive and negative associations with dietary carbohydrates, fats, and proteins. Notably, in the interaction analysis between diabetes and the aHEI, we found some lysophosphatidylcholines (LPC) that showed a similar direction with respect to aHEI in non-diabetic individuals and T2D subjects, while an opposite direction was observed in T1D subjects. The study highlights the significant association between lipidomic profiles and dietary habits in people with and without diabetes, particularly emphasizing the role of healthy dietary choices, as reflected by the aHEI, in modulating lipid concentrations. These findings underscore the importance of dietary interventions to improve metabolic health outcomes, especially in the context of diabetes management.
Collapse
Affiliation(s)
- Marina Idalia Rojo-López
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (M.B.-A.); (J.R.); (M.A.); (J.J.)
| | - Maria Barranco-Altirriba
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (M.B.-A.); (J.R.); (M.A.); (J.J.)
- B2SLab, Departament d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, 08028 Barcelona, Spain
- Networking Biomedical Research Centre in the Subject Area of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Barcelona, Spain
| | - Joana Rossell
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (M.B.-A.); (J.R.); (M.A.); (J.J.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Maria Antentas
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (M.B.-A.); (J.R.); (M.A.); (J.J.)
| | - Esmeralda Castelblanco
- Department of Internal Medicine, Endocrinology, Metabolism and Lipid Research Division, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Oscar Yanes
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Department of Electronic Engineering, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Ralf J. M. Weber
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (R.J.M.W.); (G.R.L.); (C.W.); (W.B.D.)
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Gavin R. Lloyd
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (R.J.M.W.); (G.R.L.); (C.W.); (W.B.D.)
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Catherine Winder
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (R.J.M.W.); (G.R.L.); (C.W.); (W.B.D.)
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Warwick B. Dunn
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (R.J.M.W.); (G.R.L.); (C.W.); (W.B.D.)
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Josep Julve
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (M.B.-A.); (J.R.); (M.A.); (J.J.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Minerva Granado-Casas
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Department of Nursing and Physiotherapy, University of Lleida, 25198 Lleida, Spain
- Research Group of Health Care (GreCS), IRBLleida, 25198 Lleida, Spain
| | - Dídac Mauricio
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041 Barcelona, Spain; (M.I.R.-L.); (M.B.-A.); (J.R.); (M.A.); (J.J.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Faculty of Medicine, University of Vic (UVIC/UCC), 08500 Vic, Spain
| |
Collapse
|
19
|
Zhang F, Shan S, Fu C, Guo S, Liu C, Wang S. Advanced Mass Spectrometry-Based Biomarker Identification for Metabolomics of Diabetes Mellitus and Its Complications. Molecules 2024; 29:2530. [PMID: 38893405 PMCID: PMC11173766 DOI: 10.3390/molecules29112530] [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: 02/08/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 06/21/2024] Open
Abstract
Over the years, there has been notable progress in understanding the pathogenesis and treatment modalities of diabetes and its complications, including the application of metabolomics in the study of diabetes, capturing attention from researchers worldwide. Advanced mass spectrometry, including gas chromatography-tandem mass spectrometry (GC-MS/MS), liquid chromatography-tandem mass spectrometry (LC-MS/MS), and ultra-performance liquid chromatography coupled to electrospray ionization quadrupole time-of-flight mass spectrometry (UPLC-ESI-Q-TOF-MS), etc., has significantly broadened the spectrum of detectable metabolites, even at lower concentrations. Advanced mass spectrometry has emerged as a powerful tool in diabetes research, particularly in the context of metabolomics. By leveraging the precision and sensitivity of advanced mass spectrometry techniques, researchers have unlocked a wealth of information within the metabolome. This technology has enabled the identification and quantification of potential biomarkers associated with diabetes and its complications, providing new ideas and methods for clinical diagnostics and metabolic studies. Moreover, it offers a less invasive, or even non-invasive, means of tracking disease progression, evaluating treatment efficacy, and understanding the underlying metabolic alterations in diabetes. This paper summarizes advanced mass spectrometry for the application of metabolomics in diabetes mellitus, gestational diabetes mellitus, diabetic peripheral neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic encephalopathy, diabetic cardiomyopathy, and diabetic foot ulcers and organizes some of the potential biomarkers of the different complications with the aim of providing ideas and methods for subsequent in-depth metabolic research and searching for new ways of treating the disease.
Collapse
Affiliation(s)
- Feixue Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Medical College, Hubei University of Science and Technology, Xianning 437100, China; (F.Z.); (C.F.); (S.G.)
| | - Shan Shan
- College of Life Science, National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang 330022, China;
| | - Chenlu Fu
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Medical College, Hubei University of Science and Technology, Xianning 437100, China; (F.Z.); (C.F.); (S.G.)
- School of Pharmacy, Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Medical College, Hubei University of Science and Technology, Xianning 437100, China; (F.Z.); (C.F.); (S.G.)
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Medical College, Hubei University of Science and Technology, Xianning 437100, China; (F.Z.); (C.F.); (S.G.)
| | - Shuanglong Wang
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, East China University of Technology, Nanchang 330013, China
| |
Collapse
|
20
|
Li H, Li L, Huang QQ, Yang SY, Zou JJ, Xiao F, Xiang Q, Liu X, Yu R. Global status and trends of metabolomics in diabetes: A literature visualization knowledge graph study. World J Diabetes 2024; 15:1021-1044. [PMID: 38766424 PMCID: PMC11099375 DOI: 10.4239/wjd.v15.i5.1021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/28/2024] [Accepted: 03/18/2024] [Indexed: 05/10/2024] Open
Abstract
BACKGROUND Diabetes is a metabolic disease characterized by hyperglycemia, which has increased the global medical burden and is also the main cause of death in most countries. AIM To understand the knowledge structure of global development status, research focus, and future trend of the relationship between diabetes and metabolomics in the past 20 years. METHODS The articles about the relationship between diabetes and metabolomics in the Web of Science Core Collection were retrieved from 2002 to October 23, 2023, and the relevant information was analyzed using CiteSpace6.2.2R (CiteSpace), VOSviewer6.1.18 (VOSviewer), and Bibliometrix software under R language. RESULTS A total of 3123 publications were included from 2002 to 2022. In the past two decades, the number of publications and citations in this field has continued to increase. The United States, China, Germany, the United Kingdom, and other relevant funds, institutions, and authors have significantly contributed to this field. Scientific Reports and PLoS One are the journals with the most publications and the most citations. Through keyword co-occurrence and cluster analysis, the closely related keywords are "insulin resistance", "risk", "obesity", "oxidative stress", "metabolomics", "metabolites" and "biomarkers". Keyword clustering included cardiovascular disease, gut microbiota, metabonomics, diabetic nephropathy, molecular docking, gestational diabetes mellitus, oxidative stress, and insulin resistance. Burst detection analysis of keyword depicted that "Gene", "microbiota", "validation", "kidney disease", "antioxidant activity", "untargeted metabolomics", "management", and "accumulation" are knowledge frontiers in recent years. CONCLUSION The relationship between metabolomics and diabetes is receiving extensive attention. Diabetic nephropathy, diabetic cardiovascular disease, and kidney disease are key diseases for future research in this field. Gut microbiota, molecular docking, and untargeted metabolomics are key research directions in the future. Antioxidant activity, gene, validation, mass spectrometry, management, and accumulation are at the forefront of knowledge frontiers in this field.
Collapse
Affiliation(s)
- Hong Li
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Liu Li
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Qiu-Qing Huang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Si-Yao Yang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Jun-Ju Zou
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Fan Xiao
- College of International Education, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Qin Xiang
- Department of Science and Technology, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Xiu Liu
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Rong Yu
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
- College of Graduate, Hunan University of Chinese Medicine, Hunan Changsha, Hunan Province, China
| |
Collapse
|
21
|
Alosaimi ME, Alotaibi BS, Abduljabbar MH, Alnemari RM, Almalki AH, Serag A. Therapeutic implications of dapagliflozin on the metabolomics profile of diabetic rats: A GC-MS investigation coupled with multivariate analysis. J Pharm Biomed Anal 2024; 242:116018. [PMID: 38341926 DOI: 10.1016/j.jpba.2024.116018] [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: 01/09/2024] [Revised: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Diabetes mellitus is a complex metabolic disorder with systemic implications, necessitating the search for reliable biomarkers and therapeutic strategies. This study investigates the metabolomics profile alterations in diabetic rats, with a focus on the therapeutic effects of Dapagliflozin, a drug known to inhibit renal glucose reabsorption, using Gas Chromatography-Mass Spectrometry analysis. METHODS A GC-MS based metabolomics approach combined with multivariate and univariate statistical analyses was utilized to study serum samples from a diabetic model of Wistar rats, treated with dapagliflozin. Metabolomics pathways analysis was also performed to identify the altered metabolic pathways associated with the disease and the intervention. RESULTS Dapagliflozin treatment in diabetic rats resulted in normalized levels of metabolites associated with insulin resistance, notably branched-chain and aromatic amino acids. Improvements in glycine metabolism were observed, suggesting a modulatory role of the drug. Additionally, reduced palmitic acid levels indicated an alleviation of lipotoxic effects. The metabolic changes indicate a restorative effect of dapagliflozin on diabetes-induced metabolic perturbations. CONCLUSIONS The comprehensive metabolomics analysis demonstrated the potential of GC-MS in revealing significant metabolic pathway alterations due to dapagliflozin treatment in diabetic model rats. The therapy induced normalization of key metabolic disturbances, providing insights that could advance personalized diabetes mellitus management and therapeutic monitoring, highlighting the utility of metabolomics in understanding drug mechanisms and effects.
Collapse
Affiliation(s)
- Manal E Alosaimi
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Maram H Abduljabbar
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, 21944 Taif, Saudi Arabia
| | - Reem M Alnemari
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Atiah H Almalki
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, 21944 Taif, Saudi Arabia; Addiction and Neuroscience Research Unit, Health Science Campus, Taif University, P.O. Box 11099, 21944 Taif, Saudi Arabia
| | - Ahmed Serag
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, 11751 Nasr City, Cairo, Egypt.
| |
Collapse
|
22
|
Bertran L, Capellades J, Abelló S, Aguilar C, Auguet T, Richart C. Untargeted lipidomics analysis in women with morbid obesity and type 2 diabetes mellitus: A comprehensive study. PLoS One 2024; 19:e0303569. [PMID: 38743756 PMCID: PMC11093320 DOI: 10.1371/journal.pone.0303569] [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: 03/03/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
There is a phenotype of obese individuals termed metabolically healthy obese that present a reduced cardiometabolic risk. This phenotype offers a valuable model for investigating the mechanisms connecting obesity and metabolic alterations such as Type 2 Diabetes Mellitus (T2DM). Previously, in an untargeted metabolomics analysis in a cohort of morbidly obese women, we observed a different lipid metabolite pattern between metabolically healthy morbid obese individuals and those with associated T2DM. To validate these findings, we have performed a complementary study of lipidomics. In this study, we assessed a liquid chromatography coupled to a mass spectrometer untargeted lipidomic analysis on serum samples from 209 women, 73 normal-weight women (control group) and 136 morbid obese women. From those, 65 metabolically healthy morbid obese and 71 with associated T2DM. In this work, we find elevated levels of ceramides, sphingomyelins, diacyl and triacylglycerols, fatty acids, and phosphoethanolamines in morbid obese vs normal weight. Conversely, decreased levels of acylcarnitines, bile acids, lyso-phosphatidylcholines, phosphatidylcholines (PC), phosphatidylinositols, and phosphoethanolamine PE (O-38:4) were noted. Furthermore, comparing morbid obese women with T2DM vs metabolically healthy MO, a distinct lipid profile emerged, featuring increased levels of metabolites: deoxycholic acid, diacylglycerol DG (36:2), triacylglycerols, phosphatidylcholines, phosphoethanolamines, phosphatidylinositols, and lyso-phosphatidylinositol LPI (16:0). To conclude, analysing both comparatives, we observed decreased levels of deoxycholic acid, PC (34:3), and PE (O-38:4) in morbid obese women vs normal-weight. Conversely, we found elevated levels of these lipids in morbid obese women with T2DM vs metabolically healthy MO. These profiles of metabolites could be explored for the research as potential markers of metabolic risk of T2DM in morbid obese women.
Collapse
Affiliation(s)
- Laia Bertran
- Department of Medicine and Surgery, Study Group on Metabolic Diseases Associated with Insulin-Resistance (GEMMAIR), Rovira i Virgili University, Hospital Universitari de Tarragona Joan XXIII, IISPV, Tarragona, Spain
| | - Jordi Capellades
- Department of Electronic, Electric and Automatic Engineering, Higher Technical School of Engineering, Rovira i Virgili University, IISPV, Tarragona, Spain
| | - Sonia Abelló
- Scientific and Technical Service, Rovira i Virgili University, Tarragona, Spain
| | - Carmen Aguilar
- Department of Medicine and Surgery, Study Group on Metabolic Diseases Associated with Insulin-Resistance (GEMMAIR), Rovira i Virgili University, Hospital Universitari de Tarragona Joan XXIII, IISPV, Tarragona, Spain
| | - Teresa Auguet
- Department of Medicine and Surgery, Study Group on Metabolic Diseases Associated with Insulin-Resistance (GEMMAIR), Rovira i Virgili University, Hospital Universitari de Tarragona Joan XXIII, IISPV, Tarragona, Spain
| | - Cristóbal Richart
- Department of Medicine and Surgery, Study Group on Metabolic Diseases Associated with Insulin-Resistance (GEMMAIR), Rovira i Virgili University, Hospital Universitari de Tarragona Joan XXIII, IISPV, Tarragona, Spain
| |
Collapse
|
23
|
Jabbar Al‐Rikabi S, Etemadi A, Morad M, Nowrouzi A, Panahi G, Mondeali M, Toorani‐ghazvini M, Nasli‐Esfahani E, Razi F, Bandarian F. Metabolomics Signature in Prediabetes and Diabetes: Insights From Tandem Mass Spectrometry Analysis. Endocrinol Diabetes Metab 2024; 7:e00484. [PMID: 38739122 PMCID: PMC11090150 DOI: 10.1002/edm2.484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
OBJECTIVE This study investigates the metabolic differences between normal, prediabetic and diabetic patients with good and poor glycaemic control (GGC and PGC). DESIGN In this study, 1102 individuals were included, and 50 metabolites were analysed using tandem mass spectrometry. The diabetes diagnosis and treatment standards of the American Diabetes Association (ADA) were used to classify patients. METHODS The nearest neighbour method was used to match controls and cases in each group on the basis of age, sex and BMI. Factor analysis was used to reduce the number of variables and find influential underlying factors. Finally, Pearson's correlation coefficient was used to check the correlation between both glucose and HbAc1 as independent factors with binary classes. RESULTS Amino acids such as glycine, serine and proline, and acylcarnitines (AcylCs) such as C16 and C18 showed significant differences between the prediabetes and normal groups. Additionally, several metabolites, including C0, C5, C8 and C16, showed significant differences between the diabetes and normal groups. Moreover, the study found that several metabolites significantly differed between the GGC and PGC diabetes groups, such as C2, C6, C10, C16 and C18. The correlation analysis revealed that glucose and HbA1c levels significantly correlated with several metabolites, including glycine, serine and C16, in both the prediabetes and diabetes groups. Additionally, the correlation analysis showed that HbA1c significantly correlated with several metabolites, such as C2, C5 and C18, in the controlled and uncontrolled diabetes groups. CONCLUSIONS These findings could help identify new biomarkers or underlying markers for the early detection and management of diabetes.
Collapse
Affiliation(s)
| | - Ali Etemadi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences InstituteTehran University of Medical SciencesTehranIran
- Medical Biotechnology Department, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Maher Mohammed Morad
- Department of Clinical Biochemistry, School of MedicineTehran University of Medical SciencesTehranIran
| | - Azin Nowrouzi
- Department of Clinical Biochemistry, School of MedicineTehran University of Medical SciencesTehranIran
| | | | - Mozhgan Mondeali
- Department of Medical Genetics, School of MedicineTehran University of Medical SciencesTehranIran
| | - Mahsa Toorani‐ghazvini
- Medical Biotechnology Department, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Ensieh Nasli‐Esfahani
- Diabetes Research CenterEndocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical SciencesTehranIran
| | - Farideh Razi
- Metabolomics and Genomics Research CenterEndocrinology and Metabolism Molecular‐Cellular Sciences Institute, Tehran University of Medical SciencesTehranIran
| | - Fatemeh Bandarian
- Metabolomics and Genomics Research CenterEndocrinology and Metabolism Molecular‐Cellular Sciences Institute, Tehran University of Medical SciencesTehranIran
| |
Collapse
|
24
|
Yu S, Han S, Shi M, Harada M, Ge J, Li X, Cai X, Heier M, Karstenmüller G, Suhre K, Gieger C, Koenig W, Rathmann W, Peters A, Wang-Sattler R. Prediction of Myocardial Infarction Using a Combined Generative Adversarial Network Model and Feature-Enhanced Loss Function. Metabolites 2024; 14:258. [PMID: 38786735 PMCID: PMC11122941 DOI: 10.3390/metabo14050258] [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: 04/03/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Accurate risk prediction for myocardial infarction (MI) is crucial for preventive strategies, given its significant impact on global mortality and morbidity. Here, we propose a novel deep-learning approach to enhance the prediction of incident MI cases by incorporating metabolomics alongside clinical risk factors. We utilized data from the KORA cohort, including the baseline S4 and follow-up F4 studies, consisting of 1454 participants without prior history of MI. The dataset comprised 19 clinical variables and 363 metabolites. Due to the imbalanced nature of the dataset (78 observed MI cases and 1376 non-MI individuals), we employed a generative adversarial network (GAN) model to generate new incident cases, augmenting the dataset and improving feature representation. To predict MI, we further utilized multi-layer perceptron (MLP) models in conjunction with the synthetic minority oversampling technique (SMOTE) and edited nearest neighbor (ENN) methods to address overfitting and underfitting issues, particularly when dealing with imbalanced datasets. To enhance prediction accuracy, we propose a novel GAN for feature-enhanced (GFE) loss function. The GFE loss function resulted in an approximate 2% improvement in prediction accuracy, yielding a final accuracy of 70%. Furthermore, we evaluated the contribution of each clinical variable and metabolite to the predictive model and identified the 10 most significant variables, including glucose tolerance, sex, and physical activity. This is the first study to construct a deep-learning approach for producing 7-year MI predictions using the newly proposed loss function. Our findings demonstrate the promising potential of our technique in identifying novel biomarkers for MI prediction.
Collapse
Affiliation(s)
- Shixiang Yu
- TUM School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (S.Y.); (S.H.); (M.S.); (J.G.)
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Siyu Han
- TUM School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (S.Y.); (S.H.); (M.S.); (J.G.)
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Mengya Shi
- TUM School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (S.Y.); (S.H.); (M.S.); (J.G.)
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Makoto Harada
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Jianhong Ge
- TUM School of Medicine and Health, Technical University of Munich, 81675 München, Germany; (S.Y.); (S.H.); (M.S.); (J.G.)
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Xuening Li
- Biocomputing R&D Department, Beijing Huanyang Bole Consulting Co., Ltd., Beijing 100010, China;
| | - Xiang Cai
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin 541214, China;
| | - Margit Heier
- KORA Study Centre, University Hospital of Augsburg, 86153 Augsburg, Germany;
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Gabi Karstenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine and Director of the Bioinformatics Core, Doha 24144, Qatar;
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, 80636 München, Germany;
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Pettenkofer School of Public Health, Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 München, Germany
| | - Rui Wang-Sattler
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| |
Collapse
|
25
|
Ratter-Rieck JM, Shi M, Suhre K, Prehn C, Adamski J, Rathmann W, Thorand B, Roden M, Peters A, Wang-Sattler R, Herder C. Omentin associates with serum metabolite profiles indicating lower diabetes risk: KORA F4 Study. BMJ Open Diabetes Res Care 2024; 12:e003865. [PMID: 38442989 PMCID: PMC11148672 DOI: 10.1136/bmjdrc-2023-003865] [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: 10/24/2023] [Accepted: 02/01/2024] [Indexed: 03/07/2024] Open
Abstract
INTRODUCTION Circulating omentin levels have been positively associated with insulin sensitivity. Although a role for adiponectin in this relationship has been suggested, underlying mechanisms remain elusive. In order to reveal the relationship between omentin and systemic metabolism, this study aimed to investigate associations of serum concentrations of omentin and metabolites. RESEARCH DESIGN AND METHODS This study is based on 1124 participants aged 61-82 years from the population-based KORA (Cooperative Health Research in the Region of Augsburg) F4 Study, for whom both serum omentin levels and metabolite concentration profiles were available. Associations were assessed with five multivariable regression models, which were stepwise adjusted for multiple potential confounders, including age, sex, body mass index, waist-to-hip ratio, lifestyle markers (physical activity, smoking behavior and alcohol consumption), serum adiponectin levels, high-density lipoprotein cholesterol, use of lipid-lowering or anti-inflammatory medication, history of myocardial infarction and stroke, homeostasis model assessment 2 of insulin resistance, diabetes status, and use of oral glucose-lowering medication and insulin. RESULTS Omentin levels significantly associated with multiple metabolites including amino acids, acylcarnitines, and lipids (eg, sphingomyelins and phosphatidylcholines (PCs)). Positive associations for several PCs, such as diacyl (PC aa C32:1) and alkyl-alkyl (PC ae C32:2), were significant in models 1-4, whereas those with hydroxytetradecenoylcarnitine (C14:1-OH) were significant in all five models. Omentin concentrations were negatively associated with several metabolite ratios, such as the valine-to-PC ae C32:2 and the serine-to-PC ae C32:2 ratios in most models. CONCLUSIONS Our results suggest that omentin may influence insulin sensitivity and diabetes risk by changing systemic lipid metabolism, but further mechanistic studies investigating effects of omentin on metabolism of insulin-sensitive tissues are needed.
Collapse
Affiliation(s)
- Jacqueline M Ratter-Rieck
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Mengya Shi
- TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Partner Neuherberg, Neuherberg, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jerzy Adamski
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Wolfgang Rathmann
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Barbara Thorand
- German Center for Diabetes Research, Partner Neuherberg, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Annette Peters
- German Center for Diabetes Research, Partner Neuherberg, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rui Wang-Sattler
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Partner Neuherberg, Neuherberg, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
26
|
Ghaffari MH, Daniel JB, Sadri H, Schuchardt S, Martín-Tereso J, Sauerwein H. Longitudinal characterization of the metabolome of dairy cows transitioning from one lactation to the next: Investigations in blood serum. J Dairy Sci 2024; 107:1263-1285. [PMID: 37777004 DOI: 10.3168/jds.2023-23841] [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: 06/05/2023] [Accepted: 09/07/2023] [Indexed: 10/02/2023]
Abstract
The objective of this study was to characterize changes in the serum metabolome and various indicators of oxidative balance in dairy cows starting 2 wk before dry-off and continuing until wk 16 of lactation. Twelve Holstein dairy cows (body weight 745 ± 71 kg, body condition score 3.43 ± 0.66; mean ± SD) were housed in a tiestall barn from 10 wk before to 16 wk after parturition. Cows were dried off 6 wk before the expected calving date (mean dry period length = 42 d). From 8 wk before calving to 16 wk after calving, blood samples were taken weekly to study redox metabolism by determining antioxidant capacity, measured as the ferric-reducing ability of plasma, reactive oxidative metabolites, oxidative stress index, oxidative damage of lipids, measured as thiobarbituric acid reactive substances, and glutathione peroxidase activity. According to these results, dairy cows had the lowest serum antioxidant capacity and greater levels of oxidative stress during the dry-off period and the early postpartum period. For metabolomics, a subset of serum samples including wk -7 (before dry-off), -5 (after dry-off), -1, 1, 5, 10, and 15 relative to calving were used. A targeted metabolomics approach was performed using liquid chromatography and flow injection with electrospray ionization triple quadrupole mass spectrometry using the MxP Quant 500 kit (Biocrates Life Sciences AG). A total of 240 metabolites in serum were used in the final data analysis. Principal component analysis revealed a clear separation by days of sampling, indicating a remarkable shift in metabolic phenotype between the dry period and late and early lactation. Changes in many non-lipid metabolites associated with one-carbon metabolism, the tricarboxylic acid cycle, the urea cycle, and AA catabolism were observed in the study, with changes in AA serum concentrations likely related to factors such as energy and nitrogen balance, digestive efficiency, and changing diets. The study confirmed an extensive remodeling of the serum lipidome in peripartum dairy cows, highlighting the importance of changes in acylcarnitine (acylCN), phosphatidylcholines (PC), and triacylglycerols (TG), as they play a crucial role in lipid metabolism. Results showed that short-chain acylCN increased after dry-off and decreased thereafter, whereas lipid-derived acylCN increased around parturition, suggesting that more fatty acids could enter mitochondria. Phospholipids and sphingolipids in serum showed changes during lactation. In particular, concentrations of sphingomyelins, PC, and lysoPC decreased around calving but increased in mid- and late lactation. In contrast, concentrations of TG remained consistently low after parturition. The serum concentrations of bile acids fluctuated during the dry period and lactation, with glycocholic acid, cholic acid, glycodeoxycholic acid, and taurocholic acid showing the greatest concentrations. These changes are likely due to the interplay of diet, liver function, and the ability of the gut microbiota to convert primary to secondary bile acids. Overall, these descriptive results may aid in hypothesis generation and in the design and interpretation of future metabolite-based studies in dairy cows. Furthermore, they contribute to our understanding of the physiological ranges in serum metabolites relative to the lactation cycle of the dairy cow.
Collapse
Affiliation(s)
- M H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - J B Daniel
- Trouw Nutrition R&D, 3800 AG, Amersfoort, the Netherlands.
| | - H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 5166616471 Tabriz, Iran
| | - S Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | | | - H Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
27
|
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: 0] [Impact Index Per Article: 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
|
28
|
Sira J, Zhang X, Gao L, Wabo TMC, Li J, Akiti C, Zhang W, Sun D. Effects of Inorganic Arsenic on Type 2 Diabetes Mellitus In Vivo: the Roles and Mechanisms of miRNAs. Biol Trace Elem Res 2024; 202:111-121. [PMID: 37131019 DOI: 10.1007/s12011-023-03669-1] [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: 01/17/2023] [Accepted: 04/12/2023] [Indexed: 05/04/2023]
Abstract
Accumulating studies have shown that chronic exposure to iAs correlates with an increased incidence of diabetes. In recent years, miRNA dysfunction has emerged both as a response to iAs exposure and independently as candidate drivers of metabolic phenotypes such as T2DM. However, few miRNAs have been profiled during the progression of diabetes after iAs exposure in vivo. In the present study, high iAs (10 mg/L NaAsO2) exposure mice models of C57BKS/Leprdb (db/db) and C57BLKS/J (WT) were established through the drinking water, the exposure duration was 14 weeks. The results showed that high iAs exposure induced no significant changes in FBG levels in either db/db or WT mice. FBI levels, C-peptide content, and HOMA-IR levels were significantly increased, and glycogen levels in the livers were significantly lower in arsenic-exposed db/db mice. HOMA-β% was decreased significantly in WT mice exposed to high iAs. In addition, more different metabolites were found in the arsenic-exposed group than the control group in db/db mice, mainly involved in the lipid metabolism pathway. Highly expressed glucose, insulin, and lipid metabolism-related miRNAs were selected, including miR-29a-3p, miR-143-3p, miR-181a-3p, miR-122-3p, miR-22-3p, and miR-16-3p. And a series of target genes were chosen for analysis, such as ptp1b, irs1, irs2, sirt1, g6pase, pepck and glut4. The results showed that, the axles of miR-181a-3p-irs2, miR-181a-3p-sirt1, miR-22-3p-sirt1, and miR-122-3p-ptp1b in db/db mice, and miR-22-3p-sirt1, miR-16-3p-glut4 in WT mice could be considered promising targets to explore the mechanisms and therapeutic aspects of T2DM after exposure to high iAs.
Collapse
Affiliation(s)
- Jackson Sira
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
- Department of Biomedical Sciences, Faculty of Sciences, University of Ngaoundéré, P.O Box 454, Ngaoundéré, Cameroon
| | - Xiaodan Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Lin Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Therese Martin Cheteu Wabo
- Department of Biomedical Sciences, Faculty of Sciences, University of Ngaoundéré, P.O Box 454, Ngaoundéré, Cameroon
- Department of Nutrition and Food Hygiene, Harbin Medical University, Harbin, 150081, China
| | - Jinyu Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Caselia Akiti
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Wei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China.
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China.
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China.
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China.
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China.
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China.
| |
Collapse
|
29
|
Zhang B, Zhang X, Luo Z, Ren J, Yu X, Zhao H, Wang Y, Zhang W, Tian W, Wei X, Ding Q, Yang H, Jin Z, Tong X, Wang J, Zhao L. Microbiome and metabolome dysbiosis analysis in impaired glucose tolerance for the prediction of progression to diabetes mellitus. J Genet Genomics 2024; 51:75-86. [PMID: 37652264 DOI: 10.1016/j.jgg.2023.08.005] [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: 04/29/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023]
Abstract
Gut microbiota and circulating metabolite dysbiosis predate important pathological changes in glucose metabolic disorders; however, comprehensive studies on impaired glucose tolerance (IGT), a diabetes mellitus (DM) precursor, are lacking. Here, we perform metagenomic sequencing and metabolomics on 47 pairs of individuals with IGT and newly diagnosed DM and 46 controls with normal glucose tolerance (NGT); patients with IGT are followed up after 4 years for progression to DM. Analysis of baseline data reveals significant differences in gut microbiota and serum metabolites among the IGT, DM, and NGT groups. In addition, 13 types of gut microbiota and 17 types of circulating metabolites showed significant differences at baseline before IGT progressed to DM, including higher levels of Eggerthella unclassified, Coprobacillus unclassified, Clostridium ramosum, L-valine, L-norleucine, and L-isoleucine, and lower levels of Eubacterium eligens, Bacteroides faecis, Lachnospiraceae bacterium 3_1_46FAA, Alistipes senegalensis, Megaspaera elsdenii, Clostridium perfringens, α-linolenic acid, 10E,12Z-octadecadienoic acid, and dodecanoic acid. A random forest model based on differential intestinal microbiota and circulating metabolites can predict the progression from IGT to DM (AUC = 0.87). These results suggest that microbiome and metabolome dysbiosis occur in individuals with IGT and have important predictive values and potential for intervention in preventing IGT from progressing to DM.
Collapse
Affiliation(s)
- Boxun Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xuan Zhang
- Faculty of Biological Science and Technology, Baotou Teacher's College, Baotou, Inner Mongolia 014030, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhen Luo
- Infinitus (China) Company Ltd, Guangzhou, Guangdong 510405, China
| | - Jixiang Ren
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Xiaotong Yu
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Haiyan Zhao
- Xinjiekou Community Health Service Center in Xicheng District, Beijing 100035, China
| | - Yitian Wang
- Department of Spleen and Stomach, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518033, China
| | - Wenhui Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiwei Tian
- Xinjiekou Community Health Service Center in Xicheng District, Beijing 100035, China
| | - Xiuxiu Wei
- Beijing University of Chinese Medicine, Beijing 100105, China
| | - Qiyou Ding
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Haoyu Yang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zishan Jin
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Beijing University of Chinese Medicine, Beijing 100105, China
| | - Xiaolin Tong
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Northeast Asia Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China.
| | - Jun Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
30
|
Trichia E, Koulman A, Stewart ID, Brage S, Griffin SJ, Griffin JL, Khaw K, Langenberg C, Wareham NJ, Imamura F, Forouhi NG. Plasma Metabolites Related to the Consumption of Different Types of Dairy Products and Their Association with New-Onset Type 2 Diabetes: Analyses in the Fenland and EPIC-Norfolk Studies, United Kingdom. Mol Nutr Food Res 2024; 68:e2300154. [PMID: 38054622 PMCID: PMC10909549 DOI: 10.1002/mnfr.202300154] [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: 03/17/2023] [Revised: 07/07/2023] [Indexed: 12/07/2023]
Abstract
SCOPE To identify metabolites associated with habitual dairy consumption and investigate their associations with type 2 diabetes (T2D) risk. METHODS AND RESULTS Metabolomics assays were conducted in the Fenland (n = 10,281) and EPIC-Norfolk (n = 1,440) studies. Using 82 metabolites assessed in both studies, we developed metabolite scores to classify self-reported consumption of milk, yogurt, cheese, butter, and total dairy (Fenland Study-discovery set; n = 6035). Internal and external validity of the scores was evaluated (Fenland-validation set, n = 4246; EPIC-Norfolk, n = 1440). The study assessed associations between each metabolite score and T2D incidence in EPIC-Norfolk (n = 641 cases; 16,350 person-years). The scores classified low and high consumers for all dairy types with internal validity, and milk, butter, and total dairy with external validity. The scores were further associated with lower incident T2D: hazard ratios (95% confidence interval) per standard deviation: milk 0.71 (0.65, 0.77); butter 0.62 (0.57, 0.68); total dairy 0.66 (0.60, 0.72). These associations persisted after adjustment for known dairy-fat biomarkers. CONCLUSION Metabolite scores identified habitual consumers of milk, butter, and total dairy products, and were associated with lower T2D risk. These findings hold promise for identifying objective indicators of the physiological response to dairy consumption.
Collapse
Affiliation(s)
- Eirini Trichia
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Albert Koulman
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Isobel D. Stewart
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Soren Brage
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Simon J. Griffin
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | | | - Kay‐Tee Khaw
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Claudia Langenberg
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Nicholas J. Wareham
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Fumiaki Imamura
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Nita G. Forouhi
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| |
Collapse
|
31
|
Reghupaty SC, Dall NR, Svensson KJ. Hallmarks of the metabolic secretome. Trends Endocrinol Metab 2024; 35:49-61. [PMID: 37845120 PMCID: PMC10841501 DOI: 10.1016/j.tem.2023.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
The identification of novel secreted factors is advancing at an unprecedented pace. However, there is a critical need to consolidate and integrate this knowledge to provide a framework of their diverse mechanisms, functional significance, and inter-relationships. Complicating this effort are challenges related to nonstandardized methods, discrepancies in sample handling, and inconsistencies in the annotation of unknown molecules. This Review aims to synthesize the rapidly expanding field of the metabolic secretome, encompassing the five major types of secreted factors: proteins, peptides, metabolites, lipids, and extracellular vesicles. By systematically defining the functions and detection of the components within the metabolic secretome, this Review provides a primer into the advances of the field, and how integration of the techniques discussed can provide a deeper understanding of the mechanisms underlying metabolic homeostasis and its disorders.
Collapse
Affiliation(s)
- Saranya C Reghupaty
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA
| | - Nicholas R Dall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA.
| |
Collapse
|
32
|
Ratautė K, Ratautas D. A Review from a Clinical Perspective: Recent Advances in Biosensors for the Detection of L-Amino Acids. BIOSENSORS 2023; 14:5. [PMID: 38248382 PMCID: PMC10813600 DOI: 10.3390/bios14010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024]
Abstract
The field of biosensors is filled with reports and designs of various sensors, with the vast majority focusing on glucose sensing. However, in addition to glucose, there are many other important analytes that are worth investigating as well. In particular, L-amino acids appear as important diagnostic markers for a number of conditions. However, the progress in L-amino acid detection and the development of biosensors for L-amino acids are still somewhat insufficient. In recent years, the need to determine L-amino acids from clinical samples has risen. More clinical data appear to demonstrate that abnormal concentrations of L-amino acids are related to various clinical conditions such as inherited metabolic disorders, dyslipidemia, type 2 diabetes, muscle damage, etc. However, to this day, the diagnostic potential of L-amino acids is not yet fully established. Most likely, this is because of the difficulties in measuring L-amino acids, especially in human blood. In this review article, we extensively investigate the 'overlooked' L-amino acids. We review typical levels of amino acids present in human blood and broadly survey the importance of L-amino acids in most common conditions which can be monitored or diagnosed from changes in L-amino acids present in human blood. We also provide an overview of recent biosensors for L-amino acid monitoring and their advantages and disadvantages, with some other alternative methods for L-amino acid quantification, and finally we outline future perspectives related to the development of biosensing devices for L-amino acid monitoring.
Collapse
Affiliation(s)
- Kristina Ratautė
- Faculty of Medicine, Vilnius University, M. K. Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania
| | - Dalius Ratautas
- Life Science Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| |
Collapse
|
33
|
Sinturel F, Chera S, Brulhart-Meynet MC, Montoya JP, Stenvers DJ, Bisschop PH, Kalsbeek A, Guessous I, Jornayvaz FR, Philippe J, Brown SA, D'Angelo G, Riezman H, Dibner C. Circadian organization of lipid landscape is perturbed in type 2 diabetic patients. Cell Rep Med 2023; 4:101299. [PMID: 38016481 PMCID: PMC10772323 DOI: 10.1016/j.xcrm.2023.101299] [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: 12/20/2022] [Revised: 06/26/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
Lipid homeostasis in humans follows a diurnal pattern in muscle and pancreatic islets, altered upon metabolic dysregulation. We employ tandem and liquid-chromatography mass spectrometry to investigate daily regulation of lipid metabolism in subcutaneous white adipose tissue (SAT) and serum of type 2 diabetic (T2D) and non-diabetic (ND) human volunteers (n = 12). Around 8% of ≈440 lipid metabolites exhibit diurnal rhythmicity in serum and SAT from ND and T2D subjects. The spectrum of rhythmic lipids differs between ND and T2D individuals, with the most substantial changes observed early morning, as confirmed by lipidomics in an independent cohort of ND and T2D subjects (n = 32) conducted at a single morning time point. Strikingly, metabolites identified as daily rhythmic in both serum and SAT from T2D subjects exhibit phase differences. Our study reveals massive temporal and tissue-specific alterations of human lipid homeostasis in T2D, providing essential clues for the development of lipid biomarkers in a temporal manner.
Collapse
Affiliation(s)
- Flore Sinturel
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland
| | - Simona Chera
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland; Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Marie-Claude Brulhart-Meynet
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Jonathan Paz Montoya
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands; Laboratory for Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Arts and Sciences (KNAW), Amsterdam, 1105 BA, the Netherlands
| | - Idris Guessous
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - François R Jornayvaz
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Department of Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Jacques Philippe
- Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Department of Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Giovanni D'Angelo
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Charna Dibner
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland.
| |
Collapse
|
34
|
Yao Y, Lei X, Wang Y, Zhang G, Huang H, Zhao Y, Shi S, Gao Y, Cai X, Gao S, Lin Y. A Mitochondrial Nanoguard Modulates Redox Homeostasis and Bioenergy Metabolism in Diabetic Peripheral Neuropathy. ACS NANO 2023; 17:22334-22354. [PMID: 37782570 DOI: 10.1021/acsnano.3c04462] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
As a major late complication of diabetes, diabetic peripheral neuropathy (DPN) is the primary reason for amputation. Nevertheless, there are no wonder drugs available. Regulating dysfunctional mitochondria is a key therapeutic target for DPN. Resveratrol (RSV) is widely proven to guard mitochondria, yet the unsatisfactory bioavailability restricts its clinical application. Tetrahedral framework nucleic acids (tFNAs) are promising carriers due to their excellent cell entrance efficiency, biological safety, and structure editability. Here, RSV was intercalated into tFNAs to form the tFNAs-RSV complexes. tFNAs-RSV achieved enhanced stability, bioavailability, and biocompatibility compared with tFNAs and RSV alone. With its treatment, reactive oxygen species (ROS) production was minimized and reductases were activated in an in vitro model of DPN. Besides, respiratory function and adenosine triphosphate (ATP) production were enhanced. tFNAs-RSV also exhibited favorable therapeutic effects on sensory dysfunction, neurovascular deterioration, demyelination, and neuroapoptosis in DPN mice. Metabolomics analysis revealed that redox regulation and energy metabolism were two principal mechanisms that were impacted during the process. Comprehensive inspections indicated that tFNAs-RSV inhibited nitrosation and oxidation and activated reductase and respiratory chain. In sum, tFNAs-RSV served as a mitochondrial nanoguard (mito-guard), representing a viable drilling target for clinical drug development of DPN.
Collapse
Affiliation(s)
- Yangxue Yao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Xiaoyu Lei
- Research Center for Nano Biomaterials, and Analytical & Testing Center, Sichuan University, Chengdu 610064, P. R. China
| | - Yun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Geru Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Hongxiao Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Yuxuan Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Yang Gao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Shaojingya Gao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| |
Collapse
|
35
|
Li X, Chen H. Characteristics of glucolipid metabolism and complications in novel cluster-based diabetes subgroups: a retrospective study. Lipids Health Dis 2023; 22:200. [PMID: 37990237 PMCID: PMC10662503 DOI: 10.1186/s12944-023-01953-6] [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: 08/23/2023] [Accepted: 10/19/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Glucolipid metabolism plays an important role in the occurrence and development of diabetes mellitus. However, there is limited research on the characteristics of glucolipid metabolism and complications in different subgroups of newly diagnosed diabetes. This study aimed to investigate the characteristics of glucolipid metabolism and complications in novel cluster-based diabetes subgroups and explore the contributions of different glucolipid metabolism indicators to the occurrence of complications and pancreatic function. METHODS This retrospective study included 547 newly diagnosed type 2 diabetes patients. Age, body mass index (BMI), glycated hemoglobin (HbA1C), homeostasis model assessment-2 beta-cell function (HOMA2-β), and homeostasis model assessment-2 insulin resistance (HOMA2-IR) were used as clustering variables. The participants were divided into 4 groups by k-means cluster analysis. The characteristics of glucolipid indicators and complications in each subgroup were analyzed. Regression analyses were used to evaluate the impact of glucolipid metabolism indicators on complications and pancreatic function. RESULTS Total cholesterol (TC), triglycerides (TG), triglyceride glucose index (TyG), HbA1C, fasting plasma glucose (FPG), and 2-h postprandial plasma glucose (2hPG) were higher in the severe insulin-resistant diabetes (SIRD) and severe insulin-deficient diabetes (SIDD) groups. Fasting insulin (FINS), fasting C-peptide (FCP), 2-h postprandial insulin (2hINS), 2-h postprandial C-peptide (2hCP), and the monocyte-to-high-density lipoprotein cholesterol ratio (MHR) were higher in mild obesity-related diabetes (MOD) and SIRD. 2hCP, FCP, and FINS were positively correlated with HOMA2-β, while FPG, TyG, HbA1C, and TG were negatively correlated with HOMA2-β. FINS, FPG, FCP, and HbA1C were positively correlated with HOMA2-IR, while high-density lipoprotein (HDL) was negatively correlated with HOMA2-IR. FINS (odds ratio (OR),1.043;95% confidence interval (CI) 1.006 ~ 1.081), FCP (OR,2.881;95%CI 2.041 ~ 4.066), and TyG (OR,1.649;95%CI 1.292 ~ 2.104) contributed to increase the risk of nonalcoholic fatty liver disease (NAFLD); 2hINS (OR,1.015;95%CI 1.008 ~ 1.022) contributed to increase the risk of atherosclerotic cardiovascular disease (ASCVD); FCP (OR,1.297;95%CI 1.027 ~ 1.637) significantly increased the risk of chronic kidney disease (CKD). CONCLUSIONS There were differences in the characteristics of glucolipid metabolism as well as complications among different subgroups of newly diagnosed type 2 diabetes. 2hCP, FCP, FINS, FPG, TyG, HbA1C, HDL and TG influenced the function of insulin. FINS, TyG, 2hINS, and FCP were associated with ASCVD, NAFLD, and CKD in newly diagnosed T2DM patients.
Collapse
Affiliation(s)
- Xinrong Li
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu Province, China
| | - Hui Chen
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
36
|
Yao Y, Schneider A, Wolf K, Zhang S, Wang-Sattler R, Peters A, Breitner S. Longitudinal associations between metabolites and immediate, short- and medium-term exposure to ambient air pollution: Results from the KORA cohort study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:165780. [PMID: 37495154 DOI: 10.1016/j.scitotenv.2023.165780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/21/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Short-term exposure to air pollution has been reported to be associated with cardiopulmonary diseases, but the underlying mechanisms remain unclear. This study aimed to investigate changes in serum metabolites associated with immediate, short- and medium-term exposures to ambient air pollution. METHODS We used data from the German population-based Cooperative Health Research in the Region of Augsburg (KORA) S4 survey (1999-2001) and two follow-up examinations (F4: 2006-08 and FF4: 2013-14). Mass-spectrometry-based targeted metabolomics was used to quantify metabolites among serum samples. Only participants with repeated metabolites measurements were included in this analysis. We collected daily averages of fine particles (PM2.5), coarse particles (PMcoarse), nitrogen dioxide (NO2), and ozone (O3) at urban background monitors located in Augsburg, Germany. Covariate-adjusted generalized additive mixed-effects models were used to examine the associations between immediate (2-day average of same day and previous day as individual's blood withdrawal), short- (2-week moving average), and medium-term exposures (8-week moving average) to air pollution and metabolites. We further performed pathway analysis for the metabolites significantly associated with air pollutants in each exposure window. RESULTS Of 9,620 observations from 4,261 study participants, we included 5,772 (60.0%) observations from 2,583 (60.6%) participants in this analysis. Out of 108 metabolites that passed quality control, multiple significant associations between metabolites and air pollutants with several exposure windows were identified at a Bonferroni corrected p-value threshold (p < 3.9 × 10-5). We found the highest number of associations for NO2, particularly at the medium-term exposure windows. Among the identified metabolic pathways based on the metabolites significantly associated with air pollutants, the glycerophospholipid metabolism was the most robust pathway in different air pollutants exposures. CONCLUSIONS Our study suggested that short- and medium-term exposure to air pollution might induce alterations of serum metabolites, particularly in metabolites involved in metabolic pathways related to inflammatory response and oxidative stress.
Collapse
Affiliation(s)
- Yueli Yao
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany.
| | - Alexandra Schneider
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kathrin Wolf
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Siqi Zhang
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Rui Wang-Sattler
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany; German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich, Germany
| | - Susanne Breitner
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany
| |
Collapse
|
37
|
Singh S, Sarma DK, Verma V, Nagpal R, Kumar M. Unveiling the future of metabolic medicine: omics technologies driving personalized solutions for precision treatment of metabolic disorders. Biochem Biophys Res Commun 2023; 682:1-20. [PMID: 37788525 DOI: 10.1016/j.bbrc.2023.09.064] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023]
Abstract
Metabolic disorders are increasingly prevalent worldwide, leading to high rates of morbidity and mortality. The variety of metabolic illnesses can be addressed through personalized medicine. The goal of personalized medicine is to give doctors the ability to anticipate the best course of treatment for patients with metabolic problems. By analyzing a patient's metabolomic, proteomic, genetic profile, and clinical data, physicians can identify relevant diagnostic, and predictive biomarkers and develop treatment plans and therapy for acute and chronic metabolic diseases. To achieve this goal, real-time modeling of clinical data and multiple omics is essential to pinpoint underlying biological mechanisms, risk factors, and possibly useful data to promote early diagnosis and prevention of complex diseases. Incorporating cutting-edge technologies like artificial intelligence and machine learning is crucial for consolidating diverse forms of data, examining multiple variables, establishing databases of clinical indicators to aid decision-making, and formulating ethical protocols to address concerns. This review article aims to explore the potential of personalized medicine utilizing omics approaches for the treatment of metabolic disorders. It focuses on the recent advancements in genomics, epigenomics, proteomics, metabolomics, and nutrigenomics, emphasizing their role in revolutionizing personalized medicine.
Collapse
Affiliation(s)
- Samradhi Singh
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal, 462030, Madhya Pradesh, India
| | - Devojit Kumar Sarma
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal, 462030, Madhya Pradesh, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, 226014, Uttar Pradesh, India
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, Tallahassee, FL, 32306, USA
| | - Manoj Kumar
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal, 462030, Madhya Pradesh, India.
| |
Collapse
|
38
|
Najafi F, Mohseni P, Pasdar Y, Niknam M, Izadi N. The association between dietary amino acid profile and the risk of type 2 diabetes: Ravansar non-communicable disease cohort study. BMC Public Health 2023; 23:2284. [PMID: 37980456 PMCID: PMC10657569 DOI: 10.1186/s12889-023-17210-5] [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: 07/16/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is one of the most common chronic diseases and the main risk factors for T2D consist of a combination of lifestyle, unhealthy diet, and genetic factors. Amino acids are considered to be a major component of dietary sources for many of the associations between dietary protein and chronic disease. Therefore, this study amied to determine the association between dietary amino acid intakes and the incidence of T2D. METHODS The present nested case-control study was conducted using data from the Ravansar Non-Communicable Disease (RaNCD) Cohort Study. The information required for this study was collected from individuals who participated in the Adult Cohort Study from the start of the study until September 2023. Over a 6-year follow-up period, data from 113 new T2D cases were available. Four controls were then randomly selected for each case using density sampling. Cases and controls were matched for sex and age at the interview. Food frequency questionnaire (FFQ) was used to collect data related to all amino acids including tryptophan, threonine, isoleucine, leucine, lysine, methionine, cysteine, phenylalanine, tyrosine, valine, arginine, histidine, alanine, aspartic acid, glutamic acid, glycine, proline, and serine were also extracted. Binary logistic regression was used to estimate the crude and adjusted odds ratio for the risk of T2D. RESULTS Using the univariable model, a significant association was found between T2D risk and branched-chain, alkaline, sulfuric, and essential amino acids in the fourth quartile. Accordingly, individuals in the fourth quartile had a 1.81- to 1.87-fold higher risk of developing new T2D than individuals in the lowest quartile (P<0.05). After adjustment for several variables, the risk of developing a new T2D was 2.70 (95% CI: 1.16-6.31), 2.68 (95% CI: 1.16-6.21), 2.98 (95% CI: 1.27-6.96), 2.45 (95% CI: 1.02-5.90), and 2.66 (95% CI: 1.13-6.25) times higher, for individuals in the fourth quartile of branched-chain, alkaline, sulfuric, alcoholic, and essential amino acids compared with those in the lowest quartile, respectively. CONCLUSIONS The results showed that the risk of developing a new T2D was higher for individuals in the fourth quartile of branched-chain amino acids, alkaline, sulfate, and essential amino acids than in the lower quartile.
Collapse
Affiliation(s)
- Farid Najafi
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Mohseni
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yahya Pasdar
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahdieh Niknam
- Research Center for Social Determinants of Health, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Izadi
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
39
|
Yang W, Jiang W, Guo S. Regulation of Macronutrients in Insulin Resistance and Glucose Homeostasis during Type 2 Diabetes Mellitus. Nutrients 2023; 15:4671. [PMID: 37960324 PMCID: PMC10647592 DOI: 10.3390/nu15214671] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Insulin resistance is an important feature of metabolic syndrome and a precursor of type 2 diabetes mellitus (T2DM). Overnutrition-induced obesity is a major risk factor for the development of insulin resistance and T2DM. The intake of macronutrients plays a key role in maintaining energy balance. The components of macronutrients distinctly regulate insulin sensitivity and glucose homeostasis. Precisely adjusting the beneficial food compound intake is important for the prevention of insulin resistance and T2DM. Here, we reviewed the effects of different components of macronutrients on insulin sensitivity and their underlying mechanisms, including fructose, dietary fiber, saturated and unsaturated fatty acids, and amino acids. Understanding the diet-gene interaction will help us to better uncover the molecular mechanisms of T2DM and promote the application of precision nutrition in practice by integrating multi-omics analysis.
Collapse
Affiliation(s)
| | | | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA; (W.Y.); (W.J.)
| |
Collapse
|
40
|
Liu Y, Wang D, Liu YP. Metabolite profiles of diabetes mellitus and response to intervention in anti-hyperglycemic drugs. Front Endocrinol (Lausanne) 2023; 14:1237934. [PMID: 38027178 PMCID: PMC10644798 DOI: 10.3389/fendo.2023.1237934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) has become a major health problem, threatening the quality of life of nearly 500 million patients worldwide. As a typical multifactorial metabolic disease, T2DM involves the changes and interactions of various metabolic pathways such as carbohydrates, amino acid, and lipids. It has been suggested that metabolites are not only the endpoints of upstream biochemical processes, but also play a critical role as regulators of disease progression. For example, excess free fatty acids can lead to reduced glucose utilization in skeletal muscle and induce insulin resistance; metabolism disorder of branched-chain amino acids contributes to the accumulation of toxic metabolic intermediates, and promotes the dysfunction of β-cell mitochondria, stress signal transduction, and apoptosis. In this paper, we discuss the role of metabolites in the pathogenesis of T2DM and their potential as biomarkers. Finally, we list the effects of anti-hyperglycemic drugs on serum/plasma metabolic profiles.
Collapse
Affiliation(s)
| | | | - Yi-Ping Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
41
|
Makki BE, Rahman S. Alzheimer's Disease in Diabetic Patients: A Lipidomic Prospect. Neuroscience 2023; 530:79-94. [PMID: 37652288 DOI: 10.1016/j.neuroscience.2023.08.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/04/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023]
Abstract
Diabetes Mellitus (DM) and Alzheimer's disease (AD) have been two of the most common chronic diseases affecting people worldwide. Type 2 DM (T2DM) is a metabolic disease depicted by insulin resistance, dyslipidemia, and chronic hyperglycemia while AD is a neurodegenerative disease marked by Amyloid β (Aβ) accumulation, neurofibrillary tangles aggregation, and tau phosphorylation. Various clinical, epidemiological, and lipidomics studies have linked those diseases claiming shared pathological pathways raising the assumption that diabetic patients are at an increased risk of developing AD later in their lives. Insulin resistance is the tipping point beyond where advanced glycation end (AGE) products and free radicals are produced leading to oxidative stress and lipid peroxidation. Additionally, different types of lipids are playing a crucial role in the development and the relationship between those diseases. Lipidomics, an analysis of lipid structure, formation, and interactions, evidently exhibits these lipid changes and their direct and indirect effect on Aβ synthesis, insulin resistance, oxidative stress, and neuroinflammation. In this review, we have discussed the pathophysiology of T2DM and AD, the interconnecting pathological pathways they share, and the lipidomics where different lipids such as cholesterol, phospholipids, sphingolipids, and sulfolipids contribute to the underlying features of both diseases. Understanding their role can be beneficial for diagnostic purposes or introducing new drugs to counter AD.
Collapse
Affiliation(s)
| | - Sarah Rahman
- School of Medicine, Tehran University of Medical Sciences, Iran
| |
Collapse
|
42
|
Wei P, He M, Wang Y, Han G. High-Fat Diet Alters Acylcarnitine Metabolism of the Retina and Retinal Pigment Epithelium/Choroidal Tissues in Laser-Induced Choroidal Neovascularization Rat Models. Mol Nutr Food Res 2023; 67:e2300080. [PMID: 37490551 DOI: 10.1002/mnfr.202300080] [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: 02/11/2023] [Revised: 06/19/2023] [Indexed: 07/27/2023]
Abstract
SCOPE Choroidal neovascularization (CNV) is age-related macular degeneration's (AMD) main pathological change. High-fat diet (HFD) is associated with a form of CNV; however, the specific mechanism is unclear. Mitochondrial dysfunction, characterized by abnormal acylcarnitine, occurs during metabolic screening of serum or other body tissues in AMD. This study investigates HFD's role in retinal and retinal pigment epithelium (RPE)/choroidal acylcarnitine metabolism in CNV formation. METHODS AND RESULTS Chow diet and HFD-BN rats are laser-treated to induce CNV. Acylcarnitine species are quantitatively characterized by ultrahigh-performance liquid chromatography-tandem mass spectrometry. Optical coherence tomography and fundus fluorescein angiography evaluate CNV severity. HFD promotes weight gain, dyslipidemia, and CNV formation. In CNV rats, few medium-chain fatty acids (MCFAs) acylcarnitine in the RPE/choroid are initially affected. When an HFD is administered to these, even MCFA acylcarnitine in the RPE/choroid is found to decline. However, in the retina, odd acylcarnitines are increased, revealing "an opposite" change within the RPE/choroid, accompanied by influencing glycolytic key enzymes. The HFD+CNV group incorporated fewer long-chain acylcarnitines, like C18:2, into the retina than controls. CONCLUSIONS HFD hastens choroidal neovascularization. The study comprehensively documented acylcarnitine profiles in a CNV rat model. Acylcarnitine's odd-even and carbon-chain length properties may guide future therapeutics.
Collapse
Affiliation(s)
- Pinghui Wei
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, 300020, P. R. China
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, 300020, P. R. China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, 300020, P. R. China
| | - Meiqin He
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300020, P. R. China
| | - Ying Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, 300020, P. R. China
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, 300020, P. R. China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, 300020, P. R. China
| | - Guoge Han
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, 300020, P. R. China
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin, 300020, P. R. China
- Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, 300020, P. R. China
| |
Collapse
|
43
|
Liu Z, Wang Q, Ma A, Feng S, Chung D, Zhao J, Ma Q, Liu B. Inference of disease-associated microbial gene modules based on metagenomic and metatranscriptomic data. Comput Biol Med 2023; 165:107458. [PMID: 37703713 DOI: 10.1016/j.compbiomed.2023.107458] [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: 07/10/2023] [Revised: 08/22/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023]
Abstract
The identification of microbial characteristics associated with diseases is crucial for disease diagnosis and therapy. However, the presence of heterogeneity, high dimensionality, and large amounts of microbial data presents tremendous challenges in discovering key microbial features. In this paper, we present IDAM, a novel computational method for inferring disease-associated gene modules from metagenomic and metatranscriptomic data. This method integrates gene context conservation (uber-operons) and regulatory mechanisms (gene co-expression patterns) within a mathematical graph model to explore gene modules associated with specific diseases. It alleviates reliance on prior meta-data. We applied IDAM to publicly available datasets from inflammatory bowel disease, melanoma, type 1 diabetes mellitus, and irritable bowel syndrome. The results demonstrated the superior performance of IDAM in inferring disease-associated characteristics compared to existing popular tools. Furthermore, we showcased the high reproducibility of the gene modules inferred by IDAM using independent cohorts with inflammatory bowel disease. We believe that IDAM can be a highly advantageous method for exploring disease-associated microbial characteristics. The source code of IDAM is freely available at https://github.com/OSU-BMBL/IDAM, and the web server can be accessed at https://bmblx.bmi.osumc.edu/idam/.
Collapse
Affiliation(s)
- Zhaoqian Liu
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China
| | - Qi Wang
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Shaohong Feng
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, 43210, USA
| | - Jing Zhao
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, 43210, USA.
| | - Bingqiang Liu
- School of Mathematics, Shandong University, Jinan, Shandong, 250100, China; Shandong National Center for Applied Mathematics, Jinan, Shandong, 250100, China.
| |
Collapse
|
44
|
Sengupta A, Ghosh S, Sharma S, Sonawat HM. Early Perturbations in Red Blood Cells in Response to Murine Malarial Parasite Infection: Proof-of-Concept 1H NMR Metabolomic Study. Life (Basel) 2023; 13:1684. [PMID: 37629541 PMCID: PMC10455252 DOI: 10.3390/life13081684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The major focus of metabolomics research has been confined to the readily available biofluids-urine and blood serum. However, red blood cells (RBCs) are also readily available, and may be a source of a wealth of information on vertebrates. However, the comprehensive metabolomic characterization of RBCs is minimal although they exhibit perturbations in various physiological states. RBCs act as the host of malarial parasites during the symptomatic stage. Thus, understanding the changes in RBC metabolism during infection is crucial for a better understanding of disease progression. METHODS The metabolome of normal RBCs obtained from Swiss mice was investigated using 1H NMR spectroscopy. Several 1 and 2-dimensional 1H NMR experiments were employed for this purpose. The information from this study was used to investigate the changes in the RBC metabolome during the early stage of infection (~1% infected RBCs) by Plasmodium bergheii ANKA. RESULTS We identified over 40 metabolites in RBCs. Several of these metabolites were quantitated using 1H NMR spectroscopy. The results indicate changes in the choline/membrane components and other metabolites during the early stage of malaria. CONCLUSIONS The paper reports the comprehensive characterization of the metabolome of mouse RBCs. Changes during the early stage of malarial infection suggest significant metabolic alteration, even at low parasite content (~1%). GENERAL SIGNIFICANCE This study should be of use in maximizing the amount of information available from metabolomic experiments on the cellular components of blood. The technique can be directly applied to real-time investigation of infectious diseases that target RBCs.
Collapse
Affiliation(s)
- Arjun Sengupta
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai 400005, India; (S.G.); (H.M.S.)
| | - Soumita Ghosh
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai 400005, India; (S.G.); (H.M.S.)
| | - Shobhona Sharma
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai 400005, India;
| | - Haripalsingh M. Sonawat
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai 400005, India; (S.G.); (H.M.S.)
| |
Collapse
|
45
|
Villasanta-Gonzalez A, Mora-Ortiz M, Alcala-Diaz JF, Rivas-Garcia L, Torres-Peña JD, Lopez-Bascon A, Calderon-Santiago M, Arenas-Larriva AP, Priego-Capote F, Malagon MM, Eichelmann F, Perez-Martinez P, Delgado-Lista J, Schulze MB, Camargo A, Lopez-Miranda J. Plasma lipidic fingerprint associated with type 2 diabetes in patients with coronary heart disease: CORDIOPREV study. Cardiovasc Diabetol 2023; 22:199. [PMID: 37537576 PMCID: PMC10401778 DOI: 10.1186/s12933-023-01933-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/21/2023] [Indexed: 08/05/2023] Open
Abstract
OBJECTIVE We aimed to identify a lipidic profile associated with type 2 diabetes mellitus (T2DM) development in coronary heart disease (CHD) patients, to provide a new, highly sensitive model which could be used in clinical practice to identify patients at T2DM risk. METHODS This study considered the 462 patients of the CORDIOPREV study (CHD patients) who were not diabetic at the beginning of the intervention. In total, 107 of them developed T2DM after a median follow-up of 60 months. They were diagnosed using the American Diabetes Association criteria. A novel lipidomic methodology employing liquid chromatography (LC) separation followed by HESI, and detection by mass spectrometry (MS) was used to annotate the lipids at the isomer level. The patients were then classified into a Training and a Validation Set (60-40). Next, a Random Survival Forest (RSF) was carried out to detect the lipidic isomers with the lowest prediction error, these lipids were then used to build a Lipidomic Risk (LR) score which was evaluated through a Cox. Finally, a production model combining the clinical variables of interest, and the lipidic species was carried out. RESULTS LC-tandem MS annotated 440 lipid species. From those, the RSF identified 15 lipid species with the lowest prediction error. These lipids were combined in an LR score which showed association with the development of T2DM. The LR hazard ratio per unit standard deviation was 2.87 and 1.43, in the Training and Validation Set respectively. Likewise, patients with higher LR Score values had lower insulin sensitivity (P = 0.006) and higher liver insulin resistance (P = 0.005). The receiver operating characteristic (ROC) curve obtained by combining clinical variables and the selected lipidic isomers using a generalised lineal model had an area under the curve (AUC) of 81.3%. CONCLUSION Our study showed the potential of comprehensive lipidomic analysis in identifying patients at risk of developing T2DM. In addition, the lipid species combined with clinical variables provided a new, highly sensitive model which can be used in clinical practice to identify patients at T2DM risk. Moreover, these results also indicate that we need to look closely at isomers to understand the role of this specific compound in T2DM development. Trials registration NCT00924937.
Collapse
Affiliation(s)
- Alejandro Villasanta-Gonzalez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - Marina Mora-Ortiz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Lorenzo Rivas-Garcia
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Asuncion Lopez-Bascon
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Monica Calderon-Santiago
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio P Arenas-Larriva
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - Feliciano Priego-Capote
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria M Malagon
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
| | - Fabian Eichelmann
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Matthias B Schulze
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- Germany Institute of Nutrition Science, University of Potsdam, Nuthetal, Germany
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain.
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain.
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain.
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain.
- Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
46
|
Chen Y, Xu W, Zhang W, Tong R, Yuan A, Li Z, Jiang H, Hu L, Huang L, Xu Y, Zhang Z, Sun M, Yan X, Chen AF, Qian K, Pu J. Plasma metabolic fingerprints for large-scale screening and personalized risk stratification of metabolic syndrome. Cell Rep Med 2023; 4:101109. [PMID: 37467725 PMCID: PMC10394172 DOI: 10.1016/j.xcrm.2023.101109] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/01/2023] [Accepted: 06/16/2023] [Indexed: 07/21/2023]
Abstract
Direct diagnosis and accurate assessment of metabolic syndrome (MetS) allow for prompt clinical interventions. However, traditional diagnostic strategies overlook the complex heterogeneity of MetS. Here, we perform metabolomic analysis in 13,554 participants from the natural cohort and identify 26 hub plasma metabolic fingerprints (PMFs) associated with MetS and its early identification (pre-MetS). By leveraging machine-learning algorithms, we develop robust diagnostic models for pre-MetS and MetS with convincing performance through independent validation. We utilize these PMFs to assess the relative contributions of the four major MetS risk factors in the general population, ranked as follows: hyperglycemia, hypertension, dyslipidemia, and obesity. Furthermore, we devise a personalized three-dimensional plasma metabolic risk (PMR) stratification, revealing three distinct risk patterns. In summary, our study offers effective screening tools for identifying pre-MetS and MetS patients in the general community, while defining the heterogeneous risk stratification of metabolic phenotypes in real-world settings.
Collapse
Affiliation(s)
- Yifan Chen
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Wei Xu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Wei Zhang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Renyang Tong
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Ancai Yuan
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Zheng Li
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Huiru Jiang
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Liuhua Hu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Lin Huang
- Country Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yudian Xu
- School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ziyue Zhang
- School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Mingze Sun
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Kun Qian
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China; School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Jun Pu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China.
| |
Collapse
|
47
|
Okekunle AP, Lee H, Provido SMP, Chung GH, Hong S, Yu SH, Lee CB, Lee JE. Dietary intakes of branched-chain amino acids and plasma lipid profiles among filipino women in Korea: the Filipino Women's Diet and Health Study (FiLWHEL). Nutr J 2023; 22:34. [PMID: 37430285 DOI: 10.1186/s12937-023-00861-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/24/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND The potential role of dietary branched-chain amino acids (BCAA) in metabolic health, including cardiovascular disease and diabetes, is evolving, and it is yet to be understood if dietary BCAA intakes are associated with plasma lipid profiles or dyslipidaemia. This study tested the association of dietary BCAA intakes with plasma lipid profiles and dyslipidaemia among Filipino women in Korea. METHODS Energy-adjusted dietary BCAA intakes (isoleucine, leucine, valine, and total BCAA) and fasting blood profiles of triglycerides (TG), total cholesterol (TC), high-density lipoprotein-cholesterol (HDL-C), and low-density lipoprotein-cholesterol (LDL-C) were determined in a sample of 423 women enrolled in the Filipino Women's Diet and Health Study (FiLWHEL). The generalized linear model was applied to estimate least-square (LS) means and 95% confidence intervals (CIs) and compare plasma TG, TC, HDL-C, and LDL-C across tertile distribution of energy-adjusted dietary BCAA intakes at P < 0.05. RESULTS Mean of energy-adjusted dietary total BCAA intake was 8.3 ± 3.9 g/d. Average plasma lipid profiles were 88.5 ± 47.4 mg/dl for TG, 179.7 ± 34.5 mg/dl for TC, 58.0 ± 13.7 mg/dl for HDL-C, and 104.0 ± 30.5 mg/dl for LDL-C. LS means, and 95% CIs across tertiles of energy-adjusted total BCAA intakes were 89.9 mg/dl, 88.8 mg/dl and 85.8 mg/dl (P-trend = 0.45) for TG, 179.1 mg/dl, 183.6 mg/dl and 176.5 mg/dl (P-trend = 0.48) for TC, 57.5 mg/dl, 59.6 mg/dl and 57.1 mg/dl (P-trend = 0.75) for HDL-C and 103.6 mg/dl, 106.2 mg/dl and 102.3 mg/dl (P-trend = 0.68) for LDL-C. Furthermore, the multivariable-adjusted prevalence ratios and 95% confidence intervals for dyslipidaemia across increasing tertile distribution of energy-adjusted total BCAA intake were; 1.00, 0.67 (0.40, 1.13) and 0.45 (0.16, 1.27; P-trend = 0.03) for the first, second and third tertile, respectively. CONCLUSIONS Higher dietary intakes of BCAA presented a statistically significant inverse trend with the prevalence of dyslipidaemia among Filipino women in this study and testing these associations in longitudinal studies may be necessary to confirm these findings.
Collapse
Grants
- 2020H1D3A1A04081265 National Research Foundation of Korea
- 2020H1D3A1A04081265 National Research Foundation of Korea
- 0448A-2021077 Seoul National University Asia Center
- 201300000001270 Hanmi Pharmaceutical Co., Ltd. Korea
- 201300000001270 Hanmi Pharmaceutical Co., Ltd. Korea
- 201300000001270 Hanmi Pharmaceutical Co., Ltd. Korea
- 201300000001270 Hanmi Pharmaceutical Co., Ltd. Korea
- 201600000000225 Chong Kun Dang Pharm., Seoul, Korea
- 201600000000225 Chong Kun Dang Pharm., Seoul, Korea
- 201600000000225 Chong Kun Dang Pharm., Seoul, Korea
- 201600000000225 Chong Kun Dang Pharm., Seoul, Korea
Collapse
Affiliation(s)
- Akinkunmi Paul Okekunle
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak- ro, Gwanak-gu, Seoul, 08826, Korea
- Research Institute of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Heejin Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak- ro, Gwanak-gu, Seoul, 08826, Korea
| | - Sherlyn Mae P Provido
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak- ro, Gwanak-gu, Seoul, 08826, Korea
| | - Grace H Chung
- Department of Child Development & Family Studies, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Sangmo Hong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153 Gyeongchun-ro, Guri, 11923, Korea
| | - Sung Hoon Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153 Gyeongchun-ro, Guri, 11923, Korea
| | - Chang Beom Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153 Gyeongchun-ro, Guri, 11923, Korea
| | - Jung Eun Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak- ro, Gwanak-gu, Seoul, 08826, Korea.
- Research Institute of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.
| |
Collapse
|
48
|
Naudin S, Sampson JN, Moore SC, Albanes D, Freedman ND, Weinstein SJ, Stolzenberg-Solomon R. Lipidomics and pancreatic cancer risk in two prospective studies. Eur J Epidemiol 2023; 38:783-793. [PMID: 37169992 PMCID: PMC11152614 DOI: 10.1007/s10654-023-01014-3] [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: 01/07/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Pancreatic ductal carcinoma (PDAC) is highly fatal with limited understanding of mechanisms underlying its carcinogenesis. We comprehensively investigated whether lipidomic measures were associated with PDAC in two prospective studies. We measured 904 lipid species and 252 fatty acids across 15 lipid classes in pre-diagnostic serum (up to 24 years) in a PDAC nested-case control study within the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO, NCT00002540) with 332 matched case-control sets including 272 having serial blood samples and Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study (ATBC, NCT00342992) with 374 matched case-control sets. Controls were matched to cases by cohort, age, sex, race, and date at blood draw. We used conditional logistic regression to calculate odds ratios (OR) and 95% confidence intervals (CI) per one-standard deviation increase in log-lipid concentrations within each cohort, and combined ORs using fixed-effects meta-analyses. Forty-three lipid species were associated with PDAC (false discovery rate, FDR ≤ 0.10), including lysophosphatidylcholines (LPC, n = 2), phosphatidylethanolamines (PE, n = 17), triacylglycerols (n = 13), phosphatidylcholines (PC, n = 3), diacylglycerols (n = 4), monoacylglycerols (MAG, n = 2), cholesteryl esters (CE, n = 1), and sphingomyelins (n = 1). LPC(18:2) and PE(O-16:0/18:2) showed significant inverse associations with PDAC at the Bonferroni threshold (P value < 5.5 × 10-5). The fatty acids LPC[18:2], LPC[16:0], PC[15:0], MAG[18:1] and CE[22:0] were significantly associated with PDAC (FDR < 0.10). Similar associations were observed in both cohorts. There was no significant association for the differences between PLCO serial lipidomic measures or heterogeneity by follow-up time overall. Results support that the pre-diagnostic serum lipidome, including 43 lipid species from 8 lipid classes and 5 fatty acids, is associated with PDAC.
Collapse
Affiliation(s)
- Sabine Naudin
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, DHHS, 9609 Medical Center Drive, NCI Shady Grove, Room 6E420, Rockville, MD, 20850, USA
| | - Joshua N Sampson
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Rockville, MD, USA
| | - Steven C Moore
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, DHHS, 9609 Medical Center Drive, NCI Shady Grove, Room 6E420, Rockville, MD, 20850, USA
| | - Demetrius Albanes
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, DHHS, 9609 Medical Center Drive, NCI Shady Grove, Room 6E420, Rockville, MD, 20850, USA
| | - Neal D Freedman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, DHHS, 9609 Medical Center Drive, NCI Shady Grove, Room 6E420, Rockville, MD, 20850, USA
| | - Stephanie J Weinstein
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, DHHS, 9609 Medical Center Drive, NCI Shady Grove, Room 6E420, Rockville, MD, 20850, USA
| | - Rachael Stolzenberg-Solomon
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, DHHS, 9609 Medical Center Drive, NCI Shady Grove, Room 6E420, Rockville, MD, 20850, USA.
| |
Collapse
|
49
|
Martín-Masot R, Jiménez-Muñoz M, Herrador-López M, Navas-López VM, Obis E, Jové M, Pamplona R, Nestares T. Metabolomic Profiling in Children with Celiac Disease: Beyond the Gluten-Free Diet. Nutrients 2023; 15:2871. [PMID: 37447198 DOI: 10.3390/nu15132871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Celiac disease (CD) is included in the group of complex or multifactorial diseases, i.e., those caused by the interaction of genetic and environmental factors. Despite a growing understanding of the pathophysiological mechanisms of the disease, diagnosis is still often delayed and there are no effective biomarkers for early diagnosis. The only current treatment, a gluten-free diet (GFD), can alleviate symptoms and restore intestinal villi, but its cellular effects remain poorly understood. To gain a comprehensive understanding of CD's progression, it is crucial to advance knowledge across various scientific disciplines and explore what transpires after disease onset. Metabolomics studies hold particular significance in unravelling the complexities of multifactorial and multisystemic disorders, where environmental factors play a significant role in disease manifestation and progression. By analyzing metabolites, we can gain insights into the reasons behind CD's occurrence, as well as better comprehend the impact of treatment initiation on patients. In this review, we present a collection of articles that showcase the latest breakthroughs in the field of metabolomics in pediatric CD, with the aim of trying to identify CD biomarkers for both early diagnosis and treatment monitoring. These advancements shed light on the potential of metabolomic analysis in enhancing our understanding of the disease and improving diagnostic and therapeutic strategies. More studies need to be designed to cover metabolic profiles in subjects at risk of developing the disease, as well as those analyzing biomarkers for follow-up treatment with a GFD.
Collapse
Affiliation(s)
- Rafael Martín-Masot
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
- Institute of Nutrition and Food Technology "José MataixVerdú" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18071 Granada, Spain
| | - María Jiménez-Muñoz
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Marta Herrador-López
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Víctor Manuel Navas-López
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Elia Obis
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Teresa Nestares
- Institute of Nutrition and Food Technology "José MataixVerdú" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18071 Granada, Spain
- Department of Physiology, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
| |
Collapse
|
50
|
Li N, Li J, Wang H, Liu J, Li W, Yang K, Huo X, Leng J, Yu Z, Hu G, Fang Z, Yang X. Aromatic Amino Acids and Their Interactions with Gut Microbiota-Related Metabolites for Risk of Gestational Diabetes: A Prospective Nested Case-Control Study in a Chinese Cohort. ANNALS OF NUTRITION & METABOLISM 2023; 79:291-300. [PMID: 37339616 DOI: 10.1159/000531481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION The aim of this study was to explore associations of aromatic amino acids (AAA) in early pregnancy with gestational diabetes mellitus (GDM), and whether high AAA and gut microbiota-related metabolites had interactive effects on GDM risk. METHODS We conducted a 1:1 case-control study (n = 486) nested in a prospective cohort of pregnant women from 2010 to 2012. According to the International Association of Diabetes and Pregnancy Study Group's criteria, 243 women were diagnosed with GDM. Binary conditional logistic regression was performed to examine associations of AAA with GDM risk. Interactions between AAA and gut microbiota-related metabolites for GDM were examined using additive interaction measures. RESULTS High phenylalanine and tryptophan were associated with increased GDM risk (OR: 1.72, 95% CI: 1.07-2.78 and 1.66, 1.02-2.71). The presence of high trimethylamine (TMA) markedly increased the OR of high phenylalanine alone up to 7.95 (2.79-22.71), while the presence of low glycoursodeoxycholic acid (GUDCA) markedly increased the OR of high tryptophan alone up to 22.88 (5.28-99.26), both with significant additive interactions. Furthermore, high lysophosphatidylcholines (LPC18:0) mediated both interactive effects. CONCLUSIONS High phenylalanine may have an additive interaction with high TMA, while high tryptophan may have an additive interaction with low GUDCA toward increased risk of GDM, both being mediated via LPC18:0.
Collapse
Affiliation(s)
- Ninghua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jinnan Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Weiqin Li
- Project Office, Tianjin Women and Children's Health Center, Tianjin, China
| | - Kai Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaoxu Huo
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Junhong Leng
- Project Office, Tianjin Women and Children's Health Center, Tianjin, China
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Zhongze Fang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|