1
|
Yin X, Li J, Bose D, Okamoto J, Kwon A, Jackson AU, Fernandes Silva L, Oravilahti A, Chu X, Stringham HM, Liu L, Peng R, Xia Z, Ripatti S, Daly M, Palotie A, Scott LJ, Burant CF, Fauman EB, Wen X, Boehnke M, Laakso M, Morrison J. Assessing the potential causal effects of 1099 plasma metabolites on 2099 binary disease endpoints. Nat Commun 2025; 16:3039. [PMID: 40155430 PMCID: PMC11953310 DOI: 10.1038/s41467-025-58129-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
Metabolites are small molecules that are useful for estimating disease risk and elucidating disease biology. Here, we perform two-sample Mendelian randomization to systematically infer the potential causal effects of 1099 plasma metabolites measured in 6136 Finnish men from the METSIM study on risk of 2099 binary disease endpoints measured in 309,154 Finnish individuals from FinnGen. We find evidence for 282 putative causal effects of 70 metabolites on 183 disease endpoints. We also identify 25 metabolites with potential causal effects across multiple disease domains, including ascorbic acid 2-sulfate affecting 26 disease endpoints in 12 disease domains. Our study suggests that N-acetyl-2-aminooctanoate and glycocholenate sulfate affect risk of atrial fibrillation through two distinct metabolic pathways and that N-methylpipecolate may mediate the putative causal effect of N6,N6-dimethyllysine on anxious personality disorder.
Collapse
Affiliation(s)
- Xianyong Yin
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jack Li
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Debraj Bose
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jeffrey Okamoto
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Annie Kwon
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Anniina Oravilahti
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Xiaomeng Chu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Lei Liu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruyi Peng
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhijie Xia
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Mark Daly
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology, and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology, and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Xiaoquan Wen
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland.
| | - Jean Morrison
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Zhang Y, Spitzer BW, Zhang Y, Wallace DA, Yu B, Qi Q, Argos M, Avilés-Santa ML, Boerwinkle E, Daviglus ML, Kaplan R, Cai J, Redline S, Sofer T. Untargeted metabolome atlas for sleep-related phenotypes in the Hispanic community health study/study of Latinos. EBioMedicine 2025; 111:105507. [PMID: 39693737 PMCID: PMC11722176 DOI: 10.1016/j.ebiom.2024.105507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Sleep is essential to maintaining health and wellbeing of individuals, influencing a variety of outcomes from mental health to cardiometabolic disease. This study aims to assess the relationships between various sleep-related phenotypes and blood metabolites. METHODS Utilising data from the Hispanic Community Health Study/Study of Latinos, we performed association analyses between 40 sleep-related phenotypes, grouped in several domains (sleep disordered breathing (SDB), sleep duration, sleep timing, self-reported insomnia symptoms, excessive daytime sleepiness (EDS), and heart rate during sleep), and 768 metabolites measured via untargeted metabolomics profiling. Network analysis was employed to visualise and interpret the associations between sleep phenotypes and metabolites. FINDINGS The patterns of statistically significant associations between sleep phenotypes and metabolites differed by superpathways, and highlighted subpathways of interest for future studies. For example, primary bile acid metabolism showed the highest cumulative percentage of statistically significant associations across all sleep phenotype domains except for SDB and EDS phenotypes. Several metabolites were associated with multiple sleep phenotypes, from a few domains. Glycochenodeoxycholate, vanillyl mandelate (VMA) and 1-stearoyl-2-oleoyl-GPE (18:0/18:1) were associated with the highest number of sleep phenotypes, while pregnenolone sulfate was associated with all sleep phenotype domains except for sleep duration. N-lactoyl amino acids such as N-lactoyl phenylalanine (lac-Phe), were associated with sleep duration, SDB, sleep timing and heart rate during sleep. INTERPRETATION This atlas of sleep-metabolite associations will facilitate hypothesis generation and further study of the metabolic underpinnings of sleep health. FUNDING R01HL161012, R35HL135818, R01AG80598.
Collapse
Affiliation(s)
- Ying Zhang
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian W Spitzer
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yu Zhang
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Danielle A Wallace
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Argos
- Department of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, IL, USA; Department of Environmental Health, School of Public Health, Boston University, Boston, MA, USA
| | - M Larissa Avilés-Santa
- Division of Clinical and Health Services Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Eric Boerwinkle
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA; Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Susan Redline
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Cao H, Tian Q, Chu L, Wu L, Gao H, Gao Q. Lycium ruthenicum Murray anthocyanin-driven neuroprotection modulates the gut microbiome and metabolome of MPTP-treated mice. Food Funct 2024; 15:12210-12227. [PMID: 39601125 DOI: 10.1039/d4fo01878h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Emerging evidence suggests that Parkinson's disease (PD) is strongly associated with altered gut microbiota. The present study investigated the prophylactic effects of anthocyanins (ACNs) from Lycium ruthenicum Murray on Parkinson's disease based on microbiomics and metabolomics. In this study, sixty-six adult male C57BL/6J mice were randomized into the control group, model group, positive drug (Madopar) group, and low-, medium- and high-dose ACN groups. Behavioral experiments were conducted and pathological indicators were determined. Fresh feces were collected for microbiomic analysis using 16S rRNA sequencing. Urine and serum were analyzed by the UPLC-MS method for untargeted metabolomics. The results demonstrated that ACNs ameliorated 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced motor deficits, dopamine neuron death, and glial cell activation, while 100 mg kg-1 and 200 mg kg-1 ACNs were more neuroprotective than 50 mg kg-1. Mice with PD-like phenotypes have an altered gut microbiota composition, and ACNs may regulate this disorder by causing an increase in Firmicutes/Bacteroidota ratio and abundance of norank_f__Eubacterium_coprostanoligenes_group and a decrease in the abundance of norank_f__Muribaculaceae, Coriobacteriaceae_UCG-002 and Parvibacter. Furthermore, ACNs increased 14 urinary key metabolites such as DIMBOA-Glc and tauroursodeoxycholic acid, decreased N,N-dimethyllysine, and increased 12 serum key metabolites such as 1-methylguanine and 1-nitro-5-glutathionyl-6-hydroxy-5,6-dihydronaphthalene, and decreased lamivudine-monophosphate and 5-butyl-2- methylpyridine. The present study reveals that ACNs are protective against MPTP-induced PD in mice by modulating anti-inflammatory flora in the gut and endogenous metabolites in serum/urine, and the key mechanisms may be related to Coriobacteriaceae_UCG-002 and glycerophospholipid metabolic pathways. Our findings provide new insights into the pathogenesis and potential treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Hongdou Cao
- School of Public Health, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Qi Tian
- School of Public Health, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Liwen Chu
- School of Public Health, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Lingyu Wu
- School of Public Health, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Hua Gao
- Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia 750000, China.
| | - Qinghan Gao
- School of Public Health, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| |
Collapse
|
4
|
Cao D, Zhang Y, Zhang S, Li J, Yang Q, Wang P. Risk of Alzheimer's disease and genetically predicted levels of 1400 plasma metabolites: a Mendelian randomization study. Sci Rep 2024; 14:26078. [PMID: 39478193 PMCID: PMC11525545 DOI: 10.1038/s41598-024-77921-6] [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: 03/29/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024] Open
Abstract
Alzheimer's disease (AD) is a metabolic disorder. Discovering the metabolic products involved in the development of AD may help not only in the early detection and prevention of AD but also in understanding its pathogenesis and treatment. This study investigated the causal association between the latest large-scale plasma metabolites (1091 metabolites and 309 metabolite ratios) and AD. Through the application of Mendelian randomization analysis methods such as inverse-variance weighted (IVW), MR-Egger, and weighted median models, 66 metabolites and metabolite ratios were identified as potentially having a causal association with AD, with 13 showing significant causal associations. During the replication validation phase, six metabolites and metabolite ratios were confirmed for their roles in AD: N-lactoyl tyrosine, argininate, and the adenosine 5'-monophosphate to flavin adenine dinucleotide ratio were found to exhibit protective effects against AD. In contrast, ergothioneine, piperine, and 1,7-dimethyluric acid were identified as contributing to an increased risk of AD. Among them, argininate showed a significant effect against AD. Replication and sensitivity analyses confirmed the robustness of these findings. Metabolic pathway analysis linked "Vitamin B6 metabolism" to AD risk. No genetic correlations were found, but colocalization analysis indicated potential AD risk elevation through top SNPs in APOE and PSEN2 genes. This provides novel insights into AD's etiology from a metabolomic viewpoint, suggesting both protective and risk metabolites.
Collapse
Affiliation(s)
- Di Cao
- Hubei University of Chinese Medicine, Wuhan, 430070, Hubei, China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, 430070, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, 430070, Hubei, China
| | - Yini Zhang
- Hubei University of Chinese Medicine, Wuhan, 430070, Hubei, China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, 430070, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, 430070, Hubei, China
| | - Shaobo Zhang
- Changchun University of Chinese Medicine, Changchun, 130000, Jilin, China
| | - Jun Li
- Department of Rehabilitation Medicine, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qiguang Yang
- The Second Affiliated Hospital of Changchun University of Chinese Medicine (Changchun Hospital of Chinese Medicine), Changchun, 130000, Jilin, China
| | - Ping Wang
- Hubei University of Chinese Medicine, Wuhan, 430070, Hubei, China.
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, 430070, Hubei, China.
- Hubei Shizhen Laboratory, Wuhan, 430070, Hubei, China.
| |
Collapse
|
5
|
Zhang Y, Spitzer BW, Zhang Y, Wallace DA, Yu B, Qi Q, Argos M, Avilés-Santa ML, Boerwinkle E, Daviglus ML, Kaplan R, Cai J, Redline S, Sofer T. Untargeted Metabolome Atlas for Sleep Phenotypes in the Hispanic Community Health Study/Study of Latinos. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.17.24307286. [PMID: 38798578 PMCID: PMC11118618 DOI: 10.1101/2024.05.17.24307286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Sleep is essential to maintaining health and wellbeing of individuals, influencing a variety of outcomes from mental health to cardiometabolic disease. This study aims to assess the relationships between various sleep phenotypes and blood metabolites. Utilizing data from the Hispanic Community Health Study/Study of Latinos, we performed association analyses between 40 sleep phenotypes, grouped in several domains (i.e., sleep disordered breathing (SDB), sleep duration, timing, insomnia symptoms, and heart rate during sleep), and 768 metabolites measured via untargeted metabolomics profiling. Network analysis was employed to visualize and interpret the associations between sleep phenotypes and metabolites. The patterns of statistically significant associations between sleep phenotypes and metabolites differed by superpathways, and highlighted subpathways of interest for future studies. For example, some xenobiotic metabolites were associated with sleep duration and heart rate phenotypes (e.g. 1H-indole-7-acetic acid, 4-allylphenol sulfate), while ketone bodies and fatty acid metabolism metabolites were associated with sleep timing measures (e.g. 3-hydroxybutyrate (BHBA), 3-hydroxyhexanoylcarnitine (1)). Heart rate phenotypes had the overall largest number of detected metabolite associations. Many of these associations were shared with both SDB and with sleep timing phenotypes, while SDB phenotypes shared relatively few metabolite associations with sleep duration measures. A number of metabolites were associated with multiple sleep phenotypes, from a few domains. The amino acids vanillylmandelate (VMA) and 1-carboxyethylisoleucine were associated with the greatest number of sleep phenotypes, from all domains other than insomnia. This atlas of sleep-metabolite associations will facilitate hypothesis generation and further study of the metabolic underpinnings of sleep health.
Collapse
Affiliation(s)
- Ying Zhang
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Brian W Spitzer
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yu Zhang
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Danielle A Wallace
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Maria Argos
- Department of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, IL, USA
- Department of Environmental Health, School of Public Health, Boston University, Boston, MA, USA
| | - M Larissa Avilés-Santa
- Division of Clinical and Health Services Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Eric Boerwinkle
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Susan Redline
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
6
|
Alhathli E, Julian T, Girach ZUA, Thompson AAR, Rhodes C, Gräf S, Errington N, Wilkins MR, Lawrie A, Wang D, Cooper‐Knock J. Mendelian Randomization Study With Clinical Follow-Up Links Metabolites to Risk and Severity of Pulmonary Arterial Hypertension. J Am Heart Assoc 2024; 13:e032256. [PMID: 38456412 PMCID: PMC11010003 DOI: 10.1161/jaha.123.032256] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/18/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) exhibits phenotypic heterogeneity and variable response to therapy. The metabolome has been implicated in the pathogenesis of PAH, but previous works have lacked power to implicate specific metabolites. Mendelian randomization (MR) is a method for causal inference between exposures and outcomes. METHODS AND RESULTS Using genome-wide association study summary statistics, we implemented MR analysis to test for potential causal relationships between serum concentration of 575 metabolites and PAH. Five metabolites were causally associated with the risk of PAH after multiple testing correction. Next, we measured serum concentration of candidate metabolites in an independent clinical cohort of 449 patients with PAH to check whether metabolite concentrations are correlated with markers of disease severity. Of the 5 candidates nominated by our MR work, serine was negatively associated and homostachydrine was positively associated with clinical severity of PAH via direct measurement in this independent clinical cohort. Finally we used conditional and orthogonal approaches to explore the biology underlying our lead metabolites. Rare variant burden testing was carried out using whole exome sequencing data from 578 PAH cases and 361 675 controls. Multivariable MR is an extension of MR that uses a single set of instrumental single-nucleotide polymorphisms to measure multiple exposures; multivariable MR is used to determine interdependence between the effects of different exposures on a single outcome. Rare variant analysis demonstrated that loss-of-function mutations within activating transcription factor 4, a transcription factor responsible for upregulation of serine synthesis under conditions of serine starvation, are associated with higher risk for PAH. Homostachydrine is a xenobiotic metabolite that is structurally related to l-proline betaine, which has previously been linked to modulation of inflammation and tissue remodeling in PAH. Our multivariable MR analysis suggests that the effect of l-proline betaine is actually mediated indirectly via homostachydrine. CONCLUSIONS Our data present a method for study of the metabolome in the context of PAH, and suggests several candidates for further evaluation and translational research.
Collapse
Affiliation(s)
- Elham Alhathli
- Sheffield Institute for Translational Neuroscience (SITraN), University of SheffieldSheffieldUK
- Department of Nursing, Faculty of Applied Medical SciencesTaif UniversityTaifSaudi Arabia
| | - Thomas Julian
- Division of Evolution, Infection and Genomics, School of Biological SciencesThe University of ManchesterManchesterUK
| | - Zain Ul Abideen Girach
- Sheffield Institute for Translational Neuroscience (SITraN), University of SheffieldSheffieldUK
| | - A. A. Roger Thompson
- Department of Infection, Immunity and Cardiovascular DiseaseUniversity of SheffieldSheffieldUK
| | | | - Stefan Gräf
- Department of Respiratory MedicineUniversity of CambridgeCambridgeUK
| | - Niamh Errington
- National Heart and Lung Institute, Imperial College LondonLondonUK
| | | | - Allan Lawrie
- National Heart and Lung Institute, Imperial College LondonLondonUK
| | - Dennis Wang
- Department of Computer ScienceUniversity of SheffieldSheffieldUK
- National Heart and Lung Institute, Imperial College LondonLondonUK
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR)SingaporeRepublic of Singapore
| | - Johnathan Cooper‐Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of SheffieldSheffieldUK
| |
Collapse
|
7
|
Zhai M, Zhang Y, Yan D, Wang Y, Li W, Sun J. Genetic Insights into the Association and Causality Between Blood Metabolites and Alzheimer's Disease. J Alzheimers Dis 2024; 98:885-896. [PMID: 38489174 DOI: 10.3233/jad-230985] [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: 03/17/2024]
Abstract
Background Alzheimer's disease (AD) is an increasing public health concern with the aging of the global population. Understanding the genetic correlation and potential causal relationships between blood metabolites and AD may provide important insights into the metabolic dysregulation underlying this neurodegenerative disorder. Objective The aim of this study was to investigate the causal relationship between blood metabolites and AD using Mendelian randomization (MR) analysis. Methods Association data were obtained from three large-scale genome-wide association studies of 486 blood metabolites (N = 7,824), AD (71,880 cases and 383,378 controls), early-onset AD (N = 303,760), and late-onset AD (N = 307,112). Causal associations between blood metabolites and AD were assessed using inverse variance weighting (IVW), MR-Egger, and weighted median methods. Bidirectional two-sample MR analysis was used to identify causal blood metabolites. MR-PRESSO, MR-Egger, and Cochran-Q were used to quantify instrumental variable heterogeneity and horizontal pleiotropy. Results Using MR and sensitivity analysis, we identified 40 blood metabolites with potential causal associations with AD. After applying false discovery rate (FDR) correction, two metabolites, gamma-glutamylphenylalanine (OR = 1.15, 95% CI: 1.06-1.24, p = 3.88×10-4, q = 0.09) and X-11317 (OR = 1.16, 95% CI: 1.08-1.26, p = 1.14×10-4, q = 0.05), retained significant associations with AD. Reverse MR analysis indicated no significant causal effect of AD on blood metabolites. No significant instrumental variable heterogeneity or horizontal pleiotropy was found. Conclusions This two-sample MR study provides compelling evidence for a potential causal relationship between blood metabolic dysregulation and susceptibility to AD. Further investigation of the biological relevance of the identified metabolites to AD and additional supporting evidence is warranted.
Collapse
Affiliation(s)
- Modi Zhai
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Zhang
- Beidahuang Industry Group General Hospital, Harbin, China
| | - Dongxue Yan
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuzhen Wang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wenzhong Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jie Sun
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Fazia T, Baldrighi GN, Nova A, Bernardinelli L. A systematic review of Mendelian randomization studies on multiple sclerosis. Eur J Neurosci 2023; 58:3172-3194. [PMID: 37463755 DOI: 10.1111/ejn.16088] [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: 04/03/2023] [Revised: 05/31/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023]
Abstract
Mendelian randomization (MR) is a powerful approach for assessing the causal effect of putative risk factors on an outcome, using genetic variants as instrumental variables. The methodology and application developed in the framework of MR have been dramatically improved, taking advantage of the many public genome-wide association study (GWAS) data. The availability of summary-level data allowed to perform numerous MR studies especially for complex diseases, pinpointing modifiable exposures causally related to increased or decreased disease risk. Multiple sclerosis (MS) is a complex multifactorial disease whose aetiology involves both genetic and non-genetic risk factors and their interplay. Previous observational studies have revealed associations between candidate modifiable exposures and MS risk; although being prone to confounding, and reverse causation, these studies were unable to draw causal conclusions. MR analysis addresses the limitations of observational studies and allows to establish reliable and accurate causal conclusions. Here, we systematically reviewed the studies evaluating the causal effect, through MR, of genetic and non-genetic exposures on MS risk. Among 107 papers found, only 42 were eligible for final evaluation and qualitative synthesis. We found that, above all, low vitamin D levels and high adult body mass index (BMI) appear to be uncontested risk factors for increased MS risk.
Collapse
Affiliation(s)
- Teresa Fazia
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | | | - Andrea Nova
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Luisa Bernardinelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
9
|
Yin X, Li J, Bose D, Okamoto J, Kwon A, Jackson AU, Silva LF, Oravilahti A, Stringham HM, Ripatti S, Daly M, Palotie A, Scott LJ, Burant CF, Fauman EB, Wen X, Boehnke M, Laakso M, Morrison J. Metabolome-wide Mendelian randomization characterizes heterogeneous and shared causal effects of metabolites on human health. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.26.23291721. [PMID: 37425837 PMCID: PMC10327254 DOI: 10.1101/2023.06.26.23291721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Metabolites are small molecules that are useful for estimating disease risk and elucidating disease biology. Nevertheless, their causal effects on human diseases have not been evaluated comprehensively. We performed two-sample Mendelian randomization to systematically infer the causal effects of 1,099 plasma metabolites measured in 6,136 Finnish men from the METSIM study on risk of 2,099 binary disease endpoints measured in 309,154 Finnish individuals from FinnGen. We identified evidence for 282 causal effects of 70 metabolites on 183 disease endpoints (FDR<1%). We found 25 metabolites with potential causal effects across multiple disease domains, including ascorbic acid 2-sulfate affecting 26 disease endpoints in 12 disease domains. Our study suggests that N-acetyl-2-aminooctanoate and glycocholenate sulfate affect risk of atrial fibrillation through two distinct metabolic pathways and that N-methylpipecolate may mediate the causal effect of N6, N6-dimethyllysine on anxious personality disorder. This study highlights the broad causal impact of plasma metabolites and widespread metabolic connections across diseases.
Collapse
|
10
|
Jie L, Ma Z, Gao Y, Shi X, Yu L, Mao J, Wang P. The mechanism of palmatine-mediated intestinal flora and host metabolism intervention in OA-OP comorbidity rats. Front Med (Lausanne) 2023; 10:1153360. [PMID: 37153081 PMCID: PMC10159182 DOI: 10.3389/fmed.2023.1153360] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/28/2023] [Indexed: 05/09/2023] Open
Abstract
Background ErXian decoction is a Chinese herbal compound that can prevent and control the course of osteoarthritis (OA) and osteoporosis (OP). OP and OA are two age-related diseases that often coexist in elderly individuals, and both are associated with dysregulation of the gut microbiome. In the initial study, Palmatine (PAL) was obtained by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and network pharmacological screening techniques, followed by 16S rRNA sequencing and serum metabolomics of intestinal contents, to explore the mechanism of PAL in the treatment of OA and OP. Methods The rats selected for this study were randomly divided into three groups: a sham group, an OA-OP group and a PAL group. The sham group was intragastrically administered normal saline solution, and the PLA group was treated with PAL for 56 days. Through microcomputed tomography (micro-CT), ELISA, 16S rRNA gene sequencing and non-targeted metabonomics research, we explored the potential mechanism of intestinal microbiota and serum metabolites in PAL treatment of OA-OP rats. Results Palmatine significantly repair bone microarchitecture of rat femur in OA-OP rats and improved cartilage damage. The analysis of intestinal microflora showed that PAL could also improve the intestinal microflora disorder of OA-OP rats. For example, the abundance of Firmicutes, Bacteroidota, Actinobacteria, Lactobacillus, unclassified_f_Lachnospiraceae, norank_f_Muribaculaceae, Lactobacillaceae, Lachnospiraceae and Muribaculaceae increased after PAL intervention. In addition, the results of metabolomics data analysis showed that PAL also change the metabolic status of OA-OP rats. After PAL intervention, metabolites such as 5-methoxytryptophol, 2-methoxy acetaminophen sulfate, beta-tyrosine, indole-3-carboxylic acid-O-sulfate and cyclodopa glucoside increased. Association analysis of metabolomics and gut microbiota (GM) showed that the communication of multiple flora and different metabolites played an important role in OP and OA. Conclusion Palmatine can improve cartilage degeneration and bone loss in OA-OP rats. The evidence we provided supports the idea that PAL improves OA-OP by altering GM and serum metabolites. In addition, the application of GM and serum metabolomics correlation analysis provides a new strategy for uncovering the mechanism of herbal treatment for bone diseases.
Collapse
Affiliation(s)
- Lishi Jie
- Department of Orthopaedics and Traumatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenyuan Ma
- Department of Orthopaedics and Traumatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yifan Gao
- Department of Orthopaedics and Traumatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoqing Shi
- Department of Orthopaedics and Traumatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Likai Yu
- Department of Orthopaedics and Traumatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Mao
- Department of Orthopaedics and Traumatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Peimin Wang
- Department of Orthopaedics and Traumatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Peimin Wang,
| |
Collapse
|