Skip to main content
medRxiv
  • Home
  • About
  • Submit
  • ALERTS / RSS
Advanced Search

Profiling the genome and proteome of metabolic dysfunction-associated steatotic liver disease identifies potential therapeutic targets

View ORCID ProfileJun Liu, Sile Hu, Lingyan Chen, Charlotte Daly, Cesar Augusto Prada Medina, View ORCID ProfileTom G Richardson, Matthew Traylor, Niall J Dempster, Richard Mbasu, Thomas Monfeuga, View ORCID ProfileMarijana Vujkovic, Philip S Tsao, Julie A Lynch, View ORCID ProfileBenjamin F. Voight, View ORCID ProfileKyong-Mi Chang, VA Million Veteran Program, Jeremy F Cobbold, Jeremy W Tomlinson, Cornelia M van Duijn, Joanna M M Howson
doi: https://doi.org/10.1101/2023.11.30.23299247
Jun Liu
1Nuffield Department of Population Health, University of Oxford, Oxford, UK
2Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
MBChB, PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Jun Liu
  • For correspondence: jun.liu{at}ndph.ox.ac.uk
Sile Hu
2Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lingyan Chen
2Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Charlotte Daly
3Department of Discovery Technology and Genomics, Novo Nordisk Research Centre Oxford, Oxford, UK
4Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Cesar Augusto Prada Medina
5AI & Digital Research, Research & Early Development, Novo Nordisk Research Centre Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tom G Richardson
2Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
6MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Tom G Richardson
Matthew Traylor
2Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Niall J Dempster
4Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
MBChB DPhil
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Richard Mbasu
3Department of Discovery Technology and Genomics, Novo Nordisk Research Centre Oxford, Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Thomas Monfeuga
5AI & Digital Research, Research & Early Development, Novo Nordisk Research Centre Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marijana Vujkovic
7Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
8Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
9Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Marijana Vujkovic
Philip S Tsao
10VA Palo Alto Health Care System, Palo Alto, CA, USA
11Department of Cardiovascular Medicine, School of Medicine, Stanford University, Stanford, CA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Julie A Lynch
12VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, Utah, USA
13Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Benjamin F. Voight
7Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
14Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
15Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
16Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Benjamin F. Voight
Kyong-Mi Chang
7Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
8Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Kyong-Mi Chang
Jeremy F Cobbold
17NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust and the University of Oxford, Oxford, UK
18Translational Gastroenterology Unit, University of Oxford, Oxford, UK
MBChB, PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jeremy W Tomlinson
4Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
17NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust and the University of Oxford, Oxford, UK
MBChB, PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Cornelia M van Duijn
1Nuffield Department of Population Health, University of Oxford, Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joanna M M Howson
2Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
PhD
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Abstract
  • Full Text
  • Info/History
  • Metrics
  • Supplementary material
  • Data/Code
  • Preview PDF
Loading

ABSTRACT

BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) affects over 25% of the population and currently has no effective treatments. Plasma proteins with causal evidence may represent promising drug targets. We aimed to identify plasma proteins in the causal pathway of MASLD and explore their interaction with obesity.

METHODS We analysed 2,941 plasma proteins in 43,978 European participants from UK Biobank. We performed genome-wide association study (GWAS) for all MASLD-associated proteins and created the largest MASLD GWAS (109,885 cases/1,014,923 controls). We performed Mendelian Randomization (MR) and integrated proteins and their encoding genes in MASLD ranges to identify candidate causal proteins. We then validated them through independent replication, exome sequencing, liver imaging, bulk and single-cell gene expression, liver biopsies, pathway, and phenome-wide data. We explored the role of obesity by MR and multivariable MR across proteins, body mass index, and MASLD.

RESULTS We found 929 proteins associated with MASLD, reported five novel genetic loci associated with MASLD, and identified 17 candidate MASLD protein targets. We identified four novel targets for MASLD (CD33, GRHPR, HMOX2, and SCG3), provided protein evidence supporting roles of AHCY, FCGR2B, ORM1, and RBKS in MASLD, and validated nine previously known targets. We found that CD33, FCGR2B, ORM1, RBKS, and SCG3 mediated the association of obesity and MASLD, and HMOX2, ORM1, and RBKS had effect on MASLD independent of obesity.

CONCLUSIONS This study identified new protein targets in the causal pathway of MASLD, providing new insights into the multi-omics architecture and pathophysiology of MASLD. These findings advise further therapeutic interventions for MASLD.

Competing Interest Statement

The authors declare the following competing interests: S.H., L.C., C.D., C.A.P.M., M.T., R.M., T.M. and J.M.M.H. are full-time employees of Novo Nordisk. T.G.R. was part-time employee of Novo Nordisk during the project running. J.L. is supported by a University of Oxford Novo Nordisk Research Fellowship. N.D. was supported by a University of Oxford Novo Nordisk Research Training Fellowship. The remaining authors declare no competing interests.

Funding Statement

This research is based on data from the MVP, Office of Research and Development, Veterans Health Administration and was supported by award MVP000 (mvp003/028b). MVP MASLD data was generated with Department of Veterans Affairs (VA) funding support (I01-BX003362: K.M.C., P.S.T., and M.V.) with additional support from resources and facilities of the VA Informatics and Computing Infrastructure (VINCI), VA HSR RES 13-457. This publication does not represent the views of the Department of Veterans Affairs or the United States Government. M.V. acknowledges support for this work from the National Institutes of Health (NIH)/National Institute of Diabetes and Digestive and Kidney Diseases, grant R01 DK134575. J.L. is supported by a Novo Nordisk Postdoctoral Fellowship and N.D. through a Novo Nordisk clinical research training fellowship run in partnership with the University of Oxford. The work has been supported by the Medical Research Council (JWT) and by the Oxford NIHR Biomedical Research Centre (JWT).

Author Declarations

I confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.

Yes

The details of the IRB/oversight body that provided approval or exemption for the research described are given below:

All participants provided electronically signed informed consent. UK Biobank has approval from the North West Multi-centre Research Ethics Committee, the Patient Information Advisory Group, and the Community Health Index Advisory Group. The current study is a part of UK Biobank project 53639 and 65851.All participants gave full, informed, written consent for the liver biopsy (NHS research ethics reference: 17/WM/0130).

I confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.

Yes

I understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).

Yes

I have followed all appropriate research reporting guidelines, such as any relevant EQUATOR Network research reporting checklist(s) and other pertinent material, if applicable.

Yes

Footnotes

  • ↵* Authors share last authorship

Data availability

UK Biobank data are publicly available to bona fide researchers upon application at http://www.ukbiobank.ac.uk/using-the-resource/. Publicly available summary statistics are obtained from https://gwas.mrcieu.ac.uk/, https://www.ebi.ac.uk/gwas/, https://www.gtexportal.org/home/datasets, and https://pan.ukbb.broadinstitute.org. Bulk liver RNA-seq data are available in Gene Expression Omnibus (GEO) under accession GSE135251. SnRNA-seq and scRNA-seq data are available in GEO under accession GSE189175, GSE185477, GSE136103, GSE212837, GSE189600, and GSE192740. Other source of data or web source were clarified in the methods. Relevant detailed results generated in this study were presented in supplementary tables. Clinical data with respect to the liver biopsy samples were assessable through contacting J.W.T.

  • ABBREVIATIONS

    MASLD
    metabolic dysfunction-associated steatotic liver disease
    NAFLD
    non-alcoholic fatty liver disease
    MASH
    metabolic dysfunction-associated steatohepatitis
    NASH
    non-alcoholic steatohepatitis
    MR
    Mendelian randomization
    MRI-PDFF
    magnetic resonance imaging - estimated proton density fat fraction
    ICD
    International Classification of Diseases
    LC-MS/MS
    liquid chromatography-tandem mass spectrometry
    MSD
    Matrix Spectral Decomposition
    GWAS
    genome-wide association study
    ALT
    alanine aminotransferase
    PheMR
    phenome-wide Mendelian randomization study
    pQTL
    protein quantitative trait locus
    eQTL
    expression quantitative trait locus
    BMI
    body mass index
    VEP
    variant effect predictor
    LOFTEE
    loss-of-function transcript effect estimator
    DEG
    differentially expressed genes
    snRNA-seq
    single nucleus RNA sequence
    scRNA-seq
    single cell RNA sequence
    age2
    age squared
    SNP
    single-nucleotide polymorphism
    NAFL
    non-alcoholic fatty liver
    GRHPR
    Glyoxylate and hydroxypyruvate reductase
    HMOX2
    heme oxygenase 2
    HMOX1
    heme oxygenase 1
    IGF
    insulin-like growth factor
    SCG3
    secretogranin III
    RBKS
    ribokinase
    ORM1
    Orosomucoid 1
    AHCY
    adenosylhomocysteinase
    FCGR2B
    Fc gamma receptor IIb.
  • Copyright 
    The copyright holder for this preprint is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC-ND 4.0 International license.
    Back to top
    PreviousNext
    Posted November 30, 2023.
    Download PDF

    Supplementary Material

    Data/Code
    Email

    Thank you for your interest in spreading the word about medRxiv.

    NOTE: Your email address is requested solely to identify you as the sender of this article.

    Enter multiple addresses on separate lines or separate them with commas.
    Profiling the genome and proteome of metabolic dysfunction-associated steatotic liver disease identifies potential therapeutic targets
    (Your Name) has forwarded a page to you from medRxiv
    (Your Name) thought you would like to see this page from the medRxiv website.
    CAPTCHA
    This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
    Share
    Profiling the genome and proteome of metabolic dysfunction-associated steatotic liver disease identifies potential therapeutic targets
    Jun Liu, Sile Hu, Lingyan Chen, Charlotte Daly, Cesar Augusto Prada Medina, Tom G Richardson, Matthew Traylor, Niall J Dempster, Richard Mbasu, Thomas Monfeuga, Marijana Vujkovic, Philip S Tsao, Julie A Lynch, Benjamin F. Voight, Kyong-Mi Chang, VA Million Veteran Program, Jeremy F Cobbold, Jeremy W Tomlinson, Cornelia M van Duijn, Joanna M M Howson
    medRxiv 2023.11.30.23299247; doi: https://doi.org/10.1101/2023.11.30.23299247
    Twitter logo Facebook logo LinkedIn logo Mendeley logo
    Citation Tools
    Profiling the genome and proteome of metabolic dysfunction-associated steatotic liver disease identifies potential therapeutic targets
    Jun Liu, Sile Hu, Lingyan Chen, Charlotte Daly, Cesar Augusto Prada Medina, Tom G Richardson, Matthew Traylor, Niall J Dempster, Richard Mbasu, Thomas Monfeuga, Marijana Vujkovic, Philip S Tsao, Julie A Lynch, Benjamin F. Voight, Kyong-Mi Chang, VA Million Veteran Program, Jeremy F Cobbold, Jeremy W Tomlinson, Cornelia M van Duijn, Joanna M M Howson
    medRxiv 2023.11.30.23299247; doi: https://doi.org/10.1101/2023.11.30.23299247

    Citation Manager Formats

    • BibTeX
    • Bookends
    • EasyBib
    • EndNote (tagged)
    • EndNote 8 (xml)
    • Medlars
    • Mendeley
    • Papers
    • RefWorks Tagged
    • Ref Manager
    • RIS
    • Zotero
    • Tweet Widget
    • Facebook Like
    • Google Plus One

    Subject Area

    • Gastroenterology
    Subject Areas
    All Articles
    • Addiction Medicine (349)
    • Allergy and Immunology (668)
    • Allergy and Immunology (668)
    • Anesthesia (181)
    • Cardiovascular Medicine (2648)
    • Dentistry and Oral Medicine (316)
    • Dermatology (223)
    • Emergency Medicine (399)
    • Endocrinology (including Diabetes Mellitus and Metabolic Disease) (942)
    • Epidemiology (12228)
    • Forensic Medicine (10)
    • Gastroenterology (759)
    • Genetic and Genomic Medicine (4103)
    • Geriatric Medicine (387)
    • Health Economics (680)
    • Health Informatics (2657)
    • Health Policy (1005)
    • Health Systems and Quality Improvement (985)
    • Hematology (363)
    • HIV/AIDS (851)
    • Infectious Diseases (except HIV/AIDS) (13695)
    • Intensive Care and Critical Care Medicine (797)
    • Medical Education (399)
    • Medical Ethics (109)
    • Nephrology (436)
    • Neurology (3882)
    • Nursing (209)
    • Nutrition (577)
    • Obstetrics and Gynecology (739)
    • Occupational and Environmental Health (695)
    • Oncology (2030)
    • Ophthalmology (585)
    • Orthopedics (240)
    • Otolaryngology (306)
    • Pain Medicine (250)
    • Palliative Medicine (75)
    • Pathology (473)
    • Pediatrics (1115)
    • Pharmacology and Therapeutics (466)
    • Primary Care Research (452)
    • Psychiatry and Clinical Psychology (3432)
    • Public and Global Health (6527)
    • Radiology and Imaging (1403)
    • Rehabilitation Medicine and Physical Therapy (814)
    • Respiratory Medicine (871)
    • Rheumatology (409)
    • Sexual and Reproductive Health (410)
    • Sports Medicine (342)
    • Surgery (448)
    • Toxicology (53)
    • Transplantation (185)
    • Urology (165)