Genome‐scale metabolic modeling reveals SARS‐CoV‐2‐induced metabolic changes and antiviral targets

Tremendous progress has been made to control the COVID-19 pandemic caused by the SARS-CoV-2 virus. However, effective therapeutic options are still rare. Drug repurposing and combination represent practical strategies to address this urgent unmet medical need. Viruses, including coronaviruses, are known to hijack host metabolism to facilitate viral proliferation, making targeting host metabolism a promising antiviral approach. Here, we describe an integrated analysis of 12 published in vitro and human patient gene expression datasets on SARS-CoV-2 infection using genome-scale metabolic modeling (GEM), revealing complicated host metabolism reprogramming during SARS-CoV-2 infection. We next applied the GEM-based metabolic transformation algorithm to predict anti-SARS-CoV-2 targets that counteract the virus-induced metabolic changes. We successfully validated these targets using published drug and genetic screen data and by performing an siRNA assay in Caco-2 cells. Further generating and analyzing RNA-sequencing data of remdesivir-treated Vero E6 cell samples, we predicted metabolic targets acting in combination with remdesivir, an approved anti-SARS-CoV-2 drug. Our study provides clinical data-supported candidate anti-SARS-CoV-2 targets for future evaluation, demonstrating host metabolism-targeting as a promising antiviral strategy.

[1]  E. Graviss,et al.  Risk factors for severity of COVID-19 in hospital patients age 18–29 years , 2021, PloS one.

[2]  T. Spector,et al.  Early detection of COVID-19 in the UK using self-reported symptoms: a large-scale, prospective, epidemiological surveillance study , 2021, The Lancet Digital Health.

[3]  Jennifer G. Abelin,et al.  Profiling SARS-CoV-2 HLA-I peptidome reveals T cell epitopes from out-of-frame ORFs , 2021, Cell.

[4]  Mary K. Lewinski,et al.  Functional landscape of SARS-CoV-2 cellular restriction , 2021, Molecular Cell.

[5]  Evan T. Sholle,et al.  Shotgun transcriptome, spatial omics, and isothermal profiling of SARS-CoV-2 infection reveals unique host responses, viral diversification, and drug interactions , 2021, Nature Communications.

[6]  Shane O’Carroll,et al.  Targeting immunometabolism to treat COVID-19 , 2021, Immunotherapy advances.

[7]  C. Demirdover COVID-19-vaccines , 2021, Reactions Weekly.

[8]  Minoru Kanehisa,et al.  KEGG: integrating viruses and cellular organisms , 2020, Nucleic Acids Res..

[9]  Silva Kasela,et al.  Identification of Required Host Factors for SARS-CoV-2 Infection in Human Cells , 2020, Cell.

[10]  Peter C. DeWeirdt,et al.  Genome-wide CRISPR Screens Reveal Host Factors Critical for SARS-CoV-2 Infection , 2020, Cell.

[11]  W. Lim,et al.  Dexamethasone in Hospitalized Patients with Covid-19 , 2021 .

[12]  J. Bloom,et al.  Metabolic reprogramming and epigenetic changes of vital organs in SARS-CoV-2–induced systemic toxicity , 2020, JCI Insight.

[13]  Yong-Gang Yao,et al.  Longitudinal transcriptome analyses show robust T cell immunity during recovery from COVID-19 , 2020, Signal Transduction and Targeted Therapy.

[14]  H. Peckham,et al.  Male sex identified by global COVID-19 meta-analysis as a risk factor for death and ITU admission , 2020, Nature Communications.

[15]  G. Cappellano,et al.  Large-Scale Plasma Analysis Revealed New Mechanisms and Molecules Associated with the Host Response to SARS-CoV-2 , 2020, International journal of molecular sciences.

[16]  F. Cheng,et al.  A network medicine approach to investigation and population-based validation of disease manifestations and drug repurposing for COVID-19 , 2020, PLoS biology.

[17]  R. Malekzadeh,et al.  Repurposed antiviral drugs for COVID-19; interim WHO SOLIDARITY trial results , 2020, medRxiv.

[18]  Min Zheng,et al.  A systematic review of SARS-CoV-2 vaccine candidates , 2020, Signal Transduction and Targeted Therapy.

[19]  Jennifer G. Abelin,et al.  SARS-CoV-2 infected cells present HLA-I peptides from canonical and out-of-frame ORFs , 2020, bioRxiv.

[20]  E. Piver,et al.  The Role of Phosphatidylinositol Phosphate Kinases during Viral Infection , 2020, Viruses.

[21]  T. Ideker,et al.  Functional Landscape of SARS-CoV-2 Cellular Restriction , 2020, bioRxiv.

[22]  T. Tuschl,et al.  Discovery of SARS-CoV-2 antiviral synergy between remdesivir and approved drugs in human lung cells , 2020, bioRxiv.

[23]  Ruth Nussinov,et al.  Artificial intelligence in COVID-19 drug repurposing , 2020, The Lancet Digital Health.

[24]  P. Shi,et al.  In vivo antiviral host transcriptional response to SARS-CoV-2 by viral load, sex, and age , 2020, PLoS biology.

[25]  S. Ciesek,et al.  Targeting pentose phosphate pathway for SARS-CoV-2 therapy , 2020, bioRxiv.

[26]  Arti T. Navare,et al.  Crippling life support for SARS-CoV-2 and other viruses through synthetic lethality , 2020, The Journal of cell biology.

[27]  Yong Wang,et al.  Novel and potent inhibitors targeting DHODH are broad-spectrum antivirals against RNA viruses including newly-emerged coronavirus SARS-CoV-2 , 2020, Protein & Cell.

[28]  Kathleen M. Jagodnik,et al.  The COVID-19 Drug and Gene Set Library , 2020, Patterns.

[29]  Eytan Ruppin,et al.  Discovery of SARS-CoV-2 Antivirals through Large-scale Drug Repositioning , 2020, Nature.

[30]  Matan Hofree,et al.  The SARS-CoV-2 Transcriptional Metabolic Signature in Lung Epithelium , 2020 .

[31]  F. Cheng,et al.  A Network Medicine Approach to Investigation and Population-based Validation of Disease Manifestations and Drug Repurposing for COVID-19 , 2020, ChemRxiv : the preprint server for chemistry.

[32]  Roland Eils,et al.  COVID-19 severity correlates with airway epithelium–immune cell interactions identified by single-cell analysis , 2020, Nature Biotechnology.

[33]  S. Fonseca,et al.  Immune and Metabolic Signatures of COVID-19 Revealed by Transcriptomics Data Reuse , 2020, Frontiers in Immunology.

[34]  Jennifer L. Bell,et al.  Effect of Dexamethasone in Hospitalized Patients with COVID-19: Preliminary Report , 2020, medRxiv.

[35]  E. Hod,et al.  COVID-19 infection alters kynurenine and fatty acid metabolism, correlating with IL-6 levels and renal status. , 2020, JCI insight.

[36]  M. Mann,et al.  Multilevel proteomics reveals host perturbations by SARS-CoV-2 and SARS-CoV , 2020, Nature.

[37]  L. Dodd,et al.  Remdesivir for the Treatment of Covid-19 — Final Report , 2020, The New England journal of medicine.

[38]  S. Ciesek,et al.  Proteomics of SARS-CoV-2-infected host cells reveals therapy targets , 2020, Nature.

[39]  R. Schwartz,et al.  Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19 , 2020, Cell.

[40]  Lin Cheng,et al.  Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19 , 2020, Nature Medicine.

[41]  Benjamin J. Polacco,et al.  A SARS-CoV-2 Protein Interaction Map Reveals Targets for Drug-Repurposing , 2020, Nature.

[42]  M. Tay,et al.  The trinity of COVID-19: immunity, inflammation and intervention , 2020, Nature Reviews Immunology.

[43]  Jean Thierry-Mieg,et al.  Shotgun Transcriptome and Isothermal Profiling of SARS-CoV-2 Infection Reveals Unique Host Responses, Viral Diversification, and Drug Interactions , 2020, bioRxiv.

[44]  Huanhuan Gao,et al.  Proteomic and Metabolomic Characterization of COVID-19 Patient Sera , 2020, Cell.

[45]  Yan Liu,et al.  Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV , 2020, Nature Communications.

[46]  G. Herrler,et al.  SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor , 2020, Cell.

[47]  Kai Zhao,et al.  A pneumonia outbreak associated with a new coronavirus of probable bat origin , 2020, Nature.

[48]  M. Shi,et al.  Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients , 2020, Emerging microbes & infections.

[49]  Edith Le Floch,et al.  Clustering and variable selection evaluation of 13 unsupervised methods for multi-omics data integration , 2019, Briefings Bioinform..

[50]  Luis Tobalina,et al.  rMTA: robust metabolic transformation analysis , 2019, Bioinform..

[51]  Gennady Korotkevich,et al.  Fast gene set enrichment analysis , 2019, bioRxiv.

[52]  G. Zlabinger,et al.  Hijacking the Supplies: Metabolism as a Novel Facet of Virus-Host Interaction , 2019, Front. Immunol..

[53]  Gi Bae Kim,et al.  Current status and applications of genome-scale metabolic models , 2019, Genome Biology.

[54]  E. Ruppin,et al.  Mitochondrial Regulation of the Hippocampal Firing Rate Set Point and Seizure Susceptibility , 2019, Neuron.

[55]  Dong Yang,et al.  SREBP-dependent lipidomic reprogramming as a broad-spectrum antiviral target , 2019, Nature Communications.

[56]  J. Chan,et al.  Characterization of the Lipidomic Profile of Human Coronavirus-Infected Cells: Implications for Lipid Metabolism Remodeling upon Coronavirus Replication , 2019, Viruses.

[57]  Christoph Hafemeister,et al.  Comprehensive integration of single cell data , 2018, bioRxiv.

[58]  K. Allegaert,et al.  (Preprint) , 2018 .

[59]  David S. Wishart,et al.  DrugBank 5.0: a major update to the DrugBank database for 2018 , 2017, Nucleic Acids Res..

[60]  Henning Hermjakob,et al.  The Reactome pathway knowledgebase , 2013, Nucleic Acids Res..

[61]  Daniel C. Zielinski,et al.  Recon3D enables a three-dimensional view of gene variation in human metabolism , 2018 .

[62]  Eytan Ruppin,et al.  An integrated computational and experimental study uncovers FUT9 as a metabolic driver of colorectal cancer , 2017, Molecular systems biology.

[63]  Rob Patro,et al.  Salmon provides fast and bias-aware quantification of transcript expression , 2017, Nature Methods.

[64]  Roland Eils,et al.  Complex heatmaps reveal patterns and correlations in multidimensional genomic data , 2016, Bioinform..

[65]  P. Linsley,et al.  MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data , 2015, Genome Biology.

[66]  M. Lagunoff,et al.  Viral activation of cellular metabolism. , 2015, Virology.

[67]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[68]  Charity W. Law,et al.  voom: precision weights unlock linear model analysis tools for RNA-seq read counts , 2014, Genome Biology.

[69]  Eytan Ruppin,et al.  Model-based identification of drug targets that revert disrupted metabolism and its application to ageing , 2013, Nature Communications.

[70]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[71]  D. Martens,et al.  Genome-scale metabolic models: reconstruction and analysis. , 2012, Methods in molecular biology.

[72]  Helga Thorvaldsdóttir,et al.  Molecular signatures database (MSigDB) 3.0 , 2011, Bioinform..

[73]  Xavier Robin,et al.  pROC: an open-source package for R and S+ to analyze and compare ROC curves , 2011, BMC Bioinformatics.

[74]  Pedro M. Valero-Mora,et al.  ggplot2: Elegant Graphics for Data Analysis , 2010 .

[75]  Luis Serrano,et al.  Correlation of mRNA and protein in complex biological samples , 2009, FEBS letters.

[76]  Markus J. Herrgård,et al.  Network-based prediction of human tissue-specific metabolism , 2008, Nature Biotechnology.

[77]  Herbert Stegemann,et al.  [The International Clinical Trials Registry Platform - ICTRP]. , 2007, Archivos latinoamericanos de nutricion.

[78]  Monica L. Mo,et al.  Global reconstruction of the human metabolic network based on genomic and bibliomic data , 2007, Proceedings of the National Academy of Sciences.

[79]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[80]  J. Samet,et al.  Food and Drug Administration , 2007, BMJ : British Medical Journal.