PT - JOURNAL ARTICLE AU - Català-Senent, José Francisco AU - Andreu, Zoraida AU - Hidalgo, Marta R. AU - Roig, Francisco José AU - Yanguas-Casás, Natalia AU - Neva-Alejo, Almudena AU - López-Cerdán, Adolfo AU - Soler-Sáez, Irene AU - de la Iglesia-Vayá, María AU - García-García, Francisco TI - A Deep Transcriptome Meta-Analysis Reveals Sex-based Molecular Differences in Multiple Sclerosis AID - 10.1101/2021.08.31.21262175 DP - 2022 Jan 01 TA - medRxiv PG - 2021.08.31.21262175 4099 - http://medrxiv.org/content/early/2022/05/31/2021.08.31.21262175.short 4100 - http://medrxiv.org/content/early/2022/05/31/2021.08.31.21262175.full AB - Background Multiple sclerosis (MS), a chronic auto-immune, inflammatory, and degenerative disease of the central nervous system, affects both males and females; however, females suffer from a higher risk of developing MS (2-3:1 ratio compared to males). Current knowledge does not allow a precise definition of the sex-based factors influencing MS. Here, we explore the role of sex in MS to identify potential molecular mechanisms underlying sex-based differences that may guide novel therapeutic approaches tailored for males or females.Methods We performed a rigorous and systematic review of whole transcriptome studies of MS that included patient information regarding sex in Gene Expression Omnibus and ArrayExpress databases following PRISMA statement guidelines. We analyzed differential gene expression for each selected study and addressed 3 meta-analyses based on genes to evaluate common features and sex bias: the first meta-analysis of 4 nervous tissue studies, a second in 5 blood studies, and a third integrating 9 studies from both tissues. Finally, we performed a gene set analysis on the meta-analyzed differential transcriptomic profiles of the nervous system to study sex-based differences in biological pathways and phenotypes (physiological and pathological states).Results After screening 122 publications, the systematic review provided a selection of 9 studies (5 in blood and 4 in nervous tissue) with a total of 474 samples (189 MS females and 109 control females; 82 MS males and 94 control males). The tissue-specific meta-analysis identified the overexpression of KIR2DL3 in blood in females and 13 genes with a sex-based differential expression pattern in the nervous system (7 overexpressed in females: ARL17B, CECR7, CEP78, STMP1, TRAF3IP2-AS1, ZNF117 and ZNF488; and 6 overexpressed in males: IFFO2, LOC401127, NUDT18, RNF10, SLC17A5, and UBXN2B). The two-tissue meta-analysis detected a single gene overexpressed in females (LOC102723701). Functional analyses revealed different altered immune scenarios in females and males. A pro-inflammatory environment and innate immune responses related to myeloid linage predominate in females, while in males, adaptative responses associated with the lymphocyte linage. Additionally, MS females displayed alterations in mitochondrial respiratory chain complexes, purine, and glutamate metabolism, while MS males displayed alterations in stress response to metal ion, amine, and amino acid transport.Conclusion We found transcriptomic and functional differences between MS males and females (especially in the immune system), which may support the development of sex-specific treatments. Our study highlights the importance of understanding sex as a variable in MS.Competing Interest StatementThe authors have declared no competing interest.Funding StatementResearch supported by Principe Felipe Research Center and GV/2020/186 grant.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:This work does not need any approval of the IRB.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.YesI 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).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesThe large volume of data and results generated in this study are freely available through the Metafun-MS web tool: http://bioinfo.cipf.es/metafun-MS and the zenodo repository: http://doi.org/10.5281/zenodo.6344450. This study analyzed transcriptomic data available in the Gene Expression Omnibus database with accession numbers GSE37750, GSE41848, GSE41849, GSE41890, GSE62584, GSE108000, GSE111972, GSE131281, and GSE135511. http://doi.org/10.5281/zenodo.6344450 BHBenjamini & Hochberg p-value adjust methodBPBiological processes of Gene OntologyCISClinical Isolated SyndromeCNScentral nervous systemDGEDifferential gene expressionGEOGenes Omnibus Expression databaseGOGene OntologyGSAGene set analysisIDFImpact of the disease in femalesIDMImpact of the disease in malesILInterleukinKEGGKyoto Encyclopedia of Genes and GenomesLFCLogarithm of the fold changeLORLogarithm of odds ratioMSMultiple sclerosisPCAPrincipal component analysisPRISMAPreferred Reporting Items for Systematic Reviews and Meta-AnalysesPPMSProgressive Primary Multiple SclerosisPRMSProgressive-Relapse Multiple SclerosisRRMSRelapsing-Remitting Multiple SclerosisSDIDSex-differential impact of the diseaseSPMSSecondary-Progressive Multiple Sclerosis