Your browser doesn't support javascript.
loading
The relationship between extreme inter-individual variation in macrophage gene expression and genetic susceptibility to inflammatory bowel disease.
O'Brien, Claire L; Summers, Kim M; Martin, Natalia M; Carter-Cusack, Dylan; Yang, Yuanhao; Barua, Rasel; Dixit, Ojas V A; Hume, David A; Pavli, Paul.
Affiliation
  • O'Brien CL; Centre for Research in Therapeutics Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.
  • Summers KM; Inflammatory Bowel Disease Research Group, Canberra Hospital, Canberra, ACT, Australia.
  • Martin NM; Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia.
  • Carter-Cusack D; Inflammatory Bowel Disease Research Group, Canberra Hospital, Canberra, ACT, Australia.
  • Yang Y; Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia.
  • Barua R; Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia.
  • Dixit OVA; Inflammatory Bowel Disease Research Group, Canberra Hospital, Canberra, ACT, Australia.
  • Hume DA; Centre for Research in Therapeutics Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.
  • Pavli P; Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia. David.Hume@uq.edu.au.
Hum Genet ; 143(3): 233-261, 2024 Mar.
Article in En | MEDLINE | ID: mdl-38421405
ABSTRACT
The differentiation of resident intestinal macrophages from blood monocytes depends upon signals from the macrophage colony-stimulating factor receptor (CSF1R). Analysis of genome-wide association studies (GWAS) indicates that dysregulation of macrophage differentiation and response to microorganisms contributes to susceptibility to chronic inflammatory bowel disease (IBD). Here, we analyzed transcriptomic variation in monocyte-derived macrophages (MDM) from affected and unaffected sib pairs/trios from 22 IBD families and 6 healthy controls. Transcriptional network analysis of the data revealed no overall or inter-sib distinction between affected and unaffected individuals in basal gene expression or the temporal response to lipopolysaccharide (LPS). However, the basal or LPS-inducible expression of individual genes varied independently by as much as 100-fold between subjects. Extreme independent variation in the expression of pairs of HLA-associated transcripts (HLA-B/C, HLA-A/F and HLA-DRB1/DRB5) in macrophages was associated with HLA genotype. Correlation analysis indicated the downstream impacts of variation in the immediate early response to LPS. For example, variation in early expression of IL1B was significantly associated with local SNV genotype and with subsequent peak expression of target genes including IL23A, CXCL1, CXCL3, CXCL8 and NLRP3. Similarly, variation in early IFNB1 expression was correlated with subsequent expression of IFN target genes. Our results support the view that gene-specific dysregulation in macrophage adaptation to the intestinal milieu is associated with genetic susceptibility to IBD.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Inflammatory Bowel Diseases / Lipopolysaccharides / Genetic Predisposition to Disease / Macrophages Limits: Female / Humans / Male Language: En Journal: Hum Genet Year: 2024 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Inflammatory Bowel Diseases / Lipopolysaccharides / Genetic Predisposition to Disease / Macrophages Limits: Female / Humans / Male Language: En Journal: Hum Genet Year: 2024 Document type: Article