Your browser doesn't support javascript.
loading
Variant-to-function mapping of late-onset Alzheimer's disease GWAS signals in human microglial cell models implicates RTFDC1 at the CASS4 locus.
Burton, Elizabeth A; Argenziano, Mariana; Cook, Kieona; Ridler, Molly; Lu, Sumei; Su, Chun; Manduchi, Elisabetta; Littleton, Sheridan H; Leonard, Michelle E; Hodge, Kenyaita M; Wang, Li-San; Schellenberg, Gerard D; Johnson, Matthew E; Pahl, Matthew C; Pippin, James A; Wells, Andrew D; Anderson, Stewart A; Brown, Christopher D; Grant, Struan F A; Chesi, Alessandra.
Afiliación
  • Burton EA; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Argenziano M; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Cook K; CAMB Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
  • Ridler M; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Lu S; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Su C; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Manduchi E; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Littleton SH; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Leonard ME; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Hodge KM; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Wang LS; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Schellenberg GD; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Johnson ME; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Pahl MC; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Pippin JA; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Wells AD; CAMB Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
  • Anderson SA; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Brown CD; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
  • Grant SFA; Emory University, Atlanta, GA, USA.
  • Chesi A; Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
bioRxiv ; 2024 Aug 22.
Article en En | MEDLINE | ID: mdl-39229212
ABSTRACT
Late-onset Alzheimer's disease (LOAD) research has principally focused on neurons over the years due to their known role in the production of amyloid beta plaques and neurofibrillary tangles. In contrast, recent genomic studies of LOAD have implicated microglia as culprits of the prolonged inflammation exacerbating the neurodegeneration observed in patient brains. Indeed, recent LOAD genome-wide association studies (GWAS) have reported multiple loci near genes related to microglial function, including TREM2, ABI3, and CR1. However, GWAS alone cannot pinpoint underlying causal variants or effector genes at such loci, as most signals reside in non-coding regions of the genome and could presumably confer their influence frequently via long-range regulatory interactions. We elected to carry out a combination of ATAC-seq and high-resolution promoter-focused Capture-C in two human microglial cell models (iPSC-derived microglia and HMC3) in order to physically map interactions between LOAD GWAS-implicated candidate causal variants and their corresponding putative effector genes. Notably, we observed consistent evidence that rs6024870 at the GWAS CASS4 locus contacted the promoter of nearby gene, RTFDC1. We subsequently observed a directionallly consistent decrease in RTFDC1 expression with the the protective minor A allele of rs6024870 via both luciferase assays in HMC3 cells and expression studies in primary human microglia. Through CRISPR-Cas9-mediated deletion of the putative regulatory region harboring rs6024870 in HMC3 cells, we observed increased pro-inflammatory cytokine secretion and decreased DNA double strand break repair related, at least in part, to RTFDC1 expression levels. Our variant-to-function approach therefore reveals that the rs6024870-harboring regulatory element at the LOAD 'CASS4' GWAS locus influences both microglial inflammatory capacity and DNA damage resolution, along with cumulative evidence implicating RTFDC1 as a novel candidate effector gene.

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: BioRxiv Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: BioRxiv Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos
...