PLoS Genetics (May 2013)

Human genetics in rheumatoid arthritis guides a high-throughput drug screen of the CD40 signaling pathway.

  • Gang Li,
  • Dorothée Diogo,
  • Di Wu,
  • Jim Spoonamore,
  • Vlado Dancik,
  • Lude Franke,
  • Fina Kurreeman,
  • Elizabeth J Rossin,
  • Grant Duclos,
  • Cathy Hartland,
  • Xuezhong Zhou,
  • Kejie Li,
  • Jun Liu,
  • Philip L De Jager,
  • Katherine A Siminovitch,
  • Alexandra Zhernakova,
  • Soumya Raychaudhuri,
  • John Bowes,
  • Steve Eyre,
  • Leonid Padyukov,
  • Peter K Gregersen,
  • Jane Worthington,
  • Rheumatoid Arthritis Consortium International (RACI),
  • Namrata Gupta,
  • Paul A Clemons,
  • Eli Stahl,
  • Nicola Tolliday,
  • Robert M Plenge

DOI
https://doi.org/10.1371/journal.pgen.1003487
Journal volume & issue
Vol. 9, no. 5
p. e1003487

Abstract

Read online

Although genetic and non-genetic studies in mouse and human implicate the CD40 pathway in rheumatoid arthritis (RA), there are no approved drugs that inhibit CD40 signaling for clinical care in RA or any other disease. Here, we sought to understand the biological consequences of a CD40 risk variant in RA discovered by a previous genome-wide association study (GWAS) and to perform a high-throughput drug screen for modulators of CD40 signaling based on human genetic findings. First, we fine-map the CD40 risk locus in 7,222 seropositive RA patients and 15,870 controls, together with deep sequencing of CD40 coding exons in 500 RA cases and 650 controls, to identify a single SNP that explains the entire signal of association (rs4810485, P = 1.4×10(-9)). Second, we demonstrate that subjects homozygous for the RA risk allele have ∼33% more CD40 on the surface of primary human CD19+ B lymphocytes than subjects homozygous for the non-risk allele (P = 10(-9)), a finding corroborated by expression quantitative trait loci (eQTL) analysis in peripheral blood mononuclear cells from 1,469 healthy control individuals. Third, we use retroviral shRNA infection to perturb the amount of CD40 on the surface of a human B lymphocyte cell line (BL2) and observe a direct correlation between amount of CD40 protein and phosphorylation of RelA (p65), a subunit of the NF-κB transcription factor. Finally, we develop a high-throughput NF-κB luciferase reporter assay in BL2 cells activated with trimerized CD40 ligand (tCD40L) and conduct an HTS of 1,982 chemical compounds and FDA-approved drugs. After a series of counter-screens and testing in primary human CD19+ B cells, we identify 2 novel chemical inhibitors not previously implicated in inflammation or CD40-mediated NF-κB signaling. Our study demonstrates proof-of-concept that human genetics can be used to guide the development of phenotype-based, high-throughput small-molecule screens to identify potential novel therapies in complex traits such as RA.