4.7 Article

R-loops cause genomic instability in T helper lymphocytes from patients with Wiskott-Aldrich syndrome

Journal

JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY
Volume 142, Issue 1, Pages 219-234

Publisher

MOSBY-ELSEVIER
DOI: 10.1016/j.jaci.2017.11.023

Keywords

Primary immunodeficiency; Wiskott-Aldrich syndrome; X-linked thrombocytopenia; T(H)1/T(H)2 differentiation; R-loops; DNA damage; genomic instability

Funding

  1. National Institutes of Health (NIH) [R01AI084957]
  2. Intramural Research Program of the NIH, the National Institute on Aging grant [Z01AG000746-08]
  3. DeJoria Wiskott-Aldrich Research Fund
  4. NATIONAL CANCER INSTITUTE [P30CA086862] Funding Source: NIH RePORTER
  5. NATIONAL INSTITUTE OF ALLERGY AND INFECTIOUS DISEASES [R01AI084957] Funding Source: NIH RePORTER
  6. NATIONAL INSTITUTE ON AGING [ZIAAG000577, Z01AG000746, ZIAAG000746] Funding Source: NIH RePORTER

Ask authors/readers for more resources

Background: Wiskott-Aldrich syndrome (WAS), X-linked thrombocytopenia (XLT), and X-linked neutropenia, which are caused by WAS mutations affecting Wiskott-Aldrich syndrome protein (WASp) expression or activity, manifest in immunodeficiency, autoimmunity, genomic instability, and lymphoid and other cancers. WASp supports filamentous actin formation in the cytoplasm and gene transcription in the nucleus. Although the genetic basis for XLT/WAS has been clarified, the relationships between mutant forms of WASp and the diverse features of these disorders remain ill-defined. Objective: We sought to define how dysfunctional gene transcription is causally linked to the degree of T-H cell deficiency and genomic instability in the XLT/WAS clinical spectrum. Methods: In human T(H)1- or T(H)2-skewing cell culture systems, cotranscriptional R-loops (RNA/DNA duplex and displaced single-stranded DNA) and DNA double-strand breaks (DSBs) were monitored in multiple samples from patients with XLT and WAS and in normal T cells depleted of WASp. Results: WASp deficiency provokes increased R-loops and R-loop-mediated DSBs in T(H)1 cells relative to T(H)2 cells. Mechanistically, chromatin occupancy of serine 2-unphosphorylated RNA polymerase II is increased, and that of topoisomerase 1, an R-loop preventing factor, is decreased at R-loop-enriched regions of IFNG and TBX21 (T(H)1 genes) in T(H)1 cells. These aberrations accompany increased unspliced (intron-retained) and decreased spliced mRNA of IFNG and TBX21 but not IL13 (T(H)2 gene). Significantly, increased cellular load of R-loops and DSBs, which are normalized on RNaseH1-mediated suppression of ectopic R-loops, inversely correlates with disease severity scores. Conclusion: Transcriptional R-loop imbalance is a novel molecular defect causative in TH1 immunodeficiency and genomic instability in patients with WAS. The study proposes that cellular R-loop load could be used as a potential biomarker for monitoring symptom severity and prognostic outcome in the XLT-WAS clinical spectrum and could be targeted therapeutically.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available