4.7 Article

Posttranscriptional modulation of TERC by PAPD5 inhibition rescues hematopoietic development in dyskeratosis congenita

Journal

BLOOD
Volume 133, Issue 12, Pages 1308-1312

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/blood-2018-11-885368

Keywords

-

Categories

Funding

  1. National Institutes of Health, National Heart, Lung, and Blood Institute T32 training grant in molecular hematology [HL007088]
  2. Philip Majerus Fellowship Fund
  3. National Science Foundation
  4. Howard Hughes Medical Institute
  5. National Institutes of Health, National Institute of General Medical Sciences [R01GM45443]
  6. American Society of Hematology Scholar Award
  7. National Institutes of Health, National Heart, Lung, and Blood Institute [4R00HL114732, 1R01HL137793]
  8. V Foundation for Cancer Research
  9. Edward Mallinckrodt Jr Foundation
  10. AA&MDS International Foundation
  11. CONCERN Foundation
  12. American Federation for Aging Research
  13. Longer Life Foundation
  14. Center for Regenerative Medicine at Washington University in St. Louis
  15. Department of Defense Bone Marrow Failure Research Program [BM160054]
  16. CDMRP [BM160054, 917355] Funding Source: Federal RePORTER

Ask authors/readers for more resources

Reduced levels of TERC, the telomerase RNA component, cause dyskeratosis congenita (DC) in patients harboring mutations in TERC, PARN, NOP10, NHP2, NAF1, or DKC1. Inhibition of the noncanonical poly(A) polymerase PAPD5, or the exosome RNA degradation complex, partially restores TERC levels in immortalized DKC1 mutant cells, but it remains unknown if modulation of posttranscriptional processing of TERC could improve hematopoietic output in DC. We used human embryonic stem cells (hESCs) with a common dyskerin mutation (DKC1_A353V), which have defective telomere maintenance and reduced definitive hematopoietic potential, to understand the effects of reducing EXOSC3 activity, or silencing PAPD5-mediated oligoadenylation, on hematopoietic progenitor specification and function in DC. Reduction of EXOSC3 or PAPD5 levels in DKC1 mutant hESCs led to functional improvements in TERC levels and telomerase activity, with concomitant telomere elongation and reduced levels of DNA damage signaling. Interestingly, the silencing of PAPD5, but not EXOSC3, significantly restored definitive hematopoietic potential in DKC1 mutant cells. Mechanistically, we show that PAPD5 inhibition is sustained in differentiated CD34(+) cells, with a concomitant increase in mature, functional, forms of TERC, indicating that regulation of PAPD5 is a potential strategy to reverse hematologic dysfunction in DC patients.

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