Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Inhibition of telomerase RNA decay rescues telomerase deficiency caused by dyskerin or PARN defects

Subjects

Abstract

Mutations in the human telomerase RNA component (hTR), the telomerase ribonucleoprotein component dyskerin (DKC1) and the poly(A) RNase (PARN) can lead to reduced levels of hTR and to dyskeratosis congenita (DC). However, the enzymes and mechanisms responsible for hTR degradation are unknown. We demonstrate that defects in dyskerin binding lead to hTR degradation by PAPD5-mediated oligoadenylation, which promotes 3′-to-5′ degradation by EXOSC10, as well as decapping and 5′-to-3′ decay by the cytoplasmic DCP2 and XRN1 enzymes. PARN increased hTR levels by deadenylating hTR, thereby limiting its degradation by EXOSC10. Telomerase activity and proper hTR localization in dyskerin- or PARN-deficient cells were rescued by knockdown of DCP2 and/or EXOSC10. Prevention of hTR RNA decay also led to a rescue of localization of DC-associated hTR mutants. These results suggest that inhibition of RNA decay pathways might be a useful therapy for some telomere pathologies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Lack of dyskerin binding reduces hTR levels through two different RNA decay mechanisms.
Figure 2: PARN knockdown reduces hTR levels, owing to competing activity of EXOSC10.
Figure 3: Rescue of telomerase activity in dyskerin- or PARN-depleted HeLa cells by coknockdown of competing nucleases.
Figure 4: Mislocalization of hTR in dyskerin- or PARN-knockdown HeLa cells can be corrected by knockdown of competing nuclease.
Figure 5: Mutant hTR localization to Cajal bodies can be rescued by knockdown of RNA decay pathways.
Figure 6

Similar content being viewed by others

References

  1. Blackburn, E.H. & Collins, K. Telomerase: an RNP enzyme synthesizes DNA. Cold Spring Harb. Perspect. Biol. 3, a003558 (2011).

    Article  Google Scholar 

  2. Armanios, M. & Blackburn, E.H. The telomere syndromes. Nat. Rev. Genet. 13, 693–704 (2012).

    Article  CAS  Google Scholar 

  3. Heiss, N.S. et al. X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions. Nat. Genet. 19, 32–38 (1998).

    Article  CAS  Google Scholar 

  4. Vulliamy, T. et al. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature 413, 432–435 (2001).

    Article  CAS  Google Scholar 

  5. Mitchell, J.R., Wood, E. & Collins, K. A telomerase component is defective in the human disease dyskeratosis congenita. Nature 402, 551–555 (1999).

    Article  CAS  Google Scholar 

  6. Vulliamy, T.J. & Dokal, I. Dyskeratosis congenita: the diverse clinical presentation of mutations in the telomerase complex. Biochimie 90, 122–130 (2008).

    Article  CAS  Google Scholar 

  7. Stuart, B.D. et al. Exome sequencing links mutations in PARN and RTEL1 with familial pulmonary fibrosis and telomere shortening. Nat. Genet. 47, 512–517 (2015).

    Article  CAS  Google Scholar 

  8. Dhanraj, S. et al. Bone marrow failure and developmental delay caused by mutations in poly(A)-specific ribonuclease (PARN). J. Med. Genet. 52, 738–748 (2015).

    Article  CAS  Google Scholar 

  9. Tummala, H. et al. Poly(A)-specific ribonuclease deficiency impacts telomere biology and causes dyskeratosis congenita. J. Clin. Invest. 125, 2151–2160 (2015).

    Article  Google Scholar 

  10. Rammelt, C., Bilen, B., Zavolan, M. & Keller, W. PAPD5, a noncanonical poly(A) polymerase with an unusual RNA-binding motif. RNA 17, 1737–1746 (2011).

    Article  CAS  Google Scholar 

  11. Vanácová, S. et al. A new yeast poly(A) polymerase complex involved in RNA quality control. PLoS Biol. 3, e189 (2005).

    Article  Google Scholar 

  12. Wyers, F. et al. Cryptic pol II transcripts are degraded by a nuclear quality control pathway involving a new poly(A) polymerase. Cell 121, 725–737 (2005).

    Article  CAS  Google Scholar 

  13. Kadaba, S. et al. Nuclear surveillance and degradation of hypomodified initiator tRNAMet in S. cerevisiae. Genes Dev. 18, 1227–1240 (2004).

    Article  CAS  Google Scholar 

  14. LaCava, J. et al. RNA degradation by the exosome is promoted by a nuclear polyadenylation complex. Cell 121, 713–724 (2005).

    Article  CAS  Google Scholar 

  15. Goldfarb, K.C. & Cech, T.R. 3′ terminal diversity of MRP RNA and other human noncoding RNAs revealed by deep sequencing. BMC Mol. Biol. 14, 23 (2013).

    Article  CAS  Google Scholar 

  16. Fu, D. & Collins, K. Distinct biogenesis pathways for human telomerase RNA and H/ACA small nucleolar RNAs. Mol. Cell 11, 1361–1372 (2003).

    Article  CAS  Google Scholar 

  17. Ueda, Y. et al. A mutation in the H/ACA box of telomerase RNA component gene (TERC) in a young patient with myelodysplastic syndrome. BMC Med. Genet. 15, 68 (2014).

    Article  Google Scholar 

  18. Cohen, S.B. & Reddel, R.R. A sensitive direct human telomerase activity assay. Nat. Methods 5, 355–360 (2008).

    Article  CAS  Google Scholar 

  19. Zhu, Y., Tomlinson, R.L., Lukowiak, A.A., Terns, R.M. & Terns, M.P. Telomerase RNA accumulates in Cajal bodies in human cancer cells. Mol. Biol. Cell 15, 81–90 (2004).

    Article  CAS  Google Scholar 

  20. Tomlinson, R.L., Ziegler, T.D., Supakorndej, T., Terns, R.M. & Terns, M.P. Cell cycle-regulated trafficking of human telomerase to telomeres. Mol. Biol. Cell 17, 955–965 (2006).

    Article  CAS  Google Scholar 

  21. Venteicher, A.S. et al. A human telomerase holoenzyme protein required for Cajal body localization and telomere synthesis. Science 323, 644–648 (2009).

    Article  CAS  Google Scholar 

  22. Zhong, F.L. et al. TPP1 OB-fold domain controls telomere maintenance by recruiting telomerase to chromosome ends. Cell 150, 481–494 (2012).

    Article  CAS  Google Scholar 

  23. Tseng, C.-K.K. Human telomerase RNA processing and quality control. Cell Reports 13, 2232–2243 (2015).

    Article  CAS  Google Scholar 

  24. Nguyen, D. et al. A polyadenylation-dependent 3′ end maturation pathway is required for the synthesis of the human telomerase RNA. Cell Rep, 13, 2244–2257 (2015).

    Article  CAS  Google Scholar 

  25. Moon, D.H. et al. Poly(A)-specific ribonuclease (PARN) mediates 3′-end maturation of the telomerase RNA component. Nat. Genet. 47, 1482–1488 (2015).

    Article  CAS  Google Scholar 

  26. Shukla, S. & Parker, R. Quality control of assembly-defective U1 snRNAs by decapping and 5′-to-3′ exonucleolytic digestion. Proc. Natl. Acad. Sci. USA 111, E3277–E3286 (2014).

    Article  CAS  Google Scholar 

  27. Teixeira, M.T., Forstemann, K., Gasser, S.M. & Lingner, J. Intracellular trafficking of yeast telomerase components. EMBO Rep. 3, 652–659 (2002).

    Article  CAS  Google Scholar 

  28. Gallardo, F., Olivier, C., Dandjinou, A.T., Wellinger, R.J. & Chartrand, P. TLC1 RNA nucleo-cytoplasmic trafficking links telomerase biogenesis to its recruitment to telomeres. EMBO J. 27, 748–757 (2008).

    Article  CAS  Google Scholar 

  29. Xi, L. & Cech, T.R. Inventory of telomerase components in human cells reveals multiple subpopulations of hTR and hTERT. Nucleic Acids Res. 42, 8565–8577 (2014).

    Article  CAS  Google Scholar 

  30. Abreu, E., Terns, R.M. & Terns, M.P. Visualization of human telomerase localization by fluorescence microscopy techniques. Methods Mol. Biol. 735, 125–137 (2011).

    Article  CAS  Google Scholar 

  31. Schmidt, J.C., Dalby, A.B. & Cech, T.R. Identification of human TERT elements necessary for telomerase recruitment to telomeres. eLife 3, e03563 (2014).

    Article  Google Scholar 

  32. Stern, J.L., Theodorescu, D., Vogelstein, B., Papadopoulos, N. & Cech, T.R. Mutation of the TERT promoter, switch to active chromatin, and monoallelic TERT expression in multiple cancers. Genes Dev. 29, 2219–2224 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank A. Zaug for his assistance with the telomerase direct activity assays; A. Webb for help with the illustrations and figures in this manuscript; S. Spencer (University of Colorado Boulder), J. Lingner (Institut Suisse de Recherche Expérimentale sur le Cancer) and S.B. Cohen (Children's Medical Research Institute) for providing materials; and members of the Parker and Cech laboratories for their comments and suggestions. J.C.S. is supported as a Merck fellow of the Damon Runyon Cancer Research Foundation (DRG-2169-13). This work was supported by US National Institutes of Health grants R01 GM45443 (R.P.) and GM099705 (T.R.C.). T.R.C. and R.P. are supported as investigators of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

S.S., J.C.S., T.R.C. and R.P. conceptualized and designed the experiments. S.S. performed the experiments. J.C.S. performed the Pol II ChIP for hTR. K.C.G. and S.S. analyzed the hTR 3′-end sequencing reads. S.S., J.C.S., T.R.C. and R.P. analyzed and interpreted data. S.S., J.C.S., T.R.C. and R.P. wrote the manuscript.

Corresponding author

Correspondence to Roy Parker.

Ethics declarations

Competing interests

T.R.C. is on the board of directors of Merck, Inc. The other authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Quality-control pathways for A377G and C408G mutant hTR are conserved in U2OS cells.

Knockdown of specific nucleases rescues levels of A377G and C408G hTR in U2OS cells. a, Western blot for knockdown of various nucleases in U2OS cells. b, Northern blot for A377G hTR levels upon knockdown of DCP2, EXOSC10 or XRN1 (mean+/-s.d., n=4 independent experiments). c, Northern blot for C408G hTR levels upon PAPD5 knockdown with or without a co-knockdown of DCP2 or XRN1 (mean+/-s.d., n=3 independent experiments). d, Northern blot for C408G hTR upon co-knockdown of EXOSC10 and PAPD5 (mean+/-s.d., n=4 independent experiments). e, Northern blot for hTR upon knockdown of various decapping enzymes in U2OS cells (mean+/-s.d., n=4 independent experiments). f, Normalized Pol II occupancy by ChIP for the wild-type and C408G hTR plasmid-borne copy in U2OS cells (wild-type arbitrarily set to 1 and C408G normalized to wild-type).

Supplementary Figure 2 hTR 3′ mature end exhibits longer A tails after PARN knockdown.

Length distribution of oligo(A) tails at the mature 3’ end of hTR under different transfection conditions. Relative amount of each two or more A-containing reads was calculated to the total number of two or more A’s containing hTR reads under each transfection condition (P<0.05 by one tail unpaired Student’s t test).

Supplementary Figure 3 Mutant hTR localizes to cyTER bodies in U2OS cells.

Mutant A377G or C408G hTR is mislocalized to the cytoplasm in cyTER bodies in U2OS cells. a, Cytoplasmic localization of A377G or C408G RNA (white arrowheads) by FISH in U2OS cells. The nucleus is labeled with an antibody that recognizes either of the nuclear envelope components lamin A or lamin C (Scale bar 5 μm). White numbers in Merge, % of cells showing hTR localization to Cajal bodies (mean+/-s.d., n=4 independent experiments). b, Cytoplasmic localization of C408G hTR by FISH (nucleus outlined with lamin A/C staining, cell outlined with pan cadherin staining) (Scale bar 5 μm). White numbers in Merge, % of cells showing hTR cytoplasmic localization (mean+/-s.d., n=4 independent experiments).

Supplementary Figure 4 FISH signal for WT and mutant hTR is specific and does not localize to P bodies or U bodies.

cyTER bodies are novel sites of hTR storage. a, Subcellular localization of WT or C408G hTR (white arrowhead) by FISH using different combinations of Alexa Fluor labeled probes (Scale bar 5 μm). b, Subcellular localization of C408G RNA puncta (white arrowhead) in U2OS cells. Other foci stained were U-bodies or Gems (SMN) or P-bodies (DCP1) (Scale bar 10μm).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 (PDF 643 kb)

Supplementary Data Set 1

Uncropped blots and autoradiograph images (PDF 11407 kb)

Supplementary Table 1

Raw data for hTR 3′ end sequencing under different conditions (XLSX 18 kb)

Supplementary Table 2

Compiled number of reads for hTR 3′ end sequencing (XLSX 18 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shukla, S., Schmidt, J., Goldfarb, K. et al. Inhibition of telomerase RNA decay rescues telomerase deficiency caused by dyskerin or PARN defects. Nat Struct Mol Biol 23, 286–292 (2016). https://doi.org/10.1038/nsmb.3184

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3184

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing