Biogenesis of telomerase RNA in homeostasis and disease

Placeholder Show Content

Abstract/Contents

Abstract
Disruptions in RNA metabolism and errors in RNA processing are increasingly linked to a diverse set of human diseases. Telomerase, the ribonucleoprotein that extends telomeres to prevent replication-dependent chromosomal shortening, is composed of the reverse transcriptase TERT and the noncoding RNA (ncRNA) telomerase RNA component (hTR). Defects in hTR or in any of the protein cofactors required for hTR biogenesis precipitate telomere shortening and the stem cell disorder dyskeratosis congenita. Despite the critical importance of hTR in telomere homeostasis, the events surrounding hTR transcription, 3' end formation, and post-transcriptional regulation remain poorly understood. To interrogate the hTR biogenesis pathway in an unbiased kinetic fashion, we developed nascent 3' RNAend --Seq, which enabled us to isolate early hTR precursors and measure processing rates in vivo under different genetic conditions. We found that hTR is transcribed as an extended molecule with a short genomically encoded tail. These extended hTR precursors convert to the mature hTR species with delayed kinetics compared to related small ncRNAs. The hTR precursors can be oligoadenylated shortly after transcription by the poly(A)polymerase PAPD5 and can be deadenylated by the disease-associated enzyme Poly(A)ribonuclease (PARN). The relative activity of these two enzymes and the extent of of hTR precursors sets the maturation rate of hTR. In the absence of PARN, oligoadenylated hTR precursors stall and are degraded, causing overall hTR loss and telomere shortening. However, in the absence of both PARN and PAPD5, hTR precursor maturation is normalized and telomere lengths are maintained. We found that the H/ACA domain of hTR together with the RNA-binding protein dyskerin could direct formation of the hTR 3' end, recruit poly(A)polymerase activity, and confer sensitivity to regulation by oligoadenylation. A screen for additional enzymes involved in hTR 3' end formation revealed the DEDDh 3'-5' RNA exonuclease ISG20L2, which works together with PARN to promote maturation of the hTR 3' end and ensure accumulation of telomerase. These data reveal that mature hTR is generated through a feedforward circuit in which post-transcriptional oligoadenylation controls RNA maturation kinetics. Perturbations of hTR maturation rates read out into changes in steady-state telomerase levels and telomere length. Alterations in RNA maturation kinetics may prove to contribute to the mechanisms of other RNA based human diseases.

Description

Type of resource text
Form electronic resource; remote; computer; online resource
Extent 1 online resource.
Place California
Place [Stanford, California]
Publisher [Stanford University]
Copyright date 2021; ©2021
Publication date 2021; 2021
Issuance monographic
Language English

Creators/Contributors

Author Roake, Caitlin Marie
Degree supervisor Artandi, Steven E
Thesis advisor Artandi, Steven E
Thesis advisor Attardi, Laura
Thesis advisor Chua, Katrin Faye
Degree committee member Attardi, Laura
Degree committee member Chua, Katrin Faye
Associated with Stanford University, Cancer Biology Program

Subjects

Genre Theses
Genre Text

Bibliographic information

Statement of responsibility Caitlin Marie Roake.
Note Submitted to the Cancer Biology Program.
Thesis Thesis Ph.D. Stanford University 2021.
Location https://purl.stanford.edu/nz642bn7013

Access conditions

Copyright
© 2021 by Caitlin Marie Roake
License
This work is licensed under a Creative Commons Attribution Non Commercial 3.0 Unported license (CC BY-NC).

Also listed in

Loading usage metrics...