2012
DOI: 10.1038/emboj.2012.215
|View full text |Cite
|
Sign up to set email alerts
|

Human CST promotes telomere duplex replication and general replication restart after fork stalling

Abstract: Mammalian CST (CTC1-STN1-TEN1) associates with telomeres and depletion of CTC1 or STN1 causes telomere defects. However, the function of mammalian CST remains poorly understood. We show here that depletion of CST subunits leads to both telomeric and non-telomeric phenotypes associated with DNA replication defects. Stable knockdown of CTC1 or STN1 increases the incidence of anaphase bridges and multi-telomeric signals, indicating genomic and telomeric instability. STN1 knockdown also delays replication through … Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1

Citation Types

28
332
0
2

Year Published

2013
2013
2024
2024

Publication Types

Select...
5
2

Relationship

1
6

Authors

Journals

citations
Cited by 192 publications
(368 citation statements)
references
References 68 publications
28
332
0
2
Order By: Relevance
“…Elsewhere in the genome, STN1 and CTC1 participate in the resolution of replication stress (10,29). Depletion of either subunit leads to formation of anaphase bridges, and STN1 depletion results in reduced replication restart after hydroxyurea (HU)-induced fork stalling (10,11,29). Consistent with this genome-wide role in replication, CTC1 and STN1 were originally identified as a pol ␣ accessory factor (AAF) that stimulates template binding and enzyme processivity (31,32).…”
mentioning
confidence: 82%
See 3 more Smart Citations
“…Elsewhere in the genome, STN1 and CTC1 participate in the resolution of replication stress (10,29). Depletion of either subunit leads to formation of anaphase bridges, and STN1 depletion results in reduced replication restart after hydroxyurea (HU)-induced fork stalling (10,11,29). Consistent with this genome-wide role in replication, CTC1 and STN1 were originally identified as a pol ␣ accessory factor (AAF) that stimulates template binding and enzyme processivity (31,32).…”
mentioning
confidence: 82%
“…Primers for TEN1 amplification, 5Ј-GGCCAAGTTCCT-GATGGG and 5Ј-CAGTGTTACTCTGGACTGAATCAT, were designed to avoid amplification of a TEN1 pseudogene. GAPDH and STN1 primers were as described (10). Reactions were performed at least in duplicate using GAPDH as the endogenous control.…”
Section: Methodsmentioning
confidence: 99%
See 2 more Smart Citations
“…CST: DNA Pol‐α‐mediated C‐strand fill‐in is absolutely required for telomere length maintenance, as telomerase by itself is insufficient to generate the proper duplex telomere (Feng et al., 2017; Gu et al., 2012). Defects in telomere replication due to disruption of the CST complex lead to replication fork stalling, as telomeres can adopt secondary structures that are difficult to replicate (Gu et al., 2012; Stewart et al., 2012). Stalled replication forks and the failure of stalled fork restart at telomeres initiate aberrant homologous recombination events that in part account for the catastrophic loss of total telomeric DNA observed in mouse cells devoid of CTC1 (Gu et al., 2012), or the activation of a DDR in mammalian cells (Wang et al., 2012).…”
Section: Introductionmentioning
confidence: 99%