2022
DOI: 10.1182/blood.2021010791
|View full text |Cite
|
Sign up to set email alerts
|

Inherited human Apollo deficiency causes severe bone marrow failure and developmental defects

Abstract: Inherited bone marrow failure syndromes (IBMFS) represent a group of disorders typified by impaired production of one or several blood cell types. The telomere biology disorders dyskeratosis congenita (DC) and its severe variant Høyeraal-Hreidarsson (HH) syndrome are rare IBMFS characterized by bone marrow failure, developmental defects, and various premature aging complications associated with critically short telomeres. Here we identified biallelic variants in the gene encoding the 5'-to-3' DNA exonuclease A… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

0
6
0

Year Published

2022
2022
2024
2024

Publication Types

Select...
6
1

Relationship

0
7

Authors

Journals

citations
Cited by 15 publications
(6 citation statements)
references
References 59 publications
0
6
0
Order By: Relevance
“…Here, we identified pathogenic variants in POT1 and DCLRE1B (alias symbol APOLLO) in two unusual severe gastritides thought to be of unknown etiology despite extensive clinical workup. Unlike other telomere complex genes which cause shortening of telomere length, these genes are associated with long and normal telomere length, respectively [7][8][9]. Data regarding involvement of POT1 and DCLRE1B genes in telomeropathies are only recently emerging with proposals that these genes should be included on cancer-risk predisposing multigene panel tests as well as genetic tests for telomere biology dysfunction [10,11].…”
Section: Discussionmentioning
confidence: 99%
“…Here, we identified pathogenic variants in POT1 and DCLRE1B (alias symbol APOLLO) in two unusual severe gastritides thought to be of unknown etiology despite extensive clinical workup. Unlike other telomere complex genes which cause shortening of telomere length, these genes are associated with long and normal telomere length, respectively [7][8][9]. Data regarding involvement of POT1 and DCLRE1B genes in telomeropathies are only recently emerging with proposals that these genes should be included on cancer-risk predisposing multigene panel tests as well as genetic tests for telomere biology dysfunction [10,11].…”
Section: Discussionmentioning
confidence: 99%
“…[39][40][41] Moreover, recent discoveries identified even candidate genes for patients with TBD features and normal TL (Apollo) or even increased TL (POT1). 10,42 Therefore, the decoding of previously unknown genomic alterations that interfere with telomere maintenance and telomeres/telomerases regulatory functions is of utmost importance to improve understanding of TBDs and close the gap of yet unknown TBD causes.…”
Section: Aq7mentioning
confidence: 99%
“… 7 However, genetic testing fails to properly diagnose an underlying TBD in at least 20% of patients, 8 simply because not all TBD-causing genes are known to date as exemplified by the very recent discovery of alterations in candidate genes such as Apollo or RPA1 . 9 , 10 In addition, the functional consequences of yet undefined variants found in TBD-genes can more accurately be determined once the in vivo read out, that is, shortened TL result, is available. In addition, rare families with cases of phenocopy (reviewed in Revy et al 11 ), that is, where screened relatives from yet unknown patients with genetic TBD have short telomeres despite of being genetically normal, will obviously be missed by next generation sequencing (NGS) screening alone.…”
Section: Introductionmentioning
confidence: 99%
“…In amniotes, the 5' exonuclease Apollo/DCLRE1B/SNM1B has evolved a YxLxP motif in its C-terminus that allows it to bind to the TRFH domain of TRF2 through the interaction with a region surrounding F120 [7][8][9][10][11] . TRF2-bound Apollo is thought to initiate 5' end resection at leading-end telomeres to allow subsequent long-range resection by Exo1 When Apollo is deleted or prevented from binding to TRF2 (e.g., when TRF2-F120A is used to complement deletion of TRF2), leading-end telomeres do not regain their normal 3' overhangs and are vulnerable to end joining [13][14][15][16][17] . Such leading-end telomere fusions are abolished by deletion of Ku70, which initially suggested that they are mediated by c-NHEJ 13,15 .…”
Section: Introductionmentioning
confidence: 99%
“…When Apollo is deleted or prevented from binding to TRF2 (e.g., when TRF2-F120A is used to complement deletion of TRF2), leading-end telomeres do not regain their normal 3' overhangs and are vulnerable to end joining [13][14][15][16][17] . Such leading-end telomere fusions are abolished by deletion of Ku70, which initially suggested that they are mediated by c-NHEJ 13,15 .…”
Section: Introductionmentioning
confidence: 99%