2019
DOI: 10.1101/746768
|View full text |Cite
Preprint
|
Sign up to set email alerts
|

Telomere Dysfunction Drives Chronic Lung Allograft Dysfunction Pathology

Abstract: Rationale:Telomere dysfunction is associated with multiple fibrotic lung processes, including chronic lung allograft dysfunction (CLAD) which is a major limitation to long-term survival following lung transplantation. Although shorter donor telomere lengths are associated with an increased risk of CLAD, it is unknown whether short telomeres are a cause or consequence of CLAD pathology. Objective:Our objective was to test whether telomere dysfunction contributes to pathologic changes seen in CLAD. Methods and R… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

1
3
0

Year Published

2020
2020
2021
2021

Publication Types

Select...
2
1

Relationship

0
3

Authors

Journals

citations
Cited by 3 publications
(4 citation statements)
references
References 49 publications
1
3
0
Order By: Relevance
“…This is in line with a previous study, which showed that bronchoalveolar lavage of fHP patients contained significantly higher carbonylated protein levels, a marker of ROS, compared to IPF and controls (55). However, another study showed that in mice with a Club cell-specific knock out of telomere repeat-binding factor-1 (Trf1), rapid aging of club cells by telomere dysfunction alone was sufficient to induce DNA damage and subsequent bronchiolocentric fibrosis (56). The excess DNA damage in fHP Club cells might therefore, regardless of the cause, be suggestive of an important role of these cells in fibrogenesis, and is in agreement with the localization of fibrosis in fHP.…”
Section: Dna Damage In Club Cells Of Fibrotic Lungssupporting
confidence: 91%
See 1 more Smart Citation
“…This is in line with a previous study, which showed that bronchoalveolar lavage of fHP patients contained significantly higher carbonylated protein levels, a marker of ROS, compared to IPF and controls (55). However, another study showed that in mice with a Club cell-specific knock out of telomere repeat-binding factor-1 (Trf1), rapid aging of club cells by telomere dysfunction alone was sufficient to induce DNA damage and subsequent bronchiolocentric fibrosis (56). The excess DNA damage in fHP Club cells might therefore, regardless of the cause, be suggestive of an important role of these cells in fibrogenesis, and is in agreement with the localization of fibrosis in fHP.…”
Section: Dna Damage In Club Cells Of Fibrotic Lungssupporting
confidence: 91%
“…The main function of surfactant is to prevent the collapse of alveoli by the reduction of the alveolar surface tension (52)(53)(54)(55). Furthermore, surfactant proteins are involved in innate host defense regulation through complement activation, thereby clearing away apoptotic cells and invading microorganisms (56,57). Even though all four subtypes of surfactant proteins are produced in the lung, to date only mutations in the SFTPA1/2 and SFTPC genes have been associated with FPF in adults (37,(58)(59)(60)(61)(62).…”
Section: Pathogenesismentioning
confidence: 99%
“…This is in line with a previous study that showed that bronchoalveolar lavage of fHP patients contained significantly higher carbonylated protein levels, a marker of ROS, compared to IPF and controls [ 47 ]. However, another study showed that in mice with a club cell-specific knockout of telomere repeat-binding factor-1 (Trf1), rapid ageing of club cells by telomere dysfunction alone was sufficient to induce DNA damage and subsequent bronchiolocentric fibrosis [ 25 ]. In addition, it was reported that in 25% of the cases with an initial diagnosis of IPF, bronchiolocentric fibrosis is indicative for a revised diagnosis of fHP [ 48 ].…”
Section: Discussionmentioning
confidence: 99%
“…However, if not fixed properly in time, the DNA damage response becomes persistent and eventually leads to cellular senescence [ 19 21 ], a process associated with fibrogenesis in IPF lungs and fibrotic mouse models [ 22 , 23 ]. Previous studies using mice models with a telomere repeat factor-1 (Trf1) knockout in alveolar or bronchiolar epithelial cells demonstrated telomere shortening and increased DNA damage foci in both cell types [ 24 , 25 ]. Similar results were found in TRF2-inactivated human cell lines showing an elevated amount of damage foci at uncapped telomeres [ 26 ].…”
Section: Introductionmentioning
confidence: 99%