Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 11(1): 3321, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620872

ABSTRACT

Human telomeres are bound by the telomere repeat binding proteins TRF1 and TRF2. Telomere shortening in human cells leads to a DNA damage response that signals replicative senescence. While insufficient loading of TRF2 at shortened telomeres contributes to the DNA damage response in senescence, the contribution of TRF1 to senescence induction has not been determined. Here we show that counter to TRF2 deficiency-mediated induction of DNA damage, TRF1 deficiency serves a protective role to limit induction of DNA damage induced by subtelomere recombination. Shortened telomeres recruit insufficient TRF1 and as a consequence inadequate tankyrase 1 to resolve sister telomere cohesion. Our findings suggest that the persistent cohesion protects short telomeres from inappropriate recombination. Ultimately, in the final division, telomeres are no longer able to maintain cohesion and subtelomere copying ensues. Thus, the gradual loss of TRF1 and concomitant persistent cohesion that occurs with telomere shortening ensures a measured approach to replicative senescence.


Subject(s)
Telomere Shortening/genetics , Telomere/genetics , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 2/genetics , Base Sequence , Cell Line , Cell Line, Tumor , Cellular Senescence/genetics , DNA Damage , HEK293 Cells , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , In Situ Hybridization, Fluorescence , Mutation , RNA Interference , Tankyrases/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 1/deficiency , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/deficiency , Telomeric Repeat Binding Protein 2/metabolism
3.
Cell Rep ; 12(2): 286-99, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26146081

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a degenerative disease of the lungs with an average survival post-diagnosis of 2-3 years. New therapeutic targets and treatments are necessary. Mutations in components of the telomere-maintenance enzyme telomerase or in proteins important for telomere protection are found in both familial and sporadic IPF cases. However, the lack of mouse models that faithfully recapitulate the human disease has hampered new advances. Here, we generate two independent mouse models that develop IPF owing to either critically short telomeres (telomerase-deficient mice) or severe telomere dysfunction in the absence of telomere shortening (mice with Trf1 deletion in type II alveolar cells). We show that both mouse models develop pulmonary fibrosis through induction of telomere damage, thus providing proof of principle of the causal role of DNA damage stemming from dysfunctional telomeres in IPF development and identifying telomeres as promising targets for new treatments.


Subject(s)
Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Telomere/metabolism , Animals , Bleomycin/toxicity , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/drug effects , DNA Repair/drug effects , Disease Models, Animal , Female , Idiopathic Pulmonary Fibrosis/metabolism , Lung/diagnostic imaging , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiography , Tamoxifen/pharmacology , Telomerase/deficiency , Telomerase/genetics , Telomere Shortening , Telomeric Repeat Binding Protein 1/deficiency , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Tumor Suppressor Protein p53/metabolism
4.
Nat Commun ; 4: 1946, 2013.
Article in English | MEDLINE | ID: mdl-23735977

ABSTRACT

TRF1 is a component of the shelterin complex that protects chromosome ends. TRF1 deficiency leads to early embryonic lethality and to severe organ atrophy when deleted in adult tissues. Here we generate a reporter mouse carrying a knock-in eGFP-TRF1 fusion allele to study the role of TRF1 in stem cell biology and tissue homeostasis. We find that eGFP-TRF1 expression in mice is maximal in known adult stem cell compartments and show that TRF1 ensures their functionality. eGFP-TRF1 is highly expressed in induced pluripotent stem cells, uncoupled from the telomere elongation associated with reprogramming. Selection of eGFP-TRF1-high induced pluripotent stem cells correlates with higher pluripotency as indicated by their ability to form teratomas and chimeras. We further show that TRF1 is necessary for both induction and maintenance of pluripotency, and that TRF1 is a direct transcriptional target of Oct3/4.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Alleles , Animals , Atrophy , Biomarkers/metabolism , Cell Compartmentation , Cell Nucleus/metabolism , Cellular Reprogramming , Chromosomal Instability , Clone Cells , Fluorescence , Gene Deletion , Gene Knock-In Techniques , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Intestine, Small/pathology , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Recombinant Fusion Proteins/metabolism , Skin/cytology , Telomere/metabolism , Telomeric Repeat Binding Protein 1/deficiency , Telomeric Repeat Binding Protein 1/genetics
5.
J Cell Biol ; 197(2): 283-300, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22508511

ABSTRACT

TRF1 protects mammalian telomeres from fusion and fragility. Depletion of TRF1 leads to telomere fusions as well as accumulation of γ-H2AX foci and activation of both the ataxia telangiectasia mutated (ATM)- and the ataxia telangiectasia and Rad3 related (ATR)-mediated deoxyribonucleic acid (DNA) damage response (DDR) pathways. 53BP1, which is also present at dysfunctional telomeres, is a target of ATM that accumulates at DNA double-strand breaks and favors nonhomologous end-joining (NHEJ) repair over ATM-dependent resection and homology-directed repair (homologous recombination [HR]). To address the role of 53BP1 at dysfunctional telomeres, we generated mice lacking TRF1 and 53BP1. 53BP1 deficiency significantly rescued telomere fusions in mouse embryonic fibroblasts (MEFs) lacking TRF1, but they showed evidence of a switch from the NHEJ- to HR-mediated repair of uncapped telomeres. Concomitantly, double-mutant MEFs showed evidence of hyperactivation of the ATR-dependent DDR. In intact mice, combined 53BP1/TRF1 deficiency in stratified epithelia resulted in earlier onset of DNA damage and increased CHK1 phosphorylation during embryonic development, leading to aggravation of skin phenotypes.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Telomere/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Checkpoint Kinase 1 , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded , DNA Damage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Homologous Recombination , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Kinases/metabolism , Telomere/pathology , Telomeric Repeat Binding Protein 1/deficiency , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2 , Tumor Suppressor p53-Binding Protein 1
6.
Genes Dev ; 23(17): 2060-75, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19679647

ABSTRACT

The telomere repeat-binding factor 1 (TERF1, referred to hereafter as TRF1) is a component of mammalian telomeres whose role in telomere biology and disease has remained elusive. Here, we report on cells and mice conditionally deleted for TRF1. TRF1-deleted mouse embryonic fibroblasts (MEFs) show rapid induction of senescence, which is concomitant with abundant telomeric gamma-H2AX foci and activation of the ATM/ATR downstream checkpoint kinases CHK1 and CHK2. DNA damage foci are rescued by both ATM and ATM/ATR inhibitors, further indicating that both signaling pathways are activated upon TRF1 deletion. Abrogation of the p53 and RB pathways bypasses senescence but leads to chromosomal instability including sister chromatid fusions, chromosome concatenation, and occurrence of multitelomeric signals (MTS). MTS are also elevated in ATR-deficient MEFs or upon treatment with aphidicolin, two conditions known to induce breakage at fragile sites, suggesting that TRF1-depleted telomeres are prone to breakage. To address the impact of these molecular defects in the organism, we deleted TRF1 in stratified epithelia of TRF1(Delta/Delta)K5-Cre mice. These mice die perinatally and show skin hyperpigmentation and epithelial dysplasia, which are associated with induction of telomere-instigated DNA damage, activation of the p53/p21 and p16 pathways, and cell cycle arrest in vivo. p53 deficiency rescues mouse survival but leads to development of squamous cell carcinomas, demonstrating that TRF1 suppresses tumorigenesis. Together, these results demonstrate that dysfunction of a telomere-binding protein is sufficient to produce severe telomeric damage in the absence of telomere shortening, resulting in premature tissue degeneration and development of neoplastic lesions.


Subject(s)
Chromosome Fragility , Protein Deficiency/complications , Skin Diseases/etiology , Skin Neoplasms/etiology , Telomere/genetics , Telomeric Repeat Binding Protein 1/deficiency , Telomeric Repeat Binding Protein 1/metabolism , Aging/metabolism , Animals , Cell Cycle/physiology , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/genetics , E2F1 Transcription Factor/metabolism , Epidermal Cells , Epidermis/pathology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Hyperpigmentation/etiology , Hyperpigmentation/genetics , Mice , Mice, Knockout , Mutation/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Skin Diseases/genetics , Skin Neoplasms/genetics , Stem Cells/pathology , Telomeric Repeat Binding Protein 1/genetics , Tumor Suppressor Protein p53/metabolism
7.
Mol Biol Cell ; 20(10): 2563-71, 2009 May.
Article in English | MEDLINE | ID: mdl-19321665

ABSTRACT

The telomere end-protection complex prevents the ends of linear eukaryotic chromosomes from degradation or inappropriate DNA repair. The homodimeric double-stranded DNA-binding protein, Trf1, is a component of this complex and is essential for mouse embryonic development. To define the requirement for Trf1 in somatic cells, we deleted Trf1 in chicken DT40 cells by gene targeting. Trf1-deficient cells proliferated as rapidly as control cells and showed telomeric localization of Trf2, Rap1, and Pot1. Telomeric G-strand overhang lengths were increased in late-passage Trf1-deficient cells, although telomere lengths were unaffected by Trf1 deficiency, as determined by denaturing Southern and quantitative FISH analysis. Although we observed some clonal variation in terminal telomere fragment lengths, this did not correlate with cellular Trf1 levels. Trf1 was not required for telomere seeding, indicating that de novo telomere formation can proceed without Trf1. The Pin2 isoform and a novel exon 4, 5-deleted isoform localized to telomeres in Trf1-deficient cells. Trf1-deficient cells were sensitive to DNA damage induced by ionizing radiation. Our data demonstrate that chicken DT40 B cells do not require Trf1 for functional telomere structure and suggest that Trf1 may have additional, nontelomeric roles involved in maintaining genome stability.


Subject(s)
Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Animals , Cell Line , Cell Proliferation/radiation effects , Chickens , Gene Targeting , In Situ Hybridization, Fluorescence , Phenotype , RNA Splicing/radiation effects , Radiation, Ionizing , Telomerase/metabolism , Telomeric Repeat Binding Protein 1/deficiency , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...