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











Database
Publication year range
1.
Cells ; 12(24)2023 12 08.
Article in English | MEDLINE | ID: mdl-38132118

ABSTRACT

Regulator of TElomere Length Helicase 1 (RTEL1) is a helicase required for telomere maintenance and genome replication and repair. RTEL1 has been previously shown to participate in the nuclear export of small nuclear RNAs. Here we show that RTEL1 deficiency leads to a nuclear envelope destabilization exclusively in cells entering S-phase and in direct connection to origin firing. We discovered that inhibiting protein import also leads to similar, albeit non-cell cycle-related, nuclear envelope disruptions. Remarkably, overexpression of wild-type RTEL1, or of its C-terminal part lacking the helicase domain, protects cells against nuclear envelope anomalies mediated by protein import inhibition. We identified distinct domains in the C-terminus of RTEL1 essential for the interaction with KPNB1 (importin ß) and NUP153, respectively, and we demonstrated that, on its own, the latter domain can promote the dynamic nuclear internalization of peptides that freely diffuse through the nuclear pore. Consistent with putative functions exerted in protein import, RTEL1 can be visualized on both sides of the nuclear pore using high-resolution microscopy. In all, our work points to an unanticipated, helicase-independent, role of RTEL1 in connecting both nucleocytoplasmic trafficking and nuclear envelope integrity to genome replication initiation in S-phase.


Subject(s)
Nuclear Envelope , beta Karyopherins , Humans , Active Transport, Cell Nucleus , Nuclear Envelope/metabolism , beta Karyopherins/metabolism , Nuclear Pore Complex Proteins/metabolism , DNA Replication , DNA Helicases/metabolism
2.
Hum Mutat ; 37(5): 469-72, 2016 May.
Article in English | MEDLINE | ID: mdl-26847928

ABSTRACT

The DNA helicase RTEL1 participates in telomere maintenance and genome stability. Biallelic mutations in the RTEL1 gene account for the severe telomere biology disorder characteristic of the Hoyeraal-Hreidarsson syndrome (HH). Here, we report a HH patient (P4) carrying two novel compound heterozygous mutations in RTEL1: a premature stop codon (c.949A>T, p.Lys317*) and an intronic deletion leading to an exon skipping and an in-frame deletion of 25 amino-acids (p.Ile398_Lys422). P4's cells exhibit short and dysfunctional telomeres similarly to other RTEL1-deficient patients. 3D structure predictions indicated that the p.Ile398_Lys422 deletion affects a part of the helicase ARCH domain, which lines the pore formed with the core HD and the iron-sulfur cluster domains and is highly specific of sequences from the eukaryotic XPD family members.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/genetics , Dyskeratosis Congenita/genetics , Fetal Growth Retardation/genetics , Intellectual Disability/genetics , Microcephaly/genetics , Mutation , Child , Codon, Terminator , Female , Humans , Models, Molecular , Protein Domains , Sequence Deletion
3.
Blood Adv ; 1(1): 36-46, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-29296694

ABSTRACT

Telomeres are repetitive hexameric sequences located at the end of linear chromosomes. They adopt a lariat-like structure, the T-loop, to prevent them from being recognized as DNA breaks by the DNA repair machinery. RTEL1 is a DNA helicase required for proper telomere replication and stability. In particular, it has been postulated that RTEL1 is involved in the opening of the T-loop during telomere replication to avoid sudden telomere deletion and telomere circle (T-circle) formation. In humans, biallelic RTEL1 mutations cause Hoyeraal-Hreidarsson syndrome (HH), a rare and severe telomere biology disorder characterized by intrauterine growth retardation, bone marrow failure, microcephaly and/or cerebellar hypoplasia, and immunodeficiency. To date, 18 different RTEL1 mutations have been described in 19 cases of HH with short telomeres. The impaired T-loop resolution has been proposed to be a major cause of telomere shortening in RTEL1 deficiency. However, the biological and clinical consequences of this disorder remain incompletely documented. Here, we describe 4 new patients harboring biallelic RTEL1 mutations, including 2 novel missense mutations located in the C-terminal end of RTEL1 (p.Cys1268Arg and p.Val1294Phe). Clinical characteristics from these 4 patients were collected as those from 4 other RTEL1-deficient patients previously reported. In addition, we assessed whether T-circles, the product of improper T-loop resolution, were detected in our RTEL1-deficient patients. Overall, our study broadens and refines the clinical and biological spectrum of human RTEL1 deficiency.

4.
Med Sci (Paris) ; 29(12): 1138-44, 2013 Dec.
Article in French | MEDLINE | ID: mdl-24356145

ABSTRACT

RTEL1 (regulator of telomere length helicase 1) is a DNA helicase that has been identified more than 10 years ago. Many works since, mainly in the nematode Caenorhabditis elegans and the mouse, have highlighted its role in chromosomal stability, maintenance of telomere length, and DNA repair. Recently, four laboratories have characterized RTEL1 mutations in patients with dyskeratosis congenita (DC) and Hoyeraal-Hreidarsson (HH) syndrome, a rare and severe variant of DC. We here summarize the current knowledge on RTEL1 and discuss the possible other functions that RTEL1 could play.


Subject(s)
DNA Helicases/physiology , Genomic Instability/physiology , Animals , DNA Helicases/genetics , DNA Repair , Dyskeratosis Congenita/genetics , Fetal Growth Retardation/genetics , Genetic Phenomena/genetics , Genetic Phenomena/physiology , Genomic Instability/genetics , Humans , Intellectual Disability/genetics , Mice , Microcephaly/genetics , Mutation , Telomere/physiology
5.
Hum Mol Genet ; 22(16): 3239-49, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23591994

ABSTRACT

Hoyeraal-Hreidarsson syndrome (HHS), a severe variant of dyskeratosis congenita (DC), is characterized by early onset bone marrow failure, immunodeficiency and developmental defects. Several factors involved in telomere length maintenance and/or protection are defective in HHS/DC, underlining the relationship between telomere dysfunction and these diseases. By combining whole-genome linkage analysis and exome sequencing, we identified compound heterozygous RTEL1 (regulator of telomere elongation helicase 1) mutations in three patients with HHS from two unrelated families. RTEL1 is a DNA helicase that participates in DNA replication, DNA repair and telomere integrity. We show that, in addition to short telomeres, RTEL1-deficient cells from patients exhibit hallmarks of genome instability, including spontaneous DNA damage, anaphase bridges and telomeric aberrations. Collectively, these results identify RTEL1 as a novel HHS-causing gene and highlight its role as a genomic caretaker in humans.


Subject(s)
DNA Helicases/genetics , Dyskeratosis Congenita/genetics , Fetal Growth Retardation/genetics , Genomic Instability , Intellectual Disability/genetics , Microcephaly/genetics , Telomere Homeostasis/genetics , Telomere Shortening , Telomere/metabolism , Cells, Cultured , Child, Preschool , DNA Damage , DNA Helicases/chemistry , DNA Helicases/deficiency , DNA Helicases/metabolism , DNA Replication , Dyskeratosis Congenita/metabolism , Exome , Female , Fetal Growth Retardation/metabolism , Genetic Linkage , Humans , Infant , Intellectual Disability/metabolism , Male , Microcephaly/metabolism , Mutation , Sequence Alignment , Sequence Analysis, RNA , Telomerase/genetics , Telomerase/metabolism , Telomere/ultrastructure
6.
Nucleic Acids Res ; 41(2): 900-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23193277

ABSTRACT

Telomeres cap the ends of chromosomes and regulate the replicative life span of human somatic cells. Telomere function is lost upon critical shortening and a p53-dependent checkpoint that detects altered telomere states at the G1/S transition was proposed to act as a regulator of the telomere damage response. We show that telomerase-negative human fibroblasts spend more time in G2 phase as they approach senescence and this delay is associated with manifestations of telomere dysfunction and the triggering of an ATM/ATR-dependent DNA damage signal. This correlates with a partial release of telomeric proteins TRF1 and TRF2. Analysis of the consequences of TRF1 and TRF2 depletion or over-expression of mutated versions revealed that telomere uncapping or telomere replication stress also led to DNA damage signalling in G2. Progression through mitosis of these cells was associated with signs of incomplete telomere terminal processing. We also observed an increase in sister chromatid-type telomere aberrations in senescing fibroblasts indicating that defects of telomere post-replicative events increased as cells age. Our results link a post-replicative damage response at eroded telomeres to G2 arrest signalling and challenge the current paradigm that the checkpoint response to short telomeres occurs primarily at the G1/S transition in human cells.


Subject(s)
G2 Phase Cell Cycle Checkpoints/genetics , Telomere Shortening , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cells, Cultured , Cellular Senescence , DNA Damage , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , G2 Phase , Humans , Mitosis , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
7.
J Cell Sci ; 123(Pt 8): 1295-305, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20332115

ABSTRACT

Mutations in the p53 tumour suppressor gene are associated clinically with tumour progression and metastasis. Downregulation of the E-cadherin cell-cell adhesion molecule is a key event for epithelial to mesenchymal transition (EMT) in tumour progression. Here, we show that wild-type p53 induced to adopt a mutant conformation, and hot-spot p53 mutants, which are both transcriptionally inactive, downregulate E-cadherin expression in the colon carcinoma cell line HCT116. Downregulation of E-cadherin occurred concomitantly with the upregulation of Slug and Zeb-1, transcriptional factors known to repress E-cadherin gene expression. In addition, knockdown of Slug and Zeb-1 expression diminished p53-mediated E-cadherin repression. Knocking down endogenous mutant p53 in MDA-MB-231 and SW620 cancer cell lines lacking E-cadherin protein restored the expression of E-cadherin. Complete loss of E-cadherin expression in HCT116 cells induced morphological alterations along with upregulation of vimentin, a mesenchymal marker. These changes characteristic of the EMT phenotype were, however, not sufficient to confer invasiveness in a three-dimensional matrix. Downregulation of E-cadherin by mutant p53 was not required to promote the invasive phenotype induced by inactivation of p53. These findings indicate that independent control of E-cadherin expression and cell motility could be essential molecular events in p53 mutant-induced invasive phenotypes.


Subject(s)
Cadherins/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Antigens, CD , Cadherins/metabolism , Cell Line, Tumor , Collagen/metabolism , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Down-Regulation/genetics , Drug Combinations , Epithelium/pathology , Humans , Laminin/metabolism , Mesoderm/pathology , Mutant Proteins/metabolism , Neoplasm Invasiveness , Phenotype , Proteoglycans/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Transcription, Genetic , Transcriptional Activation/genetics , Vimentin/genetics , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL