Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Nucleic Acids Res ; 51(13): 6702-6722, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37246640

ABSTRACT

Telomeres are the nucleoprotein structures at the ends of linear chromosomes. Telomeres are transcribed into long non-coding Telomeric Repeat-Containing RNA (TERRA), whose functions rely on its ability to associate with telomeric chromatin. The conserved THO complex (THOC) was previously identified at human telomeres. It links transcription with RNA processing, decreasing the accumulation of co-transcriptional DNA:RNA hybrids throughout the genome. Here, we explore the role of THOC at human telomeres, as a regulator of TERRA localization to chromosome ends. We show that THOC counteracts TERRA association with telomeres via R-loops formed co-transcriptionally and also post-transcriptionally, in trans. We demonstrate that THOC binds nucleoplasmic TERRA, and that RNaseH1 loss, which increases telomeric R-loops, promotes THOC occupancy at telomeres. Additionally, we show that THOC counteracts lagging and mainly leading strand telomere fragility, suggesting that TERRA R-loops can interfere with replication fork progression. Finally, we observed that THOC suppresses telomeric sister-chromatid exchange and C-circle accumulation in ALT cancer cells, which maintain telomeres by recombination. Altogether, our findings reveal crucial roles of THOC in telomeric homeostasis through the co- and post-transcriptional regulation of TERRA R-loops.


Subject(s)
RNA, Long Noncoding , Telomerase , Humans , Telomerase/genetics , Telomerase/metabolism , R-Loop Structures , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis , RNA, Long Noncoding/genetics , Recombination, Genetic
2.
Genes Dev ; 36(17-18): 951-953, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36347559

ABSTRACT

Although telomeres are essential for chromosome stability, they represent fragile structures in our genome. Telomere shortening occurs during aging in cells lacking telomerase due to the end replication problem. In addition, recent work uncovered that the bulk of telomeric DNA poses severe hurdles for the semiconservative DNA replication machinery, requiring the assistance of an increasing number of specialized factors that prevent accidental telomere loss or damage events. In this issue of Genes & Development, Yang and colleagues (pp. 956-969) discover that TFIIH, a basic component of the PolII transcription initiation and nucleotide excision repair machinery, facilitates telomere replication. TFIIH is recruited to telomeres by the shelterin component TRF1, taking on at telomeres a moonlighting function.


Subject(s)
Telomerase , Telomeric Repeat Binding Protein 1 , Telomere/genetics , Telomere/metabolism , Telomere Shortening , Telomere-Binding Proteins/metabolism , Telomerase/metabolism , Shelterin Complex
3.
Methods Mol Biol ; 2528: 159-171, 2022.
Article in English | MEDLINE | ID: mdl-35704191

ABSTRACT

R-loops are three-stranded nucleic acid structures composed of a DNA-RNA hybrid and a displaced DNA strand. The long noncoding RNA TERRA forms R-loops at telomeres influencing the telomeric chromatin composition and impacting on telomere maintenance mechanisms by semiconservative DNA replication, homology directed DNA repair and telomerase. Here, we describe a method to detect R-loops at telomeres, which involves immunoprecipitation with the R-loop recognizing S9.6 antibody, followed by detection of telomeric DNA by either dot-blot hybridization with a radiolabeled telomeric probe, or qPCR using DNA primers that are specific for subtelomeric sequences.


Subject(s)
R-Loop Structures , RNA, Long Noncoding , DNA/chemistry , Humans , Nucleic Acid Hybridization , RNA, Long Noncoding/genetics , Telomere/genetics
4.
Nucleic Acids Res ; 49(21): 12119-12135, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34747482

ABSTRACT

Telomere shortening can cause detrimental diseases and contribute to aging. It occurs due to the end replication problem in cells lacking telomerase. Furthermore, recent studies revealed that telomere shortening can be attributed to difficulties of the semi-conservative DNA replication machinery to replicate the bulk of telomeric DNA repeats. To investigate telomere replication in a comprehensive manner, we develop QTIP-iPOND - Quantitative Telomeric chromatin Isolation Protocol followed by isolation of Proteins On Nascent DNA - which enables purification of proteins that associate with telomeres specifically during replication. In addition to the core replisome, we identify a large number of proteins that specifically associate with telomere replication forks. Depletion of several of these proteins induces telomere fragility validating their importance for telomere replication. We also find that at telomere replication forks the single strand telomere binding protein POT1 is depleted, whereas histone H1 is enriched. Our work reveals the dynamic changes of the telomeric proteome during replication, providing a valuable resource of telomere replication proteins. To our knowledge, this is the first study that examines the replisome at a specific region of the genome.


Subject(s)
DNA Replication , Telomerase/metabolism , Telomere/metabolism , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , Shelterin Complex/metabolism , Telomere Shortening , Telomere-Binding Proteins/metabolism
5.
Bioessays ; 43(10): e2100157, 2021 10.
Article in English | MEDLINE | ID: mdl-34436787

ABSTRACT

It has become apparent that difficulties to replicate telomeres concern not only the very ends of eukaryotic chromosomes. The challenges already start when the replication fork enters the telomeric repeats. The obstacles encountered consist mainly of noncanonical nucleic acid structures that interfere with replication if not resolved. Replication stress at telomeres promotes the formation of so-called fragile telomeres displaying an abnormal appearance in metaphase chromosomes though their exact molecular nature remains to be elucidated. A substantial number of factors is required to counteract fragility. In this review we promote the hypothesis that telomere fragility is not caused directly by an initial insult during replication but it results as a secondary consequence of DNA repair of damaged replication forks by the homologous DNA recombination machinery. Incomplete DNA synthesis at repair sites or partial chromatin condensation may become apparent as telomere fragility. Fragility and DNA repair during telomere replication emerges as a common phenomenon which exacerbates in multiple disease conditions.


Subject(s)
DNA Replication , Telomere , Chromatin/genetics , DNA Repair/genetics , DNA Replication/genetics , Homologous Recombination , Telomere/genetics
6.
Cell Cycle ; 20(18): 1745-1759, 2021 09.
Article in English | MEDLINE | ID: mdl-34432566

ABSTRACT

Telomeres protect chromosome ends from nucleolytic degradation, uncontrolled recombination by DNA repair enzymes and checkpoint signaling, and they provide mechanisms for their maintenance by semiconservative DNA replication, telomerase and homologous recombination. The telomeric long noncoding RNA TERRA is transcribed from a large number of chromosome ends. TERRA has been implicated in modulating telomeric chromatin structure and checkpoint signaling, and in telomere maintenance by homology directed repair, and telomerase - when telomeres are damaged or very short. Recent work indicates that TERRA association with telomeres involves the formation of DNA:RNA hybrid structures that can be formed post transcription by the RAD51 DNA recombinase, which in turn may trigger homologous recombination between telomeric repeats and telomere elongation. In this review, we describe the mechanisms of TERRA recruitment to telomeres, R-loop formation and its regulation by shelterin proteins. We discuss the consequences of R-loop formation, with regard to telomere maintenance by DNA recombination and how this may impinge on telomere replication while counteracting telomere shortening in normal cells and in ALT cancer cells, which maintain telomeres in the absence of telomerase.


Subject(s)
Neoplasms/metabolism , R-Loop Structures , RNA, Long Noncoding/metabolism , Repetitive Sequences, Nucleic Acid , Shelterin Complex/metabolism , Signal Transduction , Telomere Homeostasis , Transcription, Genetic , Animals , Cell Cycle , DNA/metabolism , Humans , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Telomerase/metabolism , Telomere Shortening
7.
Int J Mol Sci ; 22(5)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673424

ABSTRACT

Telomerase negative cancer cell types use the Alternative Lengthening of Telomeres (ALT) pathway to elongate telomeres ends. Here, we show that silencing human DNA polymerase (Pol λ) in ALT cells represses ALT activity and induces telomeric stress. In addition, replication stress in the absence of Pol λ, strongly affects the survival of ALT cells. In vitro, Pol λ can promote annealing of even a single G-rich telomeric repeat to its complementary strand and use it to prime DNA synthesis. The noncoding telomeric repeat containing RNA TERRA and replication protein A negatively regulate this activity, while the Protection of Telomeres protein 1 (POT1)/TPP1 heterodimer stimulates Pol λ. Pol λ associates with telomeres and colocalizes with TPP1 in cells. In summary, our data suggest a role of Pol λ in the maintenance of telomeres by the ALT mechanism.


Subject(s)
Aminopeptidases/metabolism , DNA Polymerase beta/metabolism , G-Quadruplexes , Serine Proteases/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/metabolism , Cell Line, Tumor , Humans , Multiprotein Complexes , Replication Protein A/metabolism , Shelterin Complex , Telomere/chemistry , Telomere/metabolism
8.
Cell Rep ; 33(5): 108347, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33147465

ABSTRACT

Telomeres are prone to damage inflicted by reactive oxygen species (ROS). Oxidized telomeric DNA and nucleotide substrates inhibit telomerase, causing telomere shortening. In addition, ROS can induce telomeric single-strand DNA breaks (SSBs). The peroxiredoxin-PRDX1 is enriched in telomeric chromatin and this counteracts ROS-induced telomere damage. Here, we identify DNA processing after oxidative stress as a main source of telomeric DNA cleavage events in the absence of PRDX1. In PRDX1-depleted cells, poly(ADP-ribose) polymerase (PARP)-dependent telomeric repair is often incomplete, giving persistent SSBs that are converted into telomeric double-strand breaks during replication, leading to rapid telomere shortening. Interestingly, PARP1 inhibition dampens telomere shortening, triggering stabilization of the homologous recombination (HR) factor BRCA1 and RAD51-mediated repair of telomeres. Overall, our results reveal that, in the absence PRDX1, incomplete PARP1-dependent DNA repair and competition between PARP1 and HR cause ROS-induced telomeric catastrophe.


Subject(s)
DNA Repair , Oxidative Stress , Peroxiredoxins/metabolism , Telomere/metabolism , BRCA1 Protein/metabolism , DNA Breaks, Single-Stranded , DNA Replication , HCT116 Cells , HeLa Cells , Humans , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Rad51 Recombinase/metabolism , Reactive Oxygen Species/metabolism
9.
EMBO J ; 39(23): e104500, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33073402

ABSTRACT

The evolutionarily conserved POT1 protein binds single-stranded G-rich telomeric DNA and has been implicated in contributing to telomeric DNA maintenance and the suppression of DNA damage checkpoint signaling. Here, we explore human POT1 function through genetics and proteomics, discovering that a complete absence of POT1 leads to severe telomere maintenance defects that had not been anticipated from previous depletion studies in human cells. Conditional deletion of POT1 in HEK293E cells gives rise to rapid telomere elongation and length heterogeneity, branched telomeric DNA structures, telomeric R-loops, and telomere fragility. We determine the telomeric proteome upon POT1-loss, implementing an improved telomeric chromatin isolation protocol. We identify a large set of proteins involved in nucleic acid metabolism that engage with telomeres upon POT1-loss. Inactivation of the homology-directed repair machinery suppresses POT1-loss-mediated telomeric DNA defects. Our results unravel as major function of human POT1 the suppression of telomere instability induced by homology-directed repair.


Subject(s)
Recombinational DNA Repair/genetics , Recombinational DNA Repair/physiology , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Cell Cycle/physiology , DNA/metabolism , DNA, Single-Stranded , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Phenotype , Proteome , Shelterin Complex , Transcriptome
10.
Nature ; 587(7833): 303-308, 2020 11.
Article in English | MEDLINE | ID: mdl-33057192

ABSTRACT

Telomeres-repeated, noncoding nucleotide motifs and associated proteins that are found at the ends of eukaryotic chromosomes-mediate genome stability and determine cellular lifespan1. Telomeric-repeat-containing RNA (TERRA) is a class of long noncoding RNAs (lncRNAs) that are transcribed from chromosome ends2,3; these RNAs in turn regulate telomeric chromatin structure and telomere maintenance through the telomere-extending enzyme telomerase4-6 and homology-directed DNA repair7,8. The mechanisms by which TERRA is recruited to chromosome ends remain poorly defined. Here we develop a reporter system with which to dissect the underlying mechanisms, and show that the UUAGGG repeats of TERRA are both necessary and sufficient to target TERRA to chromosome ends. TERRA preferentially associates with short telomeres through the formation of telomeric DNA-RNA hybrid (R-loop) structures that can form in trans. Telomere association and R-loop formation trigger telomere fragility and are promoted by the recombinase RAD51 and its interacting partner BRCA2, but counteracted by the RNA-surveillance factors RNaseH1 and TRF1. RAD51 physically interacts with TERRA and catalyses R-loop formation with TERRA in vitro, suggesting a direct involvement of this DNA recombinase in the recruitment of TERRA by strand invasion. Together, our findings reveal a RAD51-dependent pathway that governs TERRA-mediated R-loop formation after transcription, providing a mechanism for the recruitment of lncRNAs to new loci in trans.


Subject(s)
R-Loop Structures , RNA, Long Noncoding/chemistry , Rad51 Recombinase/metabolism , Telomere/chemistry , Telomere/metabolism , Base Sequence , Biocatalysis , Genes, Reporter , HeLa Cells , Humans , RNA, Long Noncoding/genetics , Ribonuclease H/metabolism , Telomere/genetics , Telomeric Repeat Binding Protein 1/metabolism
11.
EMBO J ; 39(7): e102668, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32080884

ABSTRACT

Structural maintenance of chromosomes flexible hinge domain-containing protein 1 (SMCHD1) has been implicated in X-chromosome inactivation, imprinting, and DNA damage repair, and mutations in SMCHD1 can cause facioscapulohumeral muscular dystrophy. More recently, SMCHD1 has also been identified as a component of telomeric chromatin. Here, we report that SMCHD1 is required for DNA damage signaling and non-homologous end joining (NHEJ) at unprotected telomeres. Co-depletion of SMCHD1 and the shelterin subunit TRF2 reduced telomeric 3'-overhang removal in time-course experiments, as well as the number of chromosome end fusions. SMCHD1-deficient cells displayed reduced ATM S1981 phosphorylation and diminished formation of γH2AX foci and of 53BP1-containing telomere dysfunction-induced foci (TIFs), indicating defects in DNA damage checkpoint signaling. Removal of TPP1 and subsequent activation of ATR signaling rescued telomere fusion events in TRF2-depleted SMCHD1 knockout cells. Together, these data indicate that SMCHD1 depletion reduces telomere fusions in TRF2-depleted cells due to defects in ATM-dependent checkpoint signaling and that SMCHD1 mediates DNA damage response activation upstream of ATM phosphorylation at uncapped telomeres.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/genetics , DNA Damage , DNA End-Joining Repair , Epistasis, Genetic , Gene Knockout Techniques , HeLa Cells , Humans , Phosphorylation , Shelterin Complex/genetics , Shelterin Complex/metabolism , Signal Transduction , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
12.
Nat Commun ; 10(1): 3884, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31467287

ABSTRACT

Genomic instability is a hallmark of cancer. Whether it also occurs in Cancer Associated Fibroblasts (CAFs) remains to be carefully investigated. Loss of CSL/RBP-Jκ, the effector of canonical NOTCH signaling with intrinsic transcription repressive function, causes conversion of dermal fibroblasts into CAFs. Here, we find that CSL down-modulation triggers DNA damage, telomere loss and chromosome end fusions that also occur in skin Squamous Cell Carcinoma (SCC)-associated CAFs, in which CSL is decreased. Separately from its role in transcription, we show that CSL is part of a multiprotein telomere protective complex, binding directly and with high affinity to telomeric DNA as well as to UPF1 and Ku70/Ku80 proteins and being required for their telomere association. Taken together, the findings point to a central role of CSL in telomere homeostasis with important implications for genomic instability of cancer stromal cells and beyond.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Squamous Cell/metabolism , Fibroblasts/metabolism , Genomic Instability , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Telomere/metabolism , Carcinoma, Squamous Cell/genetics , DNA Damage , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Homeostasis , Humans , Ku Autoantigen/metabolism , Membrane Proteins , Molecular Docking Simulation , Mutagenesis , RNA Helicases/metabolism , Receptors, Notch/metabolism , Signal Transduction , Skin/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Trans-Activators/metabolism
13.
RNA ; 25(11): 1470-1480, 2019 11.
Article in English | MEDLINE | ID: mdl-31350341

ABSTRACT

The telomeric long noncoding RNA TERRA has been implicated in regulating telomere maintenance by telomerase and homologous recombination, and in influencing telomeric protein composition during the cell cycle and the telomeric DNA damage response. TERRA transcription starts at subtelomeric regions resembling the CpG islands of eukaryotic genes extending toward chromosome ends. TERRA contains chromosome-specific subtelomeric sequences at its 5' end and long tracts of UUAGGG-repeats toward the 3' end. Conflicting studies have been published as to whether TERRA is expressed from one or several chromosome ends. Here, we quantify TERRA species by RT-qPCR in normal and several cancerous human cell lines. By using chromosome-specific subtelomeric DNA primers, we demonstrate that TERRA is expressed from a large number of telomeres. Deficiency in DNA methyltransferases leads to TERRA up-regulation only at the subset of chromosome ends that contain CpG-island sequences, revealing differential regulation of TERRA promoters by DNA methylation. However, independently of the differences in TERRA expression, short telomeres were uniformly present in a DNA methyltransferase deficient cell line, indicating that telomere length was not dictated by TERRA expression in cis Bioinformatic analyses indicated the presence of a large number of putative transcription factors binding sites at TERRA promoters, and we identified a subset of them that repress TERRA expression. Altogether, our study confirms that TERRA corresponds to a large gene family transcribed from multiple chromosome ends where we identified two types of TERRA promoters, only one of which is regulated by DNA methylation.


Subject(s)
Chromosomes, Human , DNA-Binding Proteins/genetics , Gene Expression Regulation , Telomere , Transcription Factors/genetics , CpG Islands , DNA Methylation , DNA Modification Methylases/metabolism , HCT116 Cells , Humans , Promoter Regions, Genetic , Up-Regulation
14.
Life Sci Alliance ; 1(4): e201800121, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30456372

ABSTRACT

Telomeres play crucial roles during tumorigenesis, inducing cellular senescence upon telomere shortening and extensive chromosome instability during telomere crisis. However, it has not been investigated if and how cellular transformation and oncogenic stress alter telomeric chromatin composition and function. Here, we transform human fibroblasts by consecutive transduction with vectors expressing hTERT, the SV40 early region, and activated H-RasV12. Pairwise comparisons of the telomeric proteome during different stages of transformation reveal up-regulation of proteins involved in chromatin remodeling, DNA repair, and replication at chromosome ends. Depletion of several of these proteins induces telomere fragility, indicating their roles in replication of telomeric DNA. Depletion of SAMHD1, which has reported roles in DNA resection and homology-directed repair, leads to telomere breakage events in cells deprived of the shelterin component TRF1. Thus, our analysis identifies factors, which accumulate at telomeres during cellular transformation to promote telomere replication and repair, resisting oncogene-borne telomere replication stress.

15.
Genes Dev ; 32(9-10): 658-669, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29773556

ABSTRACT

Telomerase counteracts telomere shortening and cellular senescence in germ, stem, and cancer cells by adding repetitive DNA sequences to the ends of chromosomes. Telomeres are susceptible to damage by reactive oxygen species (ROS), but the consequences of oxidation of telomeres on telomere length and the mechanisms that protect from ROS-mediated telomere damage are not well understood. In particular, 8-oxoguanine nucleotides at 3' ends of telomeric substrates inhibit telomerase in vitro, whereas, at internal positions, they suppress G-quadruplex formation and were therefore proposed to promote telomerase activity. Here, we disrupt the peroxiredoxin 1 (PRDX1) and 7,8-dihydro-8-oxoguanine triphosphatase (MTH1) genes in cancer cells and demonstrate that PRDX1 and MTH1 cooperate to prevent accumulation of oxidized guanine in the genome. Concomitant disruption of PRDX1 and MTH1 leads to ROS concentration-dependent continuous shortening of telomeres, which is due to efficient inhibition of telomere extension by telomerase. Our results identify antioxidant systems that are required to protect telomeres from oxidation and are necessary to allow telomere maintenance by telomerase conferring immortality to cancer cells.


Subject(s)
DNA Repair Enzymes/metabolism , Peroxiredoxins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Reactive Oxygen Species/metabolism , Telomerase/metabolism , Telomere Shortening/genetics , DNA Damage/genetics , DNA Repair Enzymes/genetics , Enzyme Activation/genetics , Gene Knockout Techniques , Genome , Guanine/metabolism , HCT116 Cells , Humans , Oxidation-Reduction , Oxidative Stress/genetics , Phosphoric Monoester Hydrolases/genetics , Telomerase/antagonists & inhibitors , Telomere Homeostasis/genetics
16.
Dev Cell ; 44(3): 281-283, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29408234

ABSTRACT

Telomerase counteracts telomere shortening, preventing cellular senescence. Telomerase deficiency causes telomere syndromes because of premature telomere exhaustion in highly proliferative cells. Paradoxically, in a recent issue of Cell, Margalef et al. (2018) demonstrate that telomerase causes telomere loss in cells lacking the RTEL1 helicase, which is defective in Hoyeraal-Hreidarsson syndrome (HHS).


Subject(s)
Telomerase/genetics , Telomere , Cellular Senescence , Dyskeratosis Congenita , Humans , Microcephaly , Mutation , Telomere Shortening
17.
Differentiation ; 99: 21-27, 2018.
Article in English | MEDLINE | ID: mdl-29274896

ABSTRACT

Telomere integrity is essential for genome stability and it regulates cell proliferation and tissue renewal. Several lines of evidence indicate that telomeres are particularly sensitive to oxidative damage. Moreover, recent studies demonstrate striking inhibitory effects of oxidative damage on telomerase activity. On the other hand, several mechanisms have been uncovered that either counteract oxidative damage at telomeres or remove the modified lesions. Here, we review the current understanding of oxidative damage and protection of telomeric DNA. We discuss how oxidative telomeric lesions impact on telomerase, the regenerative capacity of stem cells and cancer. Finally, we propose how through a better understanding of the involved pathways it may become possible to target telomerase in cancer cells in future cancer therapies.


Subject(s)
Oxidation-Reduction/drug effects , Oxidative Stress/genetics , Telomerase/genetics , Telomere/genetics , Animals , Humans , Neoplasms/genetics , Stem Cells/cytology , Telomere/metabolism
18.
Genes Dev ; 31(6): 567-577, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28381410

ABSTRACT

Telomeres are specialized nucleoprotein structures that protect chromosome ends from DNA damage response (DDR) and DNA rearrangements. The telomeric shelterin protein TRF2 suppresses the DDR, and this function has been attributed to its abilities to trigger t-loop formation or prevent massive decompaction and loss of density of telomeric chromatin. Here, we applied stochastic optical reconstruction microscopy (STORM) to measure the sizes and shapes of functional human telomeres of different lengths and dysfunctional telomeres that elicit a DDR. Telomeres have an ovoid appearance with considerable plasticity in shape. Examination of many telomeres demonstrated that depletion of TRF2, TRF1, or both affected the sizes of only a small subset of telomeres. Costaining of telomeres with DDR markers further revealed that the majority of DDR signaling telomeres retained a normal size. Thus, DDR signaling at telomeres does not require decompaction. We propose that telomeres are monitored by the DDR machinery in the absence of telomere expansion and that the DDR is triggered by changes at the molecular level in structure and protein composition.


Subject(s)
DNA Damage , Telomere/ultrastructure , Chromatin/physiology , Fluorescent Antibody Technique , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Telomeric Repeat Binding Protein 1/analysis , Telomeric Repeat Binding Protein 1/immunology , Telomeric Repeat Binding Protein 1/physiology , Telomeric Repeat Binding Protein 2/physiology
19.
Science ; 355(6325): 578-579, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28183935
20.
Methods ; 114: 39-45, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27530378

ABSTRACT

Telomeres, the heterochromatic structures that protect the ends of the chromosomes, are transcribed into a class of long non-coding RNAs, telomeric repeat-containing RNAs (TERRA), whose transcriptional regulation and functions are not well understood. The identification of TERRA adds a novel level of structural and functional complexity at telomeres, opening up a new field of research. TERRA molecules are expressed at several chromosome ends with transcription starting from the subtelomeric DNA proceeding into the telomeric tracts. TERRA is heterogeneous in length and its expression is regulated during the cell cycle and upon telomere damage. Little is known about the mechanisms of regulation at the level of transcription and post transcription by RNA stability. Furthermore, it remains to be determined to what extent the regulation at different chromosome ends may differ. We present an overview on the methodology of how RT-qPCR and primer pairs that are specific for different subtelomeric sequences can be used to detect and quantify TERRA expressed from different chromosome ends.


Subject(s)
RNA, Untranslated/genetics , Real-Time Polymerase Chain Reaction/methods , Repetitive Sequences, Nucleic Acid , Telomere , HeLa Cells , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...