Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 3038, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37263999

ABSTRACT

Telomeres are environment-sensitive regulators of health and aging. Here,we present telomere DNA length analysis of two reef-building coral genera revealing that the long- and short-term water thermal regime is a key driver of between-colony variation across the Pacific Ocean. Notably, there are differences between the two studied genera. The telomere DNA lengths of the short-lived, more stress-sensitive Pocillopora spp. colonies were largely determined by seasonal temperature variation, whereas those of the long-lived, more stress-resistant Porites spp. colonies were insensitive to seasonal patterns, but rather influenced by past thermal anomalies. These results reveal marked differences in telomere DNA length regulation between two evolutionary distant coral genera exhibiting specific life-history traits. We propose that environmentally regulated mechanisms of telomere maintenance are linked to organismal performances, a matter of paramount importance considering the effects of climate change on health.


Subject(s)
Anthozoa , Animals , Anthozoa/genetics , Coral Reefs , Temperature , Seasons , DNA/genetics
2.
Genetics ; 223(3)2023 03 02.
Article in English | MEDLINE | ID: mdl-36563016

ABSTRACT

Telomeres are ribonucleoproteins that cap chromosome-ends and their DNA length is controlled by counteracting elongation and shortening processes. The budding yeast Saccharomyces cerevisiae has been a leading model to study telomere DNA length control and dynamics. Its telomeric DNA is maintained at a length that slightly varies between laboratory strains, but little is known about its variation at the species level. The recent publication of the genomes of over 1,000 S. cerevisiae strains enabled us to explore telomere DNA length variation at an unprecedented scale. Here, we developed a bioinformatic pipeline (YeaISTY) to estimate telomere DNA length from whole-genome sequences and applied it to the sequenced S. cerevisiae collection. Our results revealed broad natural telomere DNA length variation among the isolates. Notably, telomere DNA length is shorter in those derived from wild rather than domesticated environments. Moreover, telomere DNA length variation is associated with mitochondrial metabolism, and this association is driven by wild strains. Overall, these findings reveal broad variation in budding yeast's telomere DNA length regulation, which might be shaped by its different ecological life-styles.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/genetics , Telomere/metabolism , Saccharomyces cerevisiae Proteins/genetics , Telomere-Binding Proteins/genetics , Base Sequence
3.
Cells ; 10(7)2021 07 11.
Article in English | MEDLINE | ID: mdl-34359923

ABSTRACT

Protecting telomere from the DNA damage response is essential to avoid the entry into cellular senescence and organismal aging. The progressive telomere DNA shortening in dividing somatic cells, programmed during development, leads to critically short telomeres that trigger replicative senescence and thereby contribute to aging. In several organisms, including mammals, telomeres are protected by a protein complex named Shelterin that counteract at various levels the DNA damage response at chromosome ends through the specific function of each of its subunits. The changes in Shelterin structure and function during development and aging is thus an intense area of research. Here, we review our knowledge on the existence of several Shelterin subcomplexes and the functional independence between them. This leads us to discuss the possibility that the multifunctionality of the Shelterin complex is determined by the formation of different subcomplexes whose composition may change during aging.


Subject(s)
Telomere-Binding Proteins/metabolism , Telomere/metabolism , Animals , DNA/metabolism , DNA Replication , Humans , Models, Molecular , Protein Structure, Quaternary , Telomere-Binding Proteins/chemistry
4.
Sci Rep ; 10(1): 19937, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203910

ABSTRACT

Aging is a multifactorial process that results in progressive loss of regenerative capacity and tissue function while simultaneously favoring the development of a large array of age-related diseases. Evidence suggests that the accumulation of senescent cells in tissue promotes both normal and pathological aging. Oxic stress is a key driver of cellular senescence. Because symbiotic long-lived reef corals experience daily hyperoxic and hypoxic transitions, we hypothesized that these long-lived animals have developed specific longevity strategies in response to light. We analyzed transcriptome variation in the reef coral Stylophora pistillata during the day-night cycle and revealed a signature of the FoxO longevity pathway. We confirmed this pathway by immunofluorescence using antibodies against coral FoxO to demonstrate its nuclear translocation. Through qPCR analysis of nycthemeral variations of candidate genes under different light regimens, we found that, among genes that were specifically up- or downregulated upon exposure to light, human orthologs of two "light-up" genes (HEY1 and LONF3) exhibited anti-senescence properties in primary human fibroblasts. Therefore, these genes are interesting candidates for counteracting skin aging. We propose a large screen for other light-up genes and an investigation of the biological response of reef corals to light (e.g., metabolic switching) to elucidate these processes and identify effective interventions for promoting healthy aging in humans.


Subject(s)
Anthozoa/physiology , Coral Reefs , Forkhead Transcription Factors/metabolism , Light , Longevity , Photosynthesis , Animals , Anthozoa/radiation effects , Forkhead Transcription Factors/genetics
5.
Nat Struct Mol Biol ; 27(4): 313-318, 2020 04.
Article in English | MEDLINE | ID: mdl-32231287

ABSTRACT

Telomeres arose from the need to stabilize natural chromosome ends, resulting in terminal chromatin structures with specific protective functions. Their constituent proteins also execute general functions within heterochromatin, mediating late replication and facilitating fork progression. Emerging insights into the mechanisms governing heterochromatin replication suggest telomeres and heterochromatin act in concert during development and aging. They also suggest a common evolutionary origin for these two chromosome regions that arose during eukaryogenesis.


Subject(s)
Chromatin/genetics , Heterochromatin/genetics , Proteins/genetics , Telomere/genetics , Chromatin/ultrastructure , DNA Replication/genetics , Heterochromatin/ultrastructure , Humans , Proteins/chemistry , Proteins/ultrastructure , Telomere/ultrastructure
6.
EMBO Rep ; 21(4): e49076, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32096305

ABSTRACT

Repressor/activator protein 1 (RAP1) is a highly evolutionarily conserved protein found at telomeres. Although yeast Rap1 is a key telomere capping protein preventing non-homologous end joining (NHEJ) and consequently telomere fusions, its role at mammalian telomeres in vivo is still controversial. Here, we demonstrate that RAP1 is required to protect telomeres in replicative senescent human cells. Downregulation of RAP1 in these cells, but not in young or dividing pre-senescent cells, leads to telomere uncapping and fusions. The anti-fusion effect of RAP1 was further explored in a HeLa cell line where RAP1 expression was depleted through an inducible CRISPR/Cas9 strategy. Depletion of RAP1 in these cells gives rise to telomere fusions only when telomerase is inhibited. We further show that the fusions triggered by RAP1 loss are dependent upon DNA ligase IV. We conclude that human RAP1 is specifically involved in protecting critically short telomeres. This has important implications for the functions of telomeres in senescent cells.


Subject(s)
Telomere , Transcription Factor AP-1 , Animals , Cellular Senescence/genetics , DNA Damage , HeLa Cells , Humans , Telomere/genetics , Telomere-Binding Proteins/genetics
7.
Mol Cell ; 70(3): 449-461.e5, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29727617

ABSTRACT

Hard-to-replicate regions of chromosomes (e.g., pericentromeres, centromeres, and telomeres) impede replication fork progression, eventually leading, in the event of replication stress, to chromosome fragility, aging, and cancer. Our knowledge of the mechanisms controlling the stability of these regions is essentially limited to telomeres, where fragility is counteracted by the shelterin proteins. Here we show that the shelterin subunit TRF2 ensures progression of the replication fork through pericentromeric heterochromatin, but not centromeric chromatin. In a process involving its N-terminal basic domain, TRF2 binds to pericentromeric Satellite III sequences during S phase, allowing the recruitment of the G-quadruplex-resolving helicase RTEL1 to facilitate fork progression. We also show that TRF2 is required for the stability of other heterochromatic regions localized throughout the genome, paving the way for future research on heterochromatic replication and its relationship with aging and cancer.


Subject(s)
DNA Replication/genetics , Genome/genetics , Heterochromatin/genetics , Telomere/genetics , Telomeric Repeat Binding Protein 2/genetics , Cell Line, Tumor , Centromere/genetics , Chromatin/genetics , DNA Helicases/genetics , G-Quadruplexes , HeLa Cells , Humans , S Phase/genetics
8.
Mol Cell ; 68(4): 643-644, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149592

ABSTRACT

In this issue of Molecular Cell, Kim et al. (2017) have studied the structure and organization of the shelterin protein complex protecting telomeres in Schizosaccharomyces pombe and humans and discovered an allosteric structural transition that drives the formation of the shelterin complex and participates in telomere length regulation.


Subject(s)
Telomere Homeostasis/physiology , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Humans , Schizosaccharomyces , Shelterin Complex , Telomere/genetics , Telomere-Binding Proteins/genetics
9.
Oncotarget ; 7(29): 46615-46627, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27366950

ABSTRACT

Telomere stability is a hallmark of immortalized cells, including cancer cells. While the telomere length is maintained in most cases by the telomerase, the activity of a protein complex called Shelterin is required to protect telomeres against unsuitable activation of the DNA damage response pathway. Within this complex, telomeric repeat binding factor 2 (TRF2) plays an essential role by blocking the ataxia telangiectasia-mutated protein (ATM) signaling pathway at telomeres and preventing chromosome end fusion. We showed that TRF2 was phosphorylated in vitro and in vivo on serine 323 by extracellular signal-regulated kinase (ERK1/2) in both normal and cancer cells. Moreover, TRF2 and activated ERK1/2 unexpectedly interacted in the cytoplasm of tumor cells and human tumor tissues. The expression of non-phosphorylatable forms of TRF2 in melanoma cells induced the DNA damage response, leading to growth arrest and tumor reversion. These findings revealed that the telomere stability is under direct control of one of the major pro-oncogenic signaling pathways (RAS/RAF/MEK/ERK) via TRF2 phosphorylation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/physiology , Telomeric Repeat Binding Protein 2/physiology , Animals , Apoptosis , Cell Line , Female , Humans , Mice , Phosphorylation , Telomere/physiology
10.
Nucleic Acids Res ; 44(4): 1962-76, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26748096

ABSTRACT

Telomere integrity is essential to maintain genome stability, and telomeric dysfunctions are associated with cancer and aging pathologies. In human, the shelterin complex binds TTAGGG DNA repeats and provides capping to chromosome ends. Within shelterin, RAP1 is recruited through its interaction with TRF2, and TRF2 is required for telomere protection through a network of nucleic acid and protein interactions. RAP1 is one of the most conserved shelterin proteins although one unresolved question is how its interaction may influence TRF2 properties and regulate its capacity to bind multiple proteins. Through a combination of biochemical, biophysical and structural approaches, we unveiled a unique mode of assembly between RAP1 and TRF2. The complete interaction scheme between the full-length proteins involves a complex biphasic interaction of RAP1 that directly affects the binding properties of the assembly. These results reveal how a non-DNA binding protein can influence the properties of a DNA-binding partner by mutual conformational adjustments.


Subject(s)
DNA-Binding Proteins/genetics , Genomic Instability , Telomere-Binding Proteins/genetics , Telomeric Repeat Binding Protein 2/genetics , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Humans , Multiprotein Complexes , Protein Binding , Shelterin Complex , Telomere/genetics , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 2/chemistry , Telomeric Repeat Binding Protein 2/metabolism
11.
Mol Cell ; 61(2): 274-86, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26774283

ABSTRACT

The shelterin proteins protect telomeres against activation of the DNA damage checkpoints and recombinational repair. We show here that a dimer of the shelterin subunit TRF2 wraps ∼ 90 bp of DNA through several lysine and arginine residues localized around its homodimerization domain. The expression of a wrapping-deficient TRF2 mutant, named Top-less, alters telomeric DNA topology, decreases the number of terminal loops (t-loops), and triggers the ATM checkpoint, while still protecting telomeres against non-homologous end joining (NHEJ). In Top-less cells, the protection against NHEJ is alleviated if the expression of the TRF2-interacting protein RAP1 is reduced. We conclude that a distinctive topological state of telomeric DNA, controlled by the TRF2-dependent DNA wrapping and linked to t-loop formation, inhibits both ATM activation and NHEJ. The presence of RAP1 at telomeres appears as a backup mechanism to prevent NHEJ when topology-mediated telomere protection is impaired.


Subject(s)
DNA/chemistry , Nucleic Acid Conformation , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Base Pairing , DNA/metabolism , DNA Damage , DNA End-Joining Repair , HeLa Cells , Humans , Lysine/metabolism , Models, Molecular , Mutation , Protein Structure, Tertiary , Shelterin Complex , Signal Transduction , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 2/chemistry
12.
Nucleic Acids Res ; 43(12): 5824-37, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-25999344

ABSTRACT

The ends of eukaryotic chromosomes need to be protected from the activation of a DNA damage response that leads the cell to replicative senescence or apoptosis. In mammals, protection is accomplished by a six-factor complex named shelterin, which organizes the terminal TTAGGG repeats in a still ill-defined structure, the telomere. The stable interaction of shelterin with telomeres mainly depends on the binding of two of its components, TRF1 and TRF2, to double-stranded telomeric repeats. Tethering of TRF proteins to telomeres occurs in a chromatin environment characterized by a very compact nucleosomal organization. In this work we show that binding of TRF1 and TRF2 to telomeric sequences is modulated by the histone octamer. By means of in vitro models, we found that TRF2 binding is strongly hampered by the presence of telomeric nucleosomes, whereas TRF1 binds efficiently to telomeric DNA in a nucleosomal context and is able to remodel telomeric nucleosomal arrays. Our results indicate that the different behavior of TRF proteins partly depends on the interaction with histone tails of their divergent N-terminal domains. We propose that the interplay between the histone octamer and TRF proteins plays a role in the steps leading to telomere deprotection.


Subject(s)
Nucleosomes/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Binding Sites , DNA/metabolism , Histones/metabolism , Nucleosomes/chemistry , Protein Binding , Protein Structure, Tertiary , Repetitive Sequences, Nucleic Acid , Telomere/chemistry , Telomeric Repeat Binding Protein 1/chemistry , Telomeric Repeat Binding Protein 2/chemistry
13.
Cell Rep ; 9(3): 1047-60, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25437559

ABSTRACT

Telomeric repeat binding factor 2 (TRF2), which plays a central role in telomere capping, is frequently increased in human tumors. We reveal here that TRF2 is expressed in the vasculature of most human cancer types, where it colocalizes with the Wilms' tumor suppressor WT1. We further show that TRF2 is a transcriptional target of WT1 and is required for proliferation, migration, and tube formation of endothelial cells. These angiogenic effects of TRF2 are uncoupled from its function in telomere capping. Instead, TRF2 binds and transactivates the promoter of the angiogenic tyrosine kinase platelet-derived growth factor receptor ß (PDGFRß). These findings reveal an unexpected role of TRF2 in neoangiogenesis and delineate a distinct function of TRF2 as a transcriptional regulator.


Subject(s)
Neovascularization, Pathologic/genetics , Promoter Regions, Genetic , Receptor, Platelet-Derived Growth Factor beta/genetics , Telomeric Repeat Binding Protein 2/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , DNA Damage , DNA Repair , Gene Knockout Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred C57BL , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Protein Binding , Telomere/metabolism , Up-Regulation/genetics , WT1 Proteins/metabolism
14.
Cell Cycle ; 13(15): 2469-74, 2014.
Article in English | MEDLINE | ID: mdl-25483196

ABSTRACT

The stability of mammalian telomeres depends upon TRF2, which prevents inappropriate repair and checkpoint activation. By using a plasmid integration assay in yeasts carrying humanized telomeres, we demonstrated that TRF2 possesses the intrinsic property to both stimulate initial homologous recombination events and to prevent their resolution via its basic N-terminal domain. In human cells, we further showed that this TRF2 domain prevents telomere shortening mediated by the resolvase-associated protein SLX4 as well as GEN1 and MUS81, 2 different types of endonucleases with resolvase activities. We propose that various types of resolvase activities are kept in check by the basic N-terminal domain of TRF2 in order to favor an accurate repair of the stalled forks that occur during telomere replication.


Subject(s)
Recombination, Genetic , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , HEK293 Cells , Holliday Junction Resolvases/metabolism , Humans , Plasmids , Recombinases/metabolism , Telomere Homeostasis , Telomeric Repeat Binding Protein 2/genetics , Transfection , Tumor Suppressor Protein p53/metabolism
15.
Front Oncol ; 3: 48, 2013.
Article in English | MEDLINE | ID: mdl-23509004

ABSTRACT

A major issue in telomere research is to understand how the integrity of chromosome ends is controlled. The fact that different types of nucleoprotein complexes have been described at the telomeres of different organisms raises the question of whether they have in common a structural identity that explains their role in chromosome protection. We will review here how telomeric nucleoprotein complexes are structured, comparing different organisms and trying to link these structures to telomere biology. It emerges that telomeres are formed by a complex and specific network of interactions between DNA, RNA, and proteins. The fact that these interactions and associated activities are reinforcing each other might help to guarantee the robustness of telomeric functions across the cell cycle and in the event of cellular perturbations. We will also discuss the recent notion that telomeres have evolved specific systems to overcome the DNA topological stress generated during their replication and transcription. This will lead to revisit the way we envisage the functioning of telomeric complexes since the regulation of topology is central to DNA stability, replication, recombination, and transcription as well as to chromosome higher-order organization.

16.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 3): 409-19, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23519416

ABSTRACT

Repressor activator protein 1 (Rap1) is an essential factor involved in transcription and telomere stability in the budding yeast Saccharomyces cerevisiae. Its interaction with DNA causes hypersensitivity to potassium permanganate, suggesting local DNA melting and/or distortion. In this study, various Rap1-DNA crystal forms were obtained using specifically designed crystal screens. Analysis of the DNA conformation showed that its distortion was not sufficient to explain the permanganate reactivity. However, anomalous data collected at the Mn edge using a Rap1-DNA crystal soaked in potassium permanganate solution indicated that the DNA conformation in the crystal was compatible with interaction with permanganate ions. Sequence-conservation analysis revealed that double-Myb-containing Rap1 proteins all carry a fully conserved Arg580 at a position that may favour interaction with permanganate ions, although it is not involved in the hypersensitive cytosine distortion. Permanganate reactivity assays with wild-type Rap1 and the Rap1[R580A] mutant demonstrated that Arg580 is essential for hypersensitivity. AFM experiments showed that wild-type Rap1 and the Rap1[R580A] mutant interact with DNA over 16 successive binding sites, leading to local DNA stiffening but not to accumulation of the observed local distortion. Therefore, Rap1 may cause permanganate hypersensitivity of DNA by forming a pocket between the reactive cytosine and Arg580, driving the permanganate ion towards the C5-C6 bond of the cytosine.


Subject(s)
DNA, Fungal/chemistry , DNA, Fungal/metabolism , Potassium Permanganate/chemistry , Potassium Permanganate/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Telomere-Binding Proteins/chemistry , Transcription Factors/chemistry , Arginine/chemistry , Crystallography, X-Ray , Cytosine/chemistry , DNA, Fungal/drug effects , Hydrogen Bonding/drug effects , Nucleic Acid Conformation/drug effects , Protein Binding/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Shelterin Complex , Solutions , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
17.
PLoS One ; 7(4): e34386, 2012.
Article in English | MEDLINE | ID: mdl-22536324

ABSTRACT

Mammalian telomeres stabilize chromosome ends as a result of their assembly into a peculiar form of chromatin comprising a complex of non-histone proteins named shelterin. TRF2, one of the shelterin components, binds to the duplex part of telomeric DNA and is essential to fold the telomeric chromatin into a protective cap. Although most of the human telomeric DNA is organized into tightly spaced nucleosomes, their role in telomere protection and how they interplay with telomere-specific factors in telomere organization is still unclear. In this study we investigated whether TRF2 can regulate nucleosome assembly at telomeres.By means of chromatin immunoprecipitation (ChIP) and Micrococcal Nuclease (MNase) mapping assay, we found that the density of telomeric nucleosomes in human cells was inversely proportional to the dosage of TRF2 at telomeres. This effect was not observed in the G1 phase of the cell cycle but appeared coincident of late or post-replicative events. Moreover, we showed that TRF2 overexpression altered nucleosome spacing at telomeres increasing internucleosomal distance. By means of an in vitro nucleosome assembly system containing purified histones and remodeling factors, we reproduced the short nucleosome spacing found in telomeric chromatin. Importantly, when in vitro assembly was performed in the presence of purified TRF2, nucleosome spacing on a telomeric DNA template increased, in agreement with in vivo MNase mapping.Our results demonstrate that TRF2 negatively regulates the number of nucleosomes at human telomeres by a cell cycle-dependent mechanism that alters internucleosomal distance. These findings raise the intriguing possibility that telomere protection is mediated, at least in part, by the TRF2-dependent regulation of nucleosome organization.


Subject(s)
Cell Cycle Checkpoints , Nucleosomes/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/physiology , Cell Line, Tumor , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Gene Expression , Humans , Protein Binding , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism
18.
Nucleic Acids Res ; 40(6): 2566-76, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22139926

ABSTRACT

TRF1 and TRF2 are key proteins in human telomeres, which, despite their similarities, have different behaviors upon DNA binding. Previous work has shown that unlike TRF1, TRF2 condenses telomeric, thus creating consequential negative torsion on the adjacent DNA, a property that is thought to lead to the stimulation of single-strand invasion and was proposed to favor telomeric DNA looping. In this report, we show that these activities, originating from the central TRFH domain of TRF2, are also displayed by the TRFH domain of TRF1 but are repressed in the full-length protein by the presence of an acidic domain at the N-terminus. Strikingly, a similar repression is observed on TRF2 through the binding of a TERRA-like RNA molecule to the N-terminus of TRF2. Phylogenetic and biochemical studies suggest that the N-terminal domains of TRF proteins originate from a gradual extension of the coding sequences of a duplicated ancestral gene with a consequential progressive alteration of the biochemical properties of these proteins. Overall, these data suggest that the N-termini of TRF1 and TRF2 have evolved to finely regulate their ability to condense DNA.


Subject(s)
Telomere/chemistry , Telomeric Repeat Binding Protein 1/chemistry , Telomeric Repeat Binding Protein 2/chemistry , Amino Acid Sequence , DNA/chemistry , DNA/metabolism , Evolution, Molecular , Humans , Molecular Sequence Data , Protein Structure, Tertiary , RNA/metabolism , Sequence Homology, Amino Acid , Telomere/metabolism , Telomeric Repeat Binding Protein 1/metabolism
19.
Mol Cell ; 39(5): 665-76, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20832719

ABSTRACT

Telomere protection in budding yeast requires the heterotrimer named CST (for Cdc13-Stn1-Ten1). Recent data show that CST components are conserved and required for telomere stability in a wide range of eukaryotes, even those utilizing the shelterin complex to protect their telomeres. A common function of these proteins might be to stimulate priming at the C-strand gap that remains after telomerase elongation, replication termination, and terminal processing. In light of the budding yeast situation, another conserved function of CST might well be the regulation of telomerase. The cohabitation at telomeres of CST and shelterin components highlights the complexity of telomere biology.


Subject(s)
Cyclin B/metabolism , DNA Replication/physiology , DNA, Fungal/metabolism , Saccharomycetales/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Cyclin B/genetics , DNA, Fungal/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Saccharomycetales/genetics , Schizosaccharomyces pombe Proteins/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere-Binding Proteins/genetics
20.
FEBS Lett ; 584(17): 3785-99, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20696167

ABSTRACT

A major issue in telomere research is to understand how the integrity of chromosome ends is controlled. Although several nucleoprotein complexes have been described at the telomeres of different organisms, it is still unclear how they confer a structural identity to chromosome ends in order to mask them from DNA repair and to ensure their proper replication. In this review, we describe how telomeric nucleoprotein complexes are structured, comparing different organisms and trying to link these structures to telomere biology. It emerges that telomeres are formed by a complex and specific network of interactions between DNA, RNA and proteins. The fact that these interactions and associated activities are reinforcing each other might help to guaranty the robustness of telomeric functions across the cell cycle and in the event of cellular perturbations. We propose that telomeric nucleoprotein complexes orient cell fate through dynamic transitions in their structures and their organization.


Subject(s)
Telomere/chemistry , Telomere/genetics , Animals , DNA/genetics , DNA Repair/genetics , Genetic Variation , Humans , Mammals , Oxytricha/genetics , Proteins/genetics , RNA/genetics , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/genetics , Tandem Repeat Sequences , Telomere/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...