Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.683
Filter
Add more filters

Publication year range
1.
Cell ; 187(15): 4030-4042.e13, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38908367

ABSTRACT

Insufficient telomerase activity, stemming from low telomerase reverse transcriptase (TERT) gene transcription, contributes to telomere dysfunction and aging pathologies. Besides its traditional function in telomere synthesis, TERT acts as a transcriptional co-regulator of genes pivotal in aging and age-associated diseases. Here, we report the identification of a TERT activator compound (TAC) that upregulates TERT transcription via the MEK/ERK/AP-1 cascade. In primary human cells and naturally aged mice, TAC-induced elevation of TERT levels promotes telomere synthesis, blunts tissue aging hallmarks with reduced cellular senescence and inflammatory cytokines, and silences p16INK4a expression via upregulation of DNMT3B-mediated promoter hypermethylation. In the brain, TAC alleviates neuroinflammation, increases neurotrophic factors, stimulates adult neurogenesis, and preserves cognitive function without evident toxicity, including cancer risk. Together, these findings underscore TERT's critical role in aging processes and provide preclinical proof of concept for physiological TERT activation as a strategy to mitigate multiple aging hallmarks and associated pathologies.


Subject(s)
Aging , DNA Methylation , Telomerase , Telomerase/metabolism , Telomerase/genetics , Humans , Animals , Mice , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Cellular Senescence , Promoter Regions, Genetic , DNA Methyltransferase 3B , Brain/metabolism , Telomere/metabolism , Mice, Inbred C57BL , Male , Transcription Factor AP-1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Neurogenesis
2.
Cell ; 187(14): 3638-3651.e18, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38838667

ABSTRACT

Telomere maintenance requires the extension of the G-rich telomeric repeat strand by telomerase and the fill-in synthesis of the C-rich strand by Polα/primase. At telomeres, Polα/primase is bound to Ctc1/Stn1/Ten1 (CST), a single-stranded DNA-binding complex. Like mutations in telomerase, mutations affecting CST-Polα/primase result in pathological telomere shortening and cause a telomere biology disorder, Coats plus (CP). We determined cryogenic electron microscopy structures of human CST bound to the shelterin heterodimer POT1/TPP1 that reveal how CST is recruited to telomeres by POT1. Our findings suggest that POT1 hinge phosphorylation is required for CST recruitment, and the complex is formed through conserved interactions involving several residues mutated in CP. Our structural and biochemical data suggest that phosphorylated POT1 holds CST-Polα/primase in an inactive, autoinhibited state until telomerase has extended the telomere ends. We propose that dephosphorylation of POT1 releases CST-Polα/primase into an active state that completes telomere replication through fill-in synthesis.


Subject(s)
DNA Polymerase I , Shelterin Complex , Telomere-Binding Proteins , Telomere , Humans , Cryoelectron Microscopy , DNA Polymerase I/metabolism , DNA Primase/metabolism , DNA Primase/genetics , Models, Molecular , Phosphorylation , Shelterin Complex/metabolism , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/metabolism
3.
Cell ; 183(5): 1162-1184, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33242416

ABSTRACT

Research on astronaut health and model organisms have revealed six features of spaceflight biology that guide our current understanding of fundamental molecular changes that occur during space travel. The features include oxidative stress, DNA damage, mitochondrial dysregulation, epigenetic changes (including gene regulation), telomere length alterations, and microbiome shifts. Here we review the known hazards of human spaceflight, how spaceflight affects living systems through these six fundamental features, and the associated health risks of space exploration. We also discuss the essential issues related to the health and safety of astronauts involved in future missions, especially planned long-duration and Martian missions.


Subject(s)
Extraterrestrial Environment , Space Flight , Astronauts , Health , Humans , Microbiota , Risk Factors
4.
Cell ; 172(1-2): 331-343.e13, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29290466

ABSTRACT

Telomerase maintains chromosome ends from humans to yeasts. Recruitment of yeast telomerase to telomeres occurs through its Ku and Est1 subunits via independent interactions with telomerase RNA (TLC1) and telomeric proteins Sir4 and Cdc13, respectively. However, the structures of the molecules comprising these telomerase-recruiting pathways remain unknown. Here, we report crystal structures of the Ku heterodimer and Est1 complexed with their key binding partners. Two major findings are as follows: (1) Ku specifically binds to telomerase RNA in a distinct, yet related, manner to how it binds DNA; and (2) Est1 employs two separate pockets to bind distinct motifs of Cdc13. The N-terminal Cdc13-binding site of Est1 cooperates with the TLC1-Ku-Sir4 pathway for telomerase recruitment, whereas the C-terminal interface is dispensable for binding Est1 in vitro yet is nevertheless essential for telomere maintenance in vivo. Overall, our results integrate previous models and provide fundamentally valuable structural information regarding telomere biology.


Subject(s)
DNA-Binding Proteins/chemistry , Molecular Docking Simulation , Saccharomyces cerevisiae Proteins/chemistry , Telomerase/chemistry , Telomere Homeostasis , Telomere-Binding Proteins/chemistry , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Protein Binding , RNA/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Telomerase/genetics , Telomerase/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
5.
Cell ; 173(4): 972-988.e23, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29656893

ABSTRACT

Repair of damaged DNA is essential for maintaining genome integrity and for preventing genome-instability-associated diseases, such as cancer. By combining proximity labeling with quantitative mass spectrometry, we generated high-resolution interaction neighborhood maps of the endogenously expressed DNA repair factors 53BP1, BRCA1, and MDC1. Our spatially resolved interaction maps reveal rich network intricacies, identify shared and bait-specific interaction modules, and implicate previously concealed regulators in this process. We identified a novel vertebrate-specific protein complex, shieldin, comprising REV7 plus three previously uncharacterized proteins, RINN1 (CTC-534A2.2), RINN2 (FAM35A), and RINN3 (C20ORF196). Recruitment of shieldin to DSBs, via the ATM-RNF8-RNF168-53BP1-RIF1 axis, promotes NHEJ-dependent repair of intrachromosomal breaks, immunoglobulin class-switch recombination (CSR), and fusion of unprotected telomeres. Shieldin functions as a downstream effector of 53BP1-RIF1 in restraining DNA end resection and in sensitizing BRCA1-deficient cells to PARP inhibitors. These findings have implications for understanding cancer-associated PARPi resistance and the evolution of antibody CSR in higher vertebrates.


Subject(s)
DNA End-Joining Repair/drug effects , DNA-Binding Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Adaptor Proteins, Signal Transducing , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Cycle Proteins , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , Immunoglobulin Class Switching/drug effects , Mad2 Proteins/antagonists & inhibitors , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Telomere-Binding Proteins/antagonists & inhibitors , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Suppressor p53-Binding Protein 1/antagonists & inhibitors , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
6.
Cell ; 173(5): 1179-1190.e13, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29775593

ABSTRACT

Telomerase is an RNA-protein complex (RNP) that extends telomeric DNA at the 3' ends of chromosomes using its telomerase reverse transcriptase (TERT) and integral template-containing telomerase RNA (TER). Its activity is a critical determinant of human health, affecting aging, cancer, and stem cell renewal. Lack of atomic models of telomerase, particularly one with DNA bound, has limited our mechanistic understanding of telomeric DNA repeat synthesis. We report the 4.8 Å resolution cryoelectron microscopy structure of active Tetrahymena telomerase bound to telomeric DNA. The catalytic core is an intricately interlocked structure of TERT and TER, including a previously structurally uncharacterized TERT domain that interacts with the TEN domain to physically enclose TER and regulate activity. This complete structure of a telomerase catalytic core and its interactions with telomeric DNA from the template to telomere-interacting p50-TEB complex provides unanticipated insights into telomerase assembly and catalytic cycle and a new paradigm for a reverse transcriptase RNP.


Subject(s)
DNA/metabolism , Telomerase/metabolism , Telomere/metabolism , Tetrahymena thermophila/metabolism , Catalytic Domain , Cryoelectron Microscopy , DNA/chemistry , Humans , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , Shelterin Complex , Tartrate-Resistant Acid Phosphatase/metabolism , Telomerase/chemistry , Telomere/chemistry , Telomere-Binding Proteins , Tetrahymena thermophila/enzymology
7.
Cell ; 174(1): 218-230.e13, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29804836

ABSTRACT

Ribonucleoprotein enzymes require dynamic conformations of their RNA constituents for regulated catalysis. Human telomerase employs a non-coding RNA (hTR) with a bipartite arrangement of domains-a template-containing core and a distal three-way junction (CR4/5) that stimulates catalysis through unknown means. Here, we show that telomerase activity unexpectedly depends upon the holoenzyme protein TCAB1, which in turn controls conformation of CR4/5. Cells lacking TCAB1 exhibit a marked reduction in telomerase catalysis without affecting enzyme assembly. Instead, TCAB1 inactivation causes unfolding of CR4/5 helices that are required for catalysis and for association with the telomerase reverse-transcriptase (TERT). CR4/5 mutations derived from patients with telomere biology disorders provoke defects in catalysis and TERT binding similar to TCAB1 inactivation. These findings reveal a conformational "activity switch" in human telomerase RNA controlling catalysis and TERT engagement. The identification of two discrete catalytic states for telomerase suggests an intramolecular means for controlling telomerase in cancers and progenitor cells.


Subject(s)
RNA, Untranslated/chemistry , Telomerase/metabolism , Biocatalysis , Cell Line , HeLa Cells , Humans , Molecular Chaperones , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , RNA, Untranslated/metabolism , Telomerase/antagonists & inhibitors , Telomerase/chemistry , Telomerase/genetics , Telomere/metabolism
8.
Annu Rev Biochem ; 86: 439-460, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28141967

ABSTRACT

Telomerase is the essential reverse transcriptase required for linear chromosome maintenance in most eukaryotes. Telomerase supplements the tandem array of simple-sequence repeats at chromosome ends to compensate for the DNA erosion inherent in genome replication. The template for telomerase reverse transcriptase is within the RNA subunit of the ribonucleoprotein complex, which in cells contains additional telomerase holoenzyme proteins that assemble the active ribonucleoprotein and promote its function at telomeres. Telomerase is distinct among polymerases in its reiterative reuse of an internal template. The template is precisely defined, processively copied, and regenerated by release of single-stranded product DNA. New specificities of nucleic acid handling that underlie the catalytic cycle of repeat synthesis derive from both active site specialization and new motif elaborations in protein and RNA subunits. Studies of telomerase provide unique insights into cellular requirements for genome stability, tissue renewal, and tumorigenesis as well as new perspectives on dynamic ribonucleoprotein machines.


Subject(s)
DNA Replication , DNA, Single-Stranded/metabolism , RNA/metabolism , Ribonucleoproteins/metabolism , Telomerase/metabolism , Telomere/enzymology , Animals , Catalytic Domain , DNA, Single-Stranded/genetics , Gene Expression Regulation , Humans , Microsatellite Repeats , Nucleic Acid Conformation , Oxytricha/genetics , Oxytricha/metabolism , RNA/genetics , Ribonucleoproteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomerase/genetics , Telomere/chemistry , Tetrahymena thermophila/genetics , Tetrahymena thermophila/metabolism
9.
Cell ; 170(1): 72-85.e14, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28666126

ABSTRACT

Maintenance of a minimal telomere length is essential to prevent cellular senescence. When critically short telomeres arise in the absence of telomerase, they can be repaired by homology-directed repair (HDR) to prevent premature senescence onset. It is unclear why specifically the shortest telomeres are targeted for HDR. We demonstrate that the non-coding RNA TERRA accumulates as HDR-promoting RNA-DNA hybrids (R-loops) preferentially at very short telomeres. The increased level of TERRA and R-loops, exclusively at short telomeres, is due to a local defect in RNA degradation by the Rat1 and RNase H2 nucleases, respectively. Consequently, the coordination of TERRA degradation with telomere replication is altered at shortened telomeres. R-loop persistence at short telomeres contributes to activation of the DNA damage response (DDR) and promotes recruitment of the Rad51 recombinase. Thus, the telomere length-dependent regulation of TERRA and TERRA R-loops is a critical determinant of the rate of replicative senescence.


Subject(s)
Cell Cycle , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Telomere/metabolism , Cellular Senescence , DNA Damage , Exoribonucleases/metabolism , Nucleic Acid Hybridization , Recombinational DNA Repair , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere/chemistry , Telomere-Binding Proteins/metabolism
10.
Cell ; 169(6): 1000-1011, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575665

ABSTRACT

Many cellular stresses activate senescence, a persistent hyporeplicative state characterized in part by expression of the p16INK4a cell-cycle inhibitor. Senescent cell production occurs throughout life and plays beneficial roles in a variety of physiological and pathological processes including embryogenesis, wound healing, host immunity, and tumor suppression. Meanwhile, the steady accumulation of senescent cells with age also has adverse consequences. These non-proliferating cells occupy key cellular niches and elaborate pro-inflammatory cytokines, contributing to aging-related diseases and morbidity. This model suggests that the abundance of senescent cells in vivo predicts "molecular," as opposed to chronologic, age and that senescent cell clearance may mitigate aging-associated pathology.


Subject(s)
Aging/pathology , Cell Cycle , Cellular Senescence , Animals , Humans , Neoplasms/immunology , Wound Healing
11.
Mol Cell ; 84(9): 1684-1698.e9, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38593805

ABSTRACT

The Bloom syndrome (BLM) helicase is critical for alternative lengthening of telomeres (ALT), a homology-directed repair (HDR)-mediated telomere maintenance mechanism that is prevalent in cancers of mesenchymal origin. The DNA substrates that BLM engages to direct telomere recombination during ALT remain unknown. Here, we determine that BLM helicase acts on lagging strand telomere intermediates that occur specifically in ALT-positive cells to assemble a replication-associated DNA damage response. Loss of ATRX was permissive for BLM localization to ALT telomeres in S and G2, commensurate with the appearance of telomere C-strand-specific single-stranded DNA (ssDNA). DNA2 nuclease deficiency increased 5'-flap formation in a BLM-dependent manner, while telomere C-strand, but not G-strand, nicks promoted ALT. These findings define the seminal events in the ALT DNA damage response, linking aberrant telomeric lagging strand DNA replication with a BLM-directed HDR mechanism that sustains telomere length in a subset of human cancers.


Subject(s)
DNA Damage , DNA Replication , RecQ Helicases , Telomere Homeostasis , Telomere , RecQ Helicases/metabolism , RecQ Helicases/genetics , Humans , Telomere/metabolism , Telomere/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Bloom Syndrome/enzymology , Bloom Syndrome/pathology , Cell Line, Tumor
12.
Annu Rev Biochem ; 85: 35-64, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27294438

ABSTRACT

The health of an organism is orchestrated by a multitude of molecular and biochemical networks responsible for ensuring homeostasis within cells and tissues. However, upon aging, a progressive failure in the maintenance of this homeostatic balance occurs in response to a variety of endogenous and environmental stresses, allowing the accumulation of damage, the physiological decline of individual tissues, and susceptibility to diseases. What are the molecular and cellular signaling events that control the aging process and how can this knowledge help design therapeutic strategies to combat age-associated diseases? Here we provide a comprehensive overview of the evolutionarily conserved biological processes that alter the rate of aging and discuss their link to disease prevention and the extension of healthy life span.


Subject(s)
DNA Damage , Longevity/genetics , Proteostasis Deficiencies/genetics , Signal Transduction , Telomere Shortening , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Caloric Restriction , Epigenesis, Genetic , Homeostasis/genetics , Humans , Inflammation , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mitochondria/metabolism , Oxidative Stress , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
13.
Genes Dev ; 38(15-16): 755-771, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39231615

ABSTRACT

Premature telomere shortening or telomere instability is associated with a group of rare and heterogeneous diseases collectively known as telomere biology disorders (TBDs). Here we identified two unrelated individuals with clinical manifestations of TBDs and short telomeres associated with the identical monoallelic variant c.767A>G; Y256C in RPA2 Although the replication protein A2 (RPA2) mutant did not affect ssDNA binding and G-quadruplex-unfolding properties of RPA, the mutation reduced the affinity of RPA2 with the ubiquitin ligase RFWD3 and reduced RPA ubiquitination. Using engineered knock-in cell lines, we found an accumulation of RPA at telomeres that did not trigger ATR activation but caused short and dysfunctional telomeres. Finally, both patients acquired, in a subset of blood cells, somatic genetic rescue events in either POT1 genes or TERT promoters known to counteract the accelerated telomere shortening. Collectively, our study indicates that variants in RPA2 represent a novel genetic cause of TBDs. Our results further support the fundamental role of the RPA complex in regulating telomere length and stability in humans.


Subject(s)
Replication Protein A , Telomere-Binding Proteins , Telomere , Humans , Replication Protein A/genetics , Replication Protein A/metabolism , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Heterozygote , Male , Female , Shelterin Complex , Telomere Shortening/genetics , Mutation , Telomerase/genetics , Telomerase/metabolism , Ubiquitination/genetics , Ubiquitin-Protein Ligases/genetics
14.
Mol Cell ; 83(10): 1743-1760.e11, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37116497

ABSTRACT

PARP1, an established anti-cancer target that regulates many cellular pathways, including DNA repair signaling, has been intensely studied for decades as a poly(ADP-ribosyl)transferase. Although recent studies have revealed the prevalence of mono-ADP-ribosylation upon DNA damage, it was unknown whether this signal plays an active role in the cell or is just a byproduct of poly-ADP-ribosylation. By engineering SpyTag-based modular antibodies for sensitive and flexible detection of mono-ADP-ribosylation, including fluorescence-based sensors for live-cell imaging, we demonstrate that serine mono-ADP-ribosylation constitutes a second wave of PARP1 signaling shaped by the cellular HPF1/PARP1 ratio. Multilevel chromatin proteomics reveals histone mono-ADP-ribosylation readers, including RNF114, a ubiquitin ligase recruited to DNA lesions through a zinc-finger domain, modulating the DNA damage response and telomere maintenance. Our work provides a technological framework for illuminating ADP-ribosylation in a wide range of applications and biological contexts and establishes mono-ADP-ribosylation by HPF1/PARP1 as an important information carrier for cell signaling.


Subject(s)
ADP-Ribosylation , Histones , Histones/genetics , Histones/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Chromatin , DNA Damage , Antibodies/genetics , Signal Transduction
15.
Mol Cell ; 83(7): 1043-1060.e10, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36854302

ABSTRACT

Repair of DNA double-strand breaks (DSBs) elicits three-dimensional (3D) chromatin topological changes. A recent finding reveals that 53BP1 assembles into a 3D chromatin topology pattern around DSBs. How this formation of a higher-order structure is configured and regulated remains enigmatic. Here, we report that SLFN5 is a critical factor for 53BP1 topological arrangement at DSBs. Using super-resolution imaging, we find that SLFN5 binds to 53BP1 chromatin domains to assemble a higher-order microdomain architecture by driving damaged chromatin dynamics at both DSBs and deprotected telomeres. Mechanistically, we propose that 53BP1 topology is shaped by two processes: (1) chromatin mobility driven by the SLFN5-LINC-microtubule axis and (2) the assembly of 53BP1 oligomers mediated by SLFN5. In mammals, SLFN5 deficiency disrupts the DSB repair topology and impairs non-homologous end joining, telomere fusions, class switch recombination, and sensitivity to poly (ADP-ribose) polymerase inhibitor. We establish a molecular mechanism that shapes higher-order chromatin topologies to safeguard genomic stability.


Subject(s)
Chromatin , DNA Repair , Animals , Chromatin/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Mammals/metabolism , Telomere-Binding Proteins/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Cell Cycle Proteins/metabolism
16.
Genes Dev ; 37(13-14): 555-569, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37495394

ABSTRACT

It has been known for decades that telomerase extends the 3' end of linear eukaryotic chromosomes and dictates the telomeric repeat sequence based on the template in its RNA. However, telomerase does not mitigate sequence loss at the 5' ends of chromosomes, which results from lagging strand DNA synthesis and nucleolytic processing. Therefore, a second enzyme is needed to keep telomeres intact: DNA polymerase α/Primase bound to Ctc1-Stn1-Ten1 (CST). CST-Polα/Primase maintains telomeres through a fill-in reaction that replenishes the lost sequences at the 5' ends. CST not only serves to maintain telomeres but also determines their length by keeping telomerase from overelongating telomeres. Here we discuss recent data on the evolution, structure, function, and recruitment of mammalian CST-Polα/Primase, highlighting the role of this complex and telomere length control in human disease.


Subject(s)
Telomerase , Animals , Humans , Telomerase/metabolism , DNA Primase/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis , DNA Replication , Mammals/genetics
17.
Cell ; 163(7): 1641-54, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26687355

ABSTRACT

Telomere crisis occurs during tumorigenesis when depletion of the telomere reserve leads to frequent telomere fusions. The resulting dicentric chromosomes have been proposed to drive genome instability. Here, we examine the fate of dicentric human chromosomes in telomere crisis. We observed that dicentric chromosomes invariably persisted through mitosis and developed into 50-200 µm chromatin bridges connecting the daughter cells. Before their resolution at 3-20 hr after anaphase, the chromatin bridges induced nuclear envelope rupture in interphase, accumulated the cytoplasmic 3' nuclease TREX1, and developed RPA-coated single stranded (ss) DNA. CRISPR knockouts showed that TREX1 contributed to the generation of the ssDNA and the resolution of the chromatin bridges. Post-crisis clones showed chromothripsis and kataegis, presumably resulting from DNA repair and APOBEC editing of the fragmented chromatin bridge DNA. We propose that chromothripsis in human cancer may arise through TREX1-mediated fragmentation of dicentric chromosomes formed in telomere crisis.


Subject(s)
Chromosomal Instability , Chromosomes, Human , Genomic Instability , Neoplasms/genetics , Telomere , Chromosome Aberrations , Cytokinesis , DNA, Single-Stranded/metabolism , Exodeoxyribonucleases/metabolism , Humans , Mitosis , Nuclear Envelope/metabolism , Phosphoproteins/metabolism
18.
Mol Cell ; 82(21): 4001-4017.e7, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36265488

ABSTRACT

Alternative lengthening of telomeres (ALT) is a homology-directed repair (HDR) mechanism of telomere elongation that controls proliferation in subsets of aggressive cancer. Recent studies have revealed that telomere repeat-containing RNA (TERRA) promotes ALT-associated HDR (ALT-HDR). Here, we report that RAD51AP1, a crucial ALT factor, interacts with TERRA and utilizes it to generate D- and R-loop HR intermediates. We also show that RAD51AP1 binds to and might stabilize TERRA-containing R-loops as RAD51AP1 depletion reduces R-loop formation at telomere DNA breaks. Proteomic analyses uncover a role for RAD51AP1-mediated TERRA R-loop homeostasis in a mechanism of chromatin-directed suppression of TERRA and prevention of transcription-replication collisions (TRCs) during ALT-HDR. Intriguingly, we find that both TERRA binding and this non-canonical function of RAD51AP1 require its intrinsic SUMO-SIM regulatory axis. These findings provide insights into the multi-contextual functions of RAD51AP1 within the ALT mechanism and regulation of TERRA.


Subject(s)
RNA, Long Noncoding , Telomere Homeostasis , Chromatin/genetics , Proteomics , Telomere/genetics , Telomere/metabolism , RNA, Long Noncoding/genetics , Homeostasis
19.
Mol Cell ; 82(21): 3985-4000.e4, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36265486

ABSTRACT

Alternative lengthening of telomeres (ALT), a telomerase-independent process maintaining telomeres, is mediated by break-induced replication (BIR). RAD52 promotes ALT by facilitating D-loop formation, but ALT also occurs through a RAD52-independent BIR pathway. Here, we show that the telomere non-coding RNA TERRA forms dynamic telomeric R-loops and contributes to ALT activity in RAD52 knockout cells. TERRA forms R-loops in vitro and at telomeres in a RAD51AP1-dependent manner. The formation of R-loops by TERRA increases G-quadruplexes (G4s) at telomeres. G4 stabilization enhances ALT even when TERRA is depleted, suggesting that G4s act downstream of R-loops to promote BIR. In vitro, the telomeric R-loops assembled by TERRA and RAD51AP1 generate G4s, which persist after R-loop resolution and allow formation of telomeric D-loops without RAD52. Thus, the dynamic telomeric R-loops formed by TERRA and RAD51AP1 enable the RAD52-independent ALT pathway, and G4s orchestrate an R- to D-loop switch at telomeres to stimulate BIR.


Subject(s)
RNA, Long Noncoding , Telomerase , Telomere Homeostasis , Telomere/genetics , Telomere/metabolism , Telomerase/genetics , Telomerase/metabolism , R-Loop Structures/genetics , DNA Repair
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL