Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39082275

RESUMEN

Inappropriate homology-directed repair (HDR) of telomeres results in catastrophic telomere loss and aberrant chromosome fusions, leading to genome instability. We have previously shown that the TRF2-RAP1 heterodimer protects telomeres from engaging in aberrant telomere HDR. Cells lacking the basic domain of TRF2 and functional RAP1 display HDR-mediated telomere clustering, resulting in the formation of ultrabright telomeres (UTs) and massive chromosome fusions. Using purified proteins, we uncover three distinct molecular pathways that the TRF2-RAP1 heterodimer utilizes to protect telomeres from engaging in aberrant HDR. We show mechanistically that TRF2-RAP1 inhibits RAD51-initiated telomeric D-loop formation. Both the TRF2 basic domain and RAP1-binding to TRF2 are required to block RAD51-mediated homology search. TRF2 recruits the BLM helicase to telomeres through its TRFH domain to promote BLM-mediated unwinding of telomere D-loops. In addition, TRF2-RAP1 inhibits BLM-DNA2-mediated 5' telomere end resection, preventing the generation of 3' single-stranded telomere overhangs necessary for RAD51-dependent HDR. Importantly, cells expressing BLM mutants unable to interact with TRF2 accumulate telomere D-loops and UTs. Our findings uncover distinct molecular mechanisms coordinated by TRF2-RAP1 to protect telomeres from engaging in aberrant HDR.

2.
Bioessays ; 46(2): e2300184, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38047499

RESUMEN

Mammalian telomeres have evolved safeguards to prevent their recognition as DNA double-stranded breaks by suppressing the activation of various DNA sensing and repair proteins. We have shown that the telomere-binding proteins TRF2 and RAP1 cooperate to prevent telomeres from undergoing aberrant homology-directed recombination by mediating t-loop protection. Our recent findings also suggest that mammalian telomere-binding proteins interact with the nuclear envelope to maintain chromosome stability. RAP1 interacts with nuclear lamins through KU70/KU80, and disruption of RAP1 and TRF2 function result in nuclear envelope rupture, promoting telomere-telomere recombination to form structures termed ultrabright telomeres. In this review, we discuss the importance of the interactions between shelterin components and the nuclear envelope to maintain telomere homeostasis and genome stability.


Asunto(s)
Membrana Nuclear , Telómero , Animales , Humanos , Membrana Nuclear/metabolismo , Telómero/genética , Telómero/metabolismo , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , ADN/metabolismo , Inestabilidad Genómica , Mamíferos/genética
4.
Nat Commun ; 14(1): 2144, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37059728

RESUMEN

Double-strand breaks (DSBs) due to genotoxic stress represent potential threats to genome stability. Dysfunctional telomeres are recognized as DSBs and are repaired by distinct DNA repair mechanisms. RAP1 and TRF2 are telomere binding proteins essential to protect telomeres from engaging in homology directed repair (HDR), but how this occurs remains unclear. In this study, we examined how the basic domain of TRF2 (TRF2B) and RAP1 cooperate to repress HDR at telomeres. Telomeres lacking TRF2B and RAP1 cluster into structures termed ultrabright telomeres (UTs). HDR factors localize to UTs, and UT formation is abolished by RNaseH1, DDX21 and ADAR1p110, suggesting that they contain DNA-RNA hybrids. Interaction between the BRCT domain of RAP1 and KU70/KU80 is also required to repress UT formation. Expressing TRF2∆B in Rap1-/- cells resulted in aberrant lamin A localization in the nuclear envelope and dramatically increased UT formation. Expressing lamin A phosphomimetic mutants induced nuclear envelope rupturing and aberrant HDR-mediated UT formation. Our results highlight the importance of shelterin and proteins in the nuclear envelope in repressing aberrant telomere-telomere recombination to maintain telomere homeostasis.


Asunto(s)
Membrana Nuclear , Proteína 2 de Unión a Repeticiones Teloméricas , Lamina Tipo A/metabolismo , Membrana Nuclear/metabolismo , Telómero/genética , Telómero/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteínas de Unión al GTP rap1/metabolismo
5.
Nat Commun ; 11(1): 5861, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33203878

RESUMEN

Telomeres protect chromosome ends from inappropriately activating the DNA damage and repair responses. Primary microcephaly is a key clinical feature of several human telomere disorder syndromes, but how microcephaly is linked to dysfunctional telomeres is not known. Here, we show that the microcephalin 1/BRCT-repeats inhibitor of hTERT (MCPH1/BRIT1) protein, mutated in primary microcephaly, specifically interacts with the TRFH domain of the telomere binding protein TRF2. The crystal structure of the MCPH1-TRF2 complex reveals that this interaction is mediated by the MCPH1 330YRLSP334 motif. TRF2-dependent recruitment of MCPH1 promotes localization of DNA damage factors and homology directed repair of dysfunctional telomeres lacking POT1-TPP1. Additionally, MCPH1 is involved in the replication stress response, promoting telomere replication fork progression and restart of stalled telomere replication forks. Our work uncovers a previously unrecognized role for MCPH1 in promoting telomere replication, providing evidence that telomere replication defects may contribute to the onset of microcephaly.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Microcefalia/genética , Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Animales , Sitios de Unión , Calorimetría , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Daño del ADN , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Fibroblastos , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Mutación , Dominios y Motivos de Interacción de Proteínas , Serina Proteasas/genética , Serina Proteasas/metabolismo , Complejo Shelterina , Telómero/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/química , Proteína 2 de Unión a Repeticiones Teloméricas/genética
6.
Cell Rep ; 29(11): 3708-3725.e5, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31825846

RESUMEN

Telomeres use shelterin to protect chromosome ends from activating the DNA damage sensor MRE11-RAD50-NBS1 (MRN), repressing ataxia-telangiectasia, mutated (ATM) and ATM and Rad3-related (ATR) dependent DNA damage checkpoint responses. The MRE11 nuclease is thought to be essential for the resection of the 5' C-strand to generate the microhomologies necessary for alternative non-homologous end joining (A-NHEJ) repair. In the present study, we uncover DNA damage signaling and repair pathways engaged by components of the replisome complex to repair dysfunctional telomeres. In cells lacking MRN, single-stranded telomeric overhangs devoid of POT1-TPP1 do not recruit replication protein A (RPA), ATR-interacting protein (ATRIP), and RAD 51. Rather, components of the replisome complex, including Claspin, Proliferating cell nuclear antigen (PCNA), and Downstream neighbor of SON (DONSON), initiate DNA-PKcs-mediated p-CHK1 activation and A-NHEJ repair. In addition, Claspin directly interacts with TRF2 and recruits EXO1 to newly replicated telomeres to promote 5' end resection. Our data indicate that MRN is dispensable for the repair of dysfunctional telomeres lacking POT1-TPP1 and highlight the contributions of the replisome in telomere repair.


Asunto(s)
Reparación del ADN por Unión de Extremidades , ADN Polimerasa Dirigida por ADN/metabolismo , Complejos Multienzimáticos/metabolismo , Telómero/metabolismo , Ácido Anhídrido Hidrolasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aminopeptidasas/deficiencia , Aminopeptidasas/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/deficiencia , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Exodesoxirribonucleasas/metabolismo , Células HEK293 , Humanos , Proteína Homóloga de MRE11/metabolismo , Ratones , Complejos Multienzimáticos/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Serina Proteasas/deficiencia , Serina Proteasas/metabolismo , Complejo Shelterina , Telómero/genética , Proteínas de Unión a Telómeros/deficiencia , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
7.
Cell Res ; 27(12): 1485-1502, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29160297

RESUMEN

Telomeres are nucleoprotein complexes that play essential roles in protecting chromosome ends. Mammalian telomeres consist of repetitive DNA sequences bound by the shelterin complex. In this complex, the POT1-TPP1 heterodimer binds to single-stranded telomeric DNAs, while TRF1 and TRF2-RAP1 interact with double-stranded telomeric DNAs. TIN2, the linchpin of this complex, simultaneously interacts with TRF1, TRF2, and TPP1 to mediate the stable assembly of the shelterin complex. However, the molecular mechanism by which TIN2 interacts with these proteins to orchestrate telomere protection remains poorly understood. Here, we report the crystal structure of the N-terminal domain of TIN2 in complex with TIN2-binding motifs from TPP1 and TRF2, revealing how TIN2 interacts cooperatively with TPP1 and TRF2. Unexpectedly, TIN2 contains a telomeric repeat factor homology (TRFH)-like domain that functions as a protein-protein interaction platform. Structure-based mutagenesis analyses suggest that TIN2 plays an important role in maintaining the stable shelterin complex required for proper telomere end protection.


Asunto(s)
Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Serina Proteasas/metabolismo , Complejo Shelterina/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/química , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/química , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/aislamiento & purificación , Humanos , Ratones , Conformación Proteica , Serina Proteasas/química , Serina Proteasas/aislamiento & purificación , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/aislamiento & purificación , Proteína 2 de Unión a Repeticiones Teloméricas/química , Proteína 2 de Unión a Repeticiones Teloméricas/aislamiento & purificación
8.
Nat Commun ; 8: 14929, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28393832

RESUMEN

Mammalian shelterin proteins POT1 and TPP1 form a stable heterodimer that protects chromosome ends and regulates telomerase-mediated telomere extension. However, how POT1 interacts with TPP1 remains unknown. Here we present the crystal structure of the C-terminal portion of human POT1 (POT1C) complexed with the POT1-binding motif of TPP1. The structure shows that POT1C contains two domains, a third OB fold and a Holliday junction resolvase-like domain. Both domains are essential for binding to TPP1. Notably, unlike the heart-shaped structure of ciliated protozoan Oxytricha nova TEBPα-ß complex, POT1-TPP1 adopts an elongated V-shaped conformation. In addition, we identify several missense mutations in human cancers that disrupt the POT1C-TPP1 interaction, resulting in POT1 instability. POT1C mutants that bind TPP1 localize to telomeres but fail to repress a DNA damage response and inappropriate repair by A-NHEJ. Our results reveal that POT1 C terminus is essential to prevent initiation of genome instability permissive for tumorigenesis.


Asunto(s)
Mutación/genética , Neoplasias/genética , Complejo Shelterina/genética , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Daño del ADN , Análisis Mutacional de ADN , Reparación del ADN , Inestabilidad Genómica , Humanos , Ratones , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Neoplasias/patología , Fosfoproteínas/metabolismo , Prostaglandina-E Sintasas , Unión Proteica , Estructura Secundaria de Proteína , Dispersión del Ángulo Pequeño , Complejo Shelterina/metabolismo , Relación Estructura-Actividad , Difracción de Rayos X
9.
Methods Mol Biol ; 1587: 127-131, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28324504

RESUMEN

Dysfunctional telomeres arising either through natural attrition due to telomerase deficiency or by the removal of telomere-binding proteins are recognized as double-stranded breaks (DSBs). Repair of DSBs is crucial for the maintenance of genome stability. In mammals, DSBs are repaired by either error-prone nonhomologous end joining (NHEJ) or error-free homologous recombination (HR) and can be visualized as chromosomal fusions.


Asunto(s)
Telómero/genética , Animales , Células Cultivadas , Análisis Citogenético/métodos , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Inestabilidad Genómica/genética , Ratones , Telomerasa/genética
10.
Methods Mol Biol ; 1587: 133-138, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28324505

RESUMEN

Telomere dysfunctions, rendered through replicative attrition of telomeric DNA or due to the removal of shelterin components, are recognized as DNA double-stranded breaks (DSBs) by the DNA damage repair (DDR) pathway. This leads to the activation of DNA damage checkpoint sensors, including the Mre11-Rad50-Nbs1 (MRN) complex, γ-H2AX and 53BP1, the ATM and ATR signal-transducing kinases, and downstream effectors, including Chk1, Chk2, and p53. Robust DNA damage response signals at dysfunctional telomeres, achieved by the complete deletion of TRF2 or by expressing dominant-negative mutant TPP1ΔRD, can be detected by their association with γ-H2AX and 53BP1 forming "telomere dysfunction induced foci (TIFs)." Induction of TIFs at telomeres provides an opportunity to quantify the extent of telomere dysfunction and monitor downstream signaling pathways.


Asunto(s)
Daño del ADN/genética , Telómero/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de Ciclo Celular/genética , Células Cultivadas , ADN/genética , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Ratones , Transducción de Señal/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética
11.
Mol Cell ; 65(5): 801-817.e4, 2017 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-28216226

RESUMEN

Telomeres employ TRF2 to protect chromosome ends from activating the DNA damage sensor MRE11-RAD50-NBS1 (MRN), thereby repressing ATM-dependent DNA damage checkpoint responses. How TRF2 prevents MRN activation at dysfunctional telomeres is unclear. Here, we show that the phosphorylation status of NBS1 determines the repair pathway choice of dysfunctional telomeres. The crystal structure of the TRF2-NBS1 complex at 3.0 Å resolution shows that the NBS1 429YQLSP433 motif interacts specifically with the TRF2TRFH domain. Phosphorylation of NBS1 serine 432 by CDK2 in S/G2 dissociates NBS1 from TRF2, promoting TRF2-Apollo/SNM1B complex formation and the protection of leading-strand telomeres. Classical-NHEJ-mediated repair of telomeres lacking TRF2 requires phosphorylated NBS1S432 to activate ATM, while interaction of de-phosphorylated NBS1S432 with TRF2 promotes alternative-NHEJ repair of telomeres lacking POT1-TPP1. Our work advances understanding of how the TRF2TRFH domain orchestrates telomere end protection and reveals how the phosphorylation status of the NBS1S432 dictates repair pathway choice of dysfunctional telomeres.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas Nucleares/metabolismo , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Exodesoxirribonucleasas , Fase G1 , Fase G2 , Células HCT116 , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Fase S , Serina Proteasas/genética , Serina Proteasas/metabolismo , Complejo Shelterina , Relación Estructura-Actividad , Telómero/genética , Telómero/patología , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/química , Proteína 2 de Unión a Repeticiones Teloméricas/genética
12.
Nat Commun ; 7: 10881, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26941064

RESUMEN

Repressor/activator protein 1 (RAP1) is a highly conserved telomere-interacting protein. Yeast Rap1 protects telomeres from non-homologous end joining (NHEJ), plays important roles in telomere length control and is involved in transcriptional gene regulation. However, a role for mammalian RAP1 in telomere end protection remains controversial. Here we present evidence that mammalian RAP1 is essential to protect telomere from homology directed repair (HDR) of telomeres. RAP1 cooperates with the basic domain of TRF2 (TRF2(B)) to repress PARP1 and SLX4 localization to telomeres. Without RAP1 and TRF2(B), PARP1 and SLX4 HR factors promote rapid telomere resection, resulting in catastrophic telomere loss and the generation of telomere-free chromosome fusions in both mouse and human cells. The RAP1 Myb domain is required to repress both telomere loss and formation of telomere-free fusions. Our results highlight the importance of the RAP1-TRF2 heterodimer in protecting telomeres from inappropriate processing by the HDR pathway.


Asunto(s)
Eliminación de Gen , Regulación de la Expresión Génica/fisiología , Proteínas Recombinantes/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/fisiología , Animales , Línea Celular , Reparación del ADN , Proteínas de Unión al ADN , Fibroblastos/metabolismo , Inestabilidad Genómica , Humanos , Ratones , Transporte de Proteínas , Proteínas Recombinantes/genética , Complejo Shelterina , Telómero/fisiología , Proteínas de Unión a Telómeros/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética
13.
Methods Mol Biol ; 1343: 175-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26420717

RESUMEN

Telomeres are repetitive DNA repeats that cap the ends of all eukaryotic chromosomes. Their proper maintenance is essential for genomic stability and cellular viability. Dysfunctional telomeres could arise through natural attrition of telomeric DNA or due to the removal of shelterin components. These uncapped chromosomal ends are recognized as DSBs by the DDR pathway, leading to the accumulation of DNA damage sensors at telomeres. The association of these DDR proteins with dysfunctional telomeres forms telomere dysfunction induced DNA damage foci (TIFs). Detection of TIFs at telomeres provides an opportunity to quantify the extent of telomere dysfunction and monitor downstream DNA damage signaling pathways. Here we describe a method for the detection of TIFs using a fluorescent in situ hybridization (FISH) approach.


Asunto(s)
Daño del ADN , Telómero/metabolismo , Células HEK293 , Humanos , Hibridación Fluorescente in Situ/métodos
14.
Nat Struct Mol Biol ; 18(12): 1400-7, 2011 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-22101936

RESUMEN

The mammalian shelterin component TPP1 has essential roles in telomere maintenance and, together with POT1, is required for the repression of DNA damage signaling at telomeres. Here we show that in Mus musculus, the E3 ubiquitin ligase Rnf8 localizes to uncapped telomeres and promotes the accumulation of DNA damage proteins 53Bp1 and γ-H2ax. In the absence of Rnf8, Tpp1 is unstable, resulting in telomere shortening and chromosome fusions through the alternative nonhomologous end-joining (A-NHEJ) repair pathway. The Rnf8 RING-finger domain is essential for Tpp1 stability and retention at telomeres. Rnf8 physically interacts with Tpp1 to generate Ubc13-dependent Lys63 polyubiquitin chains that stabilize Tpp1 at telomeres. The conserved Tpp1 residue Lys233 is important for Rnf8-mediated Tpp1 ubiquitylation and localization to telomeres. Thus, Tpp1 is a newly identified substrate for Rnf8, indicating a previously unrecognized role for Rnf8 in telomere end protection.


Asunto(s)
Telómero/química , Ubiquitina-Proteína Ligasas/fisiología , Animales , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas de los Mamíferos/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Ratones , Mapeo de Interacción de Proteínas , Estabilidad Proteica , Proteínas de Unión a Telómeros , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina-Proteína Ligasas/análisis , Ubiquitina-Proteína Ligasas/química , Ubiquitinación
15.
Methods Mol Biol ; 735: 145-50, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21461819

RESUMEN

Telomere dysfunctions, rendered through replicative attrition of telomeric DNA or due to the inhibition of shelterin components, are recognized as DNA double-stranded breaks (DSBs) by the DNA damage repair (DDR) pathway. This leads to the activation of DNA damage checkpoint sensors, including the Mre11-Rad50-Nbs1 (MRN) complex, γ-H2AX and 53BP1, the ATM and ATR signal-transducing kinases and downstream effectors, including Chk1, Chk2, and p53. Robust DNA damage response signals at dysfunctional telomeres, achieved by the complete deletion of TRF2 or by expressing dominant negative mutant TPP1(ΔRD), can be detected by their association with γ-H2AX and 53BP1 forming "telomere dysfunction induced foci (TIFs)." Induction of TIFs at telomeres provides an opportunity to quantify the extent of telomere dysfunction and monitor the signaling pathways.


Asunto(s)
Daño del ADN , Telómero/genética , Telómero/patología , Animales , ADN/genética , ADN/metabolismo , Reparación del ADN , Fibroblastos , Células HEK293 , Humanos , Hibridación Fluorescente in Situ , Ratones , Retroviridae/fisiología , Telómero/metabolismo , Tripeptidil Peptidasa 1
16.
Nature ; 471(7339): 532-6, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21399625

RESUMEN

Maintenance of telomeres requires both DNA replication and telomere 'capping' by shelterin. These two processes use two single-stranded DNA (ssDNA)-binding proteins, replication protein A (RPA) and protection of telomeres 1 (POT1). Although RPA and POT1 each have a critical role at telomeres, how they function in concert is not clear. POT1 ablation leads to activation of the ataxia telangiectasia and Rad3-related (ATR) checkpoint kinase at telomeres, suggesting that POT1 antagonizes RPA binding to telomeric ssDNA. Unexpectedly, we found that purified POT1 and its functional partner TPP1 are unable to prevent RPA binding to telomeric ssDNA efficiently. In cell extracts, we identified a novel activity that specifically displaces RPA, but not POT1, from telomeric ssDNA. Using purified protein, here we show that the heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) recapitulates the RPA displacing activity. The RPA displacing activity is inhibited by the telomeric repeat-containing RNA (TERRA) in early S phase, but is then unleashed in late S phase when TERRA levels decline at telomeres. Interestingly, TERRA also promotes POT1 binding to telomeric ssDNA by removing hnRNPA1, suggesting that the re-accumulation of TERRA after S phase helps to complete the RPA-to-POT1 switch on telomeric ssDNA. Together, our data suggest that hnRNPA1, TERRA and POT1 act in concert to displace RPA from telomeric ssDNA after DNA replication, and promote telomere capping to preserve genomic integrity.


Asunto(s)
ADN de Cadena Simple/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , ARN/metabolismo , Proteína de Replicación A/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/genética , Telómero/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Unión Competitiva , Proteínas de Ciclo Celular/metabolismo , Extractos Celulares , Replicación del ADN , Células HeLa , Ribonucleoproteína Nuclear Heterogénea A1 , Humanos , Unión Proteica , ARN/genética , Fase S , Complejo Shelterina
17.
Nat Struct Mol Biol ; 18(2): 213-21, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21217703

RESUMEN

Repressor activator protein 1 (RAP1) is the most highly conserved telomere protein. It is involved in protecting chromosome ends in fission yeast and promoting gene silencing in Saccharomyces cerevisiae, whereas it represses homology-directed recombination at telomeres in mammals. To understand how RAP1 has such diverse functions at telomeres, we solved the crystal or solution structures of the RAP1 C-terminal (RCT) domains of RAP1 from multiple organisms in complex with their respective protein-binding partners. Our analysis establishes RAP1(RCT) as an evolutionarily conserved protein-protein interaction module. In mammalian and fission yeast cells, this module interacts with TRF2 and Taz1, respectively, targeting RAP1 to chromosome ends for telomere protection. In contrast, S. cerevisiae RAP1 uses its RCT domain to recruit Sir3 to telomeres to mediate gene silencing. Together, our results show that, depending on the organism, the evolutionarily conserved RAP1 RCT motif has diverse functional roles at telomeres.


Asunto(s)
Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cristalografía por Rayos X , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Saccharomycetales/química , Saccharomycetales/genética , Saccharomycetales/metabolismo , Schizosaccharomyces/química , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Complejo Shelterina , Proteínas de Unión a Telómeros/genética , Proteína 2 de Unión a Repeticiones Teloméricas/química , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
18.
EMBO J ; 29(15): 2598-610, 2010 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-20588252

RESUMEN

Repair of DNA double-stranded breaks (DSBs) is crucial for the maintenance of genome stability. DSBs are repaired by either error prone non-homologous end-joining (NHEJ) or error-free homologous recombination. NHEJ precedes either by a classic, Lig4-dependent process (C-NHEJ) or an alternative, Lig4-independent one (A-NHEJ). Dysfunctional telomeres arising either through natural attrition due to telomerase deficiency or by removal of telomere-binding proteins are recognized as DSBs. In this report, we studied which end-joining pathways are required to join dysfunctional telomeres. In agreement with earlier studies, depletion of Trf2 resulted in end-to-end chromosome fusions mediated by the C-NHEJ pathway. In contrast, removal of Tpp1-Pot1a/b initiated robust chromosome fusions that are mediated by A-NHEJ. C-NHEJ is also dispensable for the fusion of naturally shortened telomeres. Our results reveal that telomeres engage distinct DNA repair pathways depending on how they are rendered dysfunctional, and that A-NHEJ is a major pathway to process dysfunctional telomeres.


Asunto(s)
Reparación del ADN , Telómero , Animales , Antígenos Nucleares/metabolismo , Células Cultivadas , Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Autoantígeno Ku , Ratones , Ratones Noqueados , Complejo Shelterina , Proteínas de Unión a Telómeros , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
19.
Cell Cycle ; 7(14): 2225-33, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18635967

RESUMEN

MDC1 and BRIT1 have been shown to function as key regulators in response to DNA damage. However, their roles in centrosomal regulation haven't been elucidated. In this study, we demonstrated the novel functions of these two molecules in regulating centrosome duplication and mitosis. We found that MDC1 and BRIT1 were integral components of the centrosome that colocalize with gamma-tubulin. Depletion of either protein led to centrosome amplification. However, the mechanisms that allow them to maintain centrosome integrity are different. MDC1-depleted cells exhibited centrosome overduplication, leading to multipolar mitosis, chromosome missegregation, and aneuploidy, whereas BRIT1 depletion led to misaligned spindles and/or lagging chromosomes with defective spindle checkpoint activation that resulted in defective cytokinesis and polyploidy. We further illustrated that both MDC1 and BRIT1 were negative regulators of Aurora A and Plk1, two centrosomal kinases involved in centrosome maturation and spindle assembly. Moreover, the levels of MDC1 and BRIT1 inversely correlated with centrosome amplification, defective mitosis and cancer metastasis in human breast cancer. Together, MDC1 and BRIT1 may function as tumor-suppressor genes, at least in part by orchestrating proper centrosome duplication and mitotic spindle assembly.


Asunto(s)
Centrosoma/metabolismo , Daño del ADN , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Aurora Quinasas , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Centrosoma/enzimología , Citocinesis , Proteínas del Citoesqueleto , Progresión de la Enfermedad , Femenino , Humanos , Mitosis , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Huso Acromático/enzimología , Quinasa Tipo Polo 1
20.
Cancer Genomics Proteomics ; 4(2): 99-106, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17804872

RESUMEN

One of the most common features of cancer is genetic instability. In response to numerous DNA-damaging insults, normal cells have evolved a complex mechanism to monitor and repair DNA damage lesions to maintain genomic integrity. The defects in DNA damage response, indeed, have been shown to associate closely with tumorigenesis. This review provides an overview on the molecular events in DNA damage signaling pathway, including cell cycle checkpoint and DNA repair. The recent research discoveries on how dysfunction in DNA damage response contributes to genomic instability and cancer development are also discussed.


Asunto(s)
Daño del ADN , Reparación del ADN , Neoplasias/genética , Neoplasias/patología , Animales , Ciclo Celular , Genoma/genética , Humanos , Neoplasias/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA