Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Nature ; 568(7753): 551-556, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30971823

RESUMEN

Synthetic lethality-an interaction between two genetic events through which the co-occurrence of these two genetic events leads to cell death, but each event alone does not-can be exploited for cancer therapeutics1. DNA repair processes represent attractive synthetic lethal targets, because many cancers exhibit an impairment of a DNA repair pathway, which can lead to dependence on specific repair proteins2. The success of poly(ADP-ribose) polymerase 1 (PARP-1) inhibitors in cancers with deficiencies in homologous recombination highlights the potential of this approach3. Hypothesizing that other DNA repair defects would give rise to synthetic lethal relationships, we queried dependencies in cancers with microsatellite instability (MSI), which results from deficient DNA mismatch repair. Here we analysed data from large-scale silencing screens using CRISPR-Cas9-mediated knockout and RNA interference, and found that the RecQ DNA helicase WRN was selectively essential in MSI models in vitro and in vivo, yet dispensable in models of cancers that are microsatellite stable. Depletion of WRN induced double-stranded DNA breaks and promoted apoptosis and cell cycle arrest selectively in MSI models. MSI cancer models required the helicase activity of WRN, but not its exonuclease activity. These findings show that WRN is a synthetic lethal vulnerability and promising drug target for MSI cancers.


Asunto(s)
Inestabilidad de Microsatélites , Repeticiones de Microsatélite/genética , Neoplasias/genética , Mutaciones Letales Sintéticas/genética , Helicasa del Síndrome de Werner/genética , Apoptosis/genética , Sistemas CRISPR-Cas/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Humanos , Modelos Genéticos , Neoplasias/patología , Interferencia de ARN , Proteína p53 Supresora de Tumor/metabolismo , Helicasa del Síndrome de Werner/deficiencia
3.
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
4.
Nucleic Acids Res ; 51(17): 9227-9247, 2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37560909

RESUMEN

Malignant cancers must activate telomere maintenance mechanisms to achieve replicative immortality. Mutations in the human Protection of Telomeres 1 (POT1) gene are frequently detected in cancers with abnormally long telomeres, suggesting that the loss of POT1 function disrupts the regulation of telomere length homeostasis to promote telomere elongation. However, our understanding of the mechanisms leading to elongated telomeres remains incomplete. The mouse genome encodes two POT1 proteins, POT1a and POT1b possessing separation of hPOT1 functions. We performed serial transplantation of Pot1b-/- sarcomas to better understand the role of POT1b in regulating telomere length maintenance. While early-generation Pot1b-/- sarcomas initially possessed shortened telomeres, late-generation Pot1b-/- cells display markedly hyper-elongated telomeres that were recognized as damaged DNA by the Replication Protein A (RPA) complex. The RPA-ATR-dependent DNA damage response at telomeres promotes telomerase recruitment to facilitate telomere hyper-elongation. POT1b, but not POT1a, was able to unfold G-quadruplex present in hyper-elongated telomeres to repress the DNA damage response. Our findings demonstrate that the repression of the RPA-ATR DDR is conserved between POT1b and human POT1, suggesting that similar mechanisms may underly the phenotypes observed in human cancers harboring human POT1 mutations.


Asunto(s)
Sarcoma , Complejo Shelterina , Ratones , Humanos , Animales , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Telómero/genética , Telómero/metabolismo , Daño del ADN , Proteína de Replicación A/metabolismo , Proteínas de Unión al ADN/genética
5.
Cell ; 135(1): 85-96, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18854157

RESUMEN

The Mre11/Rad50/NBS1 (MRN) complex maintains genomic stability by bridging DNA ends and initiating DNA damage signaling through activation of the ATM kinase. Mre11 possesses DNA nuclease activities that are highly conserved in evolution but play unknown roles in mammals. To define the functions of Mre11, we engineered targeted mouse alleles that either abrogate nuclease activities or inactivate the entire MRN complex. Mre11 nuclease deficiency causes a striking array of phenotypes indistinguishable from the absence of MRN, including early embryonic lethality and dramatic genomic instability. We identify a crucial role for the nuclease activities in homology-directed double-strand-break repair and a contributing role in activating the ATR kinase. However, the nuclease activities are not required to activate ATM after DNA damage or telomere deprotection. Therefore, nucleolytic processing by Mre11 is an essential function of fundamental importance in DNA repair, distinct from MRN control of ATM signaling.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Inestabilidad Genómica , Secuencia de Aminoácidos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular Transformada , Proliferación Celular , Roturas del ADN de Doble Cadena , Daño del ADN , Enzimas Reparadoras del ADN/química , Proteínas de Unión al ADN/química , Fibroblastos/metabolismo , Proteína Homóloga de MRE11 , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Recombinación Genética , Telómero/metabolismo , Proteínas Supresoras de Tumor/metabolismo
6.
Transpl Infect Dis ; 24(5): e13971, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36254507

RESUMEN

BACKGROUND: Antimicrobial stewardship in solid organ transplant (SOT) recipients is important to prevent antimicrobial-associated complications, but traditional stewardship principles are challenging to implement for SOT patients. Newer methodologies to optimize stewardship efforts are needed. METHODS: PubMed was searched using the keywords "cell free DNA," "metagenomic sequencing," "host biomarker," "antimicrobial stewardship," and "SOT." RESULTS: Metagenomic sequencing of cell free DNA has the potential to be a stewardship tool for SOT recipients. Various studies have shown its use for antimicrobial de-escalation and duration shortening. Host gene expression profiles can differentiate between infectious and noninfectious syndromes and may assist in stewardship efforts. However, information in immunocompromised hosts is conflicting. CONCLUSION: Microbial cell free DNA sequencing and host gene expression profiling show promise as stewardship tools in SOT recipients. Future studies on antimicrobial stewardship in SOT recipients should focus on their clinical use and feasibility.


Asunto(s)
Antiinfecciosos , Programas de Optimización del Uso de los Antimicrobianos , Ácidos Nucleicos Libres de Células , Trasplante de Órganos , Antibacterianos/uso terapéutico , Antiinfecciosos/uso terapéutico , Programas de Optimización del Uso de los Antimicrobianos/métodos , Biomarcadores , Humanos , Trasplante de Órganos/efectos adversos , Trasplante de Órganos/métodos , Receptores de Trasplantes
7.
Transpl Infect Dis ; 24(3): e13822, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35253965

RESUMEN

BACKGROUND: Mycoplasma hominis can cause significant infections after solid organ transplantation (SOT). Treatment should be guided by susceptibility testing, but conventional lab methods are laborious with prolonged turnaround time (TAT). This case series compares the phenotypic and genotypic susceptibility profiles of M. hominis isolates identified from SOT patients. METHODS: This is a single-center retrospective study evaluating SOT recipients with confirmed M. hominis infections. Patients' demographic, clinical, microbiological, and radiographic data were collected. Culture of M. hominis isolates was performed according to current Clinical and Laboratory Standards Institute guidelines. Phenotypic susceptibility testing was performed by University of Alabama Diagnostic Mycoplasma Laboratory. Whole genome sequencing (WGS) was performed followed by bioinformatic analysis of known genetic determinants of resistance. RESULTS: Seven SOT recipients with M. hominis infections were identified. Two out of seven (28.5%) patients had resistance detected by phenotypic susceptibility testing (Case 5 to levofloxacin and Case 7 to tetracycline). Genomic analyses confirmed the presence of mutations in the parC and parE topoisomerase genes at positions conferring to fluoroquinolone resistance in the isolate from Case 5, while the tetracycline-resistant isolate from Case 7 harbored the tetM gene. The median TAT from the date of specimen collection was 24 days for phenotypic susceptibility testing and 14 days for genotypic susceptibility testing. All seven patients received antimicrobials directed toward M. hominis and recovered with complete resolution of infection. CONCLUSIONS: WGS may offer a novel and more rapid methodology for M. hominis susceptibility testing to help optimize antimicrobial usage, but more data are needed.


Asunto(s)
Antiinfecciosos , Infecciones por Mycoplasma , Trasplante de Órganos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/uso terapéutico , Humanos , Pruebas de Sensibilidad Microbiana , Infecciones por Mycoplasma/diagnóstico , Infecciones por Mycoplasma/tratamiento farmacológico , Infecciones por Mycoplasma/microbiología , Mycoplasma hominis/genética , Trasplante de Órganos/efectos adversos , Estudios Retrospectivos , Tetraciclina/uso terapéutico , Resultado del Tratamiento
8.
Med Mycol ; 59(9): 939-942, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34143187

RESUMEN

Coccidioidal meningitis (CM) is a life-threatening infection with limited treatment options. Small series have reported success with isavuconazole; however, limited data exist on cerebrospinal fluid (CSF) penetration. Paired plasma and CSF isavuconazole concentrations were measured. Eleven CSF levels were tested, (7 ventricular, 4 lumbar) in three CM patients. Ventricular CSF levels were undetectable despite detectable plasma levels. All lumbar CSF levels were detectable (mean 1.00 µg/ml). Three pairs of lumbar CSF/plasma concentrations taken within 1 h of each other yielded a mean CSF/plasma ratio of 0.31. Isavuconazole was detectable in lumbar but not ventricular CSF in three patients treated for refractory CM. LAY SUMMARY: Isavuconazole is a triazole antifungal that has been used as salvage therapy in the treatment of coccidioidal meningitis (CM). Few data exist characterizing its concentration in the cerebrospinal fluid (CSF). We report tandem plasma and CSF concentrations of isavuconazole in three patients with CM.


Asunto(s)
Antifúngicos/uso terapéutico , Líquido Cefalorraquídeo/efectos de los fármacos , Coccidioidomicosis/tratamiento farmacológico , Meningitis Fúngica/tratamiento farmacológico , Plasma/efectos de los fármacos , Piridinas/uso terapéutico , Triazoles/uso terapéutico , Adulto , Anciano , Antifúngicos/farmacocinética , Monitoreo de Drogas , Femenino , Humanos , Masculino , Nitrilos/sangre , Nitrilos/líquido cefalorraquídeo , Nitrilos/farmacocinética , Nitrilos/uso terapéutico , Piridinas/sangre , Piridinas/líquido cefalorraquídeo , Resultado del Tratamiento , Triazoles/sangre , Triazoles/líquido cefalorraquídeo , Adulto Joven
9.
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
10.
Nature ; 471(7336): 119-23, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21368836

RESUMEN

Misrepair of DNA double-strand breaks produced by the V(D)J recombinase (the RAG1/RAG2 proteins) at immunoglobulin (Ig) and T cell receptor (Tcr) loci has been implicated in pathogenesis of lymphoid malignancies in humans and in mice. Defects in DNA damage response factors such as ataxia telangiectasia mutated (ATM) protein and combined deficiencies in classical non-homologous end joining and p53 predispose to RAG-initiated genomic rearrangements and lymphomagenesis. Although we showed previously that RAG1/RAG2 shepherd the broken DNA ends to classical non-homologous end joining for proper repair, roles for the RAG proteins in preserving genomic stability remain poorly defined. Here we show that the RAG2 carboxy (C) terminus, although dispensable for recombination, is critical for maintaining genomic stability. Thymocytes from 'core' Rag2 homozygotes (Rag2(c/c) mice) show dramatic disruption of Tcrα/δ locus integrity. Furthermore, all Rag2(c/c) p53(-/-) mice, unlike Rag1(c/c) p53(-/-) and p53(-/-) animals, rapidly develop thymic lymphomas bearing complex chromosomal translocations, amplifications and deletions involving the Tcrα/δ and Igh loci. We also find these features in lymphomas from Atm(-/-) mice. We show that, like ATM-deficiency, core RAG2 severely destabilizes the RAG post-cleavage complex. These results reveal a novel genome guardian role for RAG2 and suggest that similar 'end release/end persistence' mechanisms underlie genomic instability and lymphomagenesis in Rag2(c/c) p53(-/-) and Atm(-/-) mice.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Inestabilidad Genómica , Linfoma/genética , Linfoma/patología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Deleción Cromosómica , Cromosomas de los Mamíferos/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Reordenamiento Génico de Linfocito T/genética , Genes de las Cadenas Pesadas de las Inmunoglobulinas/genética , Genes p53/genética , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Ratones , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptores de Antígenos de Linfocitos T/genética , Recombinación Genética/genética , Timo/citología , Translocación Genética/genética , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
11.
Mol Cell ; 35(3): 352-64, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19683498

RESUMEN

Histone acetyltransferases (HATs) play important roles in gene regulation and DNA repair by influencing the accessibility of chromatin to transcription factors and repair proteins. Here, we show that deletion of Gcn5 leads to telomere dysfunction in mouse and human cells. Biochemical studies reveal that depletion of Gcn5 or ubiquitin-specific protease 22 (Usp22), which is another bona fide component of the Gcn5-containing SAGA complex, increases ubiquitination and turnover of TRF1, a primary component of the telomeric shelterin complex. Inhibition of the proteasome or overexpression of USP22 opposes this effect. The USP22 deubiquitinating module requires association with SAGA complexes for activity, and we find that depletion of Gcn5 compromises this association in mammalian cells. Thus, our results indicate that Gcn5 regulates TRF1 levels through effects on Usp22 activity and SAGA integrity.


Asunto(s)
Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Tioléster Hidrolasas/metabolismo , Factores de Transcripción p300-CBP/fisiología , Animales , Células Cultivadas , Aberraciones Cromosómicas , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Eliminación de Gen , Humanos , Ratones , Modelos Biológicos , Inhibidores de Proteasoma , Estabilidad Proteica , Complejo Shelterina , Proteínas de Unión a Telómeros/genética , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Tioléster Hidrolasas/genética , Ubiquitina Tiolesterasa , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
12.
Proc Natl Acad Sci U S A ; 111(30): E3062-71, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024194

RESUMEN

Tumor suppressor p53 plays an important role in mediating growth inhibition upon telomere dysfunction. Here, we show that loss of the p53 target gene cyclin-dependent kinase inhibitor 1A (CDKN1A, also known as p21(WAF1/CIP1)) increases apoptosis induction following telomerase inhibition in a variety of cancer cell lines and mouse xenografts. This effect is highly specific to p21, as loss of other checkpoint proteins and CDK inhibitors did not affect apoptosis. In telomerase, inhibited cell loss of p21 leads to E2F1- and p53-mediated transcriptional activation of p53-upregulated modulator of apoptosis, resulting in increased apoptosis. Combined genetic or pharmacological inhibition of telomerase and p21 synergistically suppresses tumor growth. Furthermore, we demonstrate that simultaneous inhibition of telomerase and p21 also suppresses growth of tumors containing mutant p53 following pharmacological restoration of p53 activity. Collectively, our results establish that inactivation of p21 leads to increased apoptosis upon telomerase inhibition and thus identify a genetic vulnerability that can be exploited to treat many human cancers containing either wild-type or mutant p53.


Asunto(s)
Apoptosis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Neoplasias Experimentales/metabolismo , Telomerasa/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Ratones , Ratones Desnudos , Mutación , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Telomerasa/genética , Telomerasa/metabolismo , Proteína p53 Supresora de Tumor/genética
13.
EMBO J ; 31(10): 2309-21, 2012 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-22531781

RESUMEN

The proper maintenance of telomeres is essential for genome stability. Mammalian telomere maintenance is governed by a number of telomere binding proteins, including the newly identified CTC1-STN1-TEN1 (CST) complex. However, the in vivo functions of mammalian CST remain unclear. To address this question, we conditionally deleted CTC1 from mice. We report here that CTC1 null mice experience rapid onset of global cellular proliferative defects and die prematurely from complete bone marrow failure due to the activation of an ATR-dependent G2/M checkpoint. Acute deletion of CTC1 does not result in telomere deprotection, suggesting that mammalian CST is not involved in capping telomeres. Rather, CTC1 facilitates telomere replication by promoting efficient restart of stalled replication forks. CTC1 deletion results in increased loss of leading C-strand telomeres, catastrophic telomere loss and accumulation of excessive ss telomere DNA. Our data demonstrate an essential role for CTC1 in promoting efficient replication and length maintenance of telomeres.


Asunto(s)
Replicación del ADN , Eliminación de Gen , Células Madre/fisiología , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Animales , Ratones , Ratones Noqueados
14.
J Neurosci ; 34(23): 7836-44, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24899707

RESUMEN

Genome stability is essential for neural development and the prevention of neurological disease. Here we determined how DNA damage signaling from dysfunctional telomeres affects neurogenesis. We found that telomere uncapping by Pot1a inactivation resulted in an Atm-dependent loss of cerebellar interneurons and granule neuron precursors in the mouse nervous system. The activation of Atm by Pot1a loss occurred in an Atr-dependent manner, revealing an Atr to Atm signaling axis in the nervous system after telomere dysfunction. In contrast to telomere lesions, Brca2 inactivation in neural progenitors also led to ablation of cerebellar interneurons, but this did not require Atm. These data reveal that neural cell loss after DNA damage selectively engages Atm signaling, highlighting how specific DNA lesions can dictate neuropathology arising in human neurodegenerative syndromes.


Asunto(s)
Daño del ADN/fisiología , Proteínas de Unión al ADN/fisiología , Neuronas/fisiología , Telómero/metabolismo , Animales , Animales Recién Nacidos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Encéfalo/citología , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Femenino , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Transgénicos , Nestina/genética , Complejo Shelterina , Proteínas de Unión a Telómeros , beta-Galactosidasa/metabolismo
15.
Nature ; 460(7257): 914-8, 2009 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-19633651

RESUMEN

Progressive telomere attrition or uncapping of the shelterin complex elicits a DNA damage response as a result of a cell's inability to distinguish dysfunctional telomeric ends from DNA double-strand breaks. Telomere deprotection activates both ataxia telangiectasia mutated (ATM) and telangiectasia and Rad3-related (ATR) kinase-dependent DNA damage response pathways, and promotes efficient non-homologous end-joining (NHEJ) of dysfunctional telomeres. The mammalian MRE11-RAD50-NBS1 (MRN; NBS1 is also known as NBN) complex interacts with ATM to sense chromosomal double-strand breaks and coordinate global DNA damage responses. Although the MRN complex accumulates at dysfunctional telomeres, it is not known whether mammalian MRN promotes repair at these sites. Here we address this question by using mouse alleles that either inactivate the entire MRN complex or eliminate only the nuclease activities of MRE11 (ref. 8). We show that cells lacking MRN do not activate ATM when telomeric repeat binding factor 2 (TRF2) is removed from telomeres, and ligase 4 (LIG4)-dependent chromosome end-to-end fusions are markedly reduced. Residual chromatid fusions involve only telomeres generated by leading strand synthesis. Notably, although cells deficient for MRE11 nuclease activity efficiently activate ATM and recruit 53BP1 (also known as TP53BP1) to deprotected telomeres, the 3' telomeric overhang persists to prevent NHEJ-mediated chromosomal fusions. Removal of shelterin proteins that protect the 3' overhang in the setting of MRE11 nuclease deficiency restores LIG4-dependent chromosome fusions. Our data indicate a critical role for the MRN complex in sensing dysfunctional telomeres, and show that in the absence of TRF2, MRE11 nuclease activity removes the 3' telomeric overhang to promote chromosome fusions. MRE11 can also protect newly replicated leading strand telomeres from NHEJ by promoting 5' strand resection to generate POT1a-TPP1-bound 3' overhangs.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Telómero/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Alelos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proteínas Cromosómicas no Histona , Aberraciones Cromosómicas , Daño del ADN , ADN Ligasa (ATP) , ADN Ligasas/metabolismo , Enzimas Reparadoras del ADN/deficiencia , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Fibroblastos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Homóloga de MRE11 , Ratones , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complejo Shelterina , Telómero/genética , Proteínas de Unión a Telómeros , Proteína 2 de Unión a Repeticiones Teloméricas/deficiencia , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
16.
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
17.
EMBO J ; 29(13): 2230-41, 2010 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-20551906

RESUMEN

Progressive telomere attrition or deficiency of the protective shelterin complex elicits a DNA damage response as a result of a cell's inability to distinguish dysfunctional telomeric ends from DNA double-strand breaks. SNMIB/Apollo is a shelterin-associated protein and a member of the SMN1/PSO2 nuclease family that localizes to telomeres through its interaction with TRF2. Here, we generated SNMIB/Apollo knockout mouse embryo fibroblasts (MEFs) to probe the function of SNMIB/Apollo at mammalian telomeres. SNMIB/Apollo null MEFs exhibit an increased incidence of G2 chromatid-type fusions involving telomeres created by leading-strand DNA synthesis, reflective of a failure to protect these telomeres after DNA replication. Mutations within SNMIB/Apollo's conserved nuclease domain failed to suppress this phenotype, suggesting that its nuclease activity is required to protect leading-strand telomeres. SNMIB/Apollo(-/-)ATM(-/-) MEFs display robust telomere fusions when Trf2 is depleted, indicating that ATM is dispensable for repair of uncapped telomeres in this setting. Our data implicate the 5'-3' exonuclease function of SNM1B/Apollo in the generation of 3' single-stranded overhangs at newly replicated leading-strand telomeres to protect them from engaging the non-homologous end-joining pathway.


Asunto(s)
Reparación del ADN , Fibroblastos/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Aminopeptidasas/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Cromosomas/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Embrión de Mamíferos/citología , Exodesoxirribonucleasas , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/metabolismo , Serina Proteasas/metabolismo , Complejo Shelterina , Proteínas de Unión a Telómeros/genética , Tripeptidil Peptidasa 1 , Proteínas Supresoras de Tumor/metabolismo
18.
Cancer Cell ; 9(2): 109-20, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16473278

RESUMEN

Mice deficient in the DNA damage sensor P53 display normal T cell development but eventually succumb to thymic lymphomas. Here, we show that inactivation of the TCR beta gene enhancer (E beta) results in a block of T cell development at stages where recombination-activating genes (RAG) are expressed. Introduction of the E beta mutation into p53-/- mice dramatically accelerates the onset of lethal thymic lymphomas that harbor RAG-dependent aberrant rearrangements, chromosome 14 and 12 translocations, and amplification of the chromosomal region 9A1-A5.3. Phenotypic and genetic analyses suggest that lymphomas emerge through a normal thymocyte development pathway. These findings provide genetic evidence that block of lymphocyte development at stages with RAG endonuclease activity can provoke lymphomagenesis on a background with deficient DNA damage responses.


Asunto(s)
Aberraciones Cromosómicas , Proteínas de Unión al ADN/metabolismo , Linfoma/genética , Linfoma/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Animales , Apoptosis , Línea Celular Tumoral , Daño del ADN , Proteínas de Unión al ADN/genética , Reordenamiento Génico de Linfocito T/genética , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/genética , Linfoma/inmunología , Linfoma/metabolismo , Ratones , Ratones Noqueados , Eliminación de Secuencia/genética , Cariotipificación Espectral , Linfocitos T/citología , Timo/citología , Timo/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
19.
Nature ; 452(7185): 365-9, 2008 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-18354482

RESUMEN

REST/NRSF (repressor-element-1-silencing transcription factor/neuron-restrictive silencing factor) negatively regulates the transcription of genes containing RE1 sites. REST is expressed in non-neuronal cells and stem/progenitor neuronal cells, in which it inhibits the expression of neuron-specific genes. Overexpression of REST is frequently found in human medulloblastomas and neuroblastomas, in which it is thought to maintain the stem character of tumour cells. Neural stem cells forced to express REST and c-Myc fail to differentiate and give rise to tumours in the mouse cerebellum. Expression of a splice variant of REST that lacks the carboxy terminus has been associated with neuronal tumours and small-cell lung carcinomas, and a frameshift mutant (REST-FS), which is also truncated at the C terminus, has oncogenic properties. Here we show, by using an unbiased screen, that REST is an interactor of the F-box protein beta-TrCP. REST is degraded by means of the ubiquitin ligase SCF(beta-TrCP) during the G2 phase of the cell cycle to allow transcriptional derepression of Mad2, an essential component of the spindle assembly checkpoint. The expression in cultured cells of a stable REST mutant, which is unable to bind beta-TrCP, inhibited Mad2 expression and resulted in a phenotype analogous to that observed in Mad2(+/-) cells. In particular, we observed defects that were consistent with faulty activation of the spindle checkpoint, such as shortened mitosis, premature sister-chromatid separation, chromosome bridges and mis-segregation in anaphase, tetraploidy, and faster mitotic slippage in the presence of a spindle inhibitor. An indistinguishable phenotype was observed by expressing the oncogenic REST-FS mutant, which does not bind beta-TrCP. Thus, SCF(beta-TrCP)-dependent degradation of REST during G2 permits the optimal activation of the spindle checkpoint, and consequently it is required for the fidelity of mitosis. The high levels of REST or its truncated variants found in certain human tumours may contribute to cellular transformation by promoting genomic instability.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Línea Celular , Fase G2 , Regulación de la Expresión Génica , Inestabilidad Genómica , Humanos , Proteínas Mad2 , Mitosis , Unión Proteica , Proteínas Represoras/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Huso Acromático/fisiología , Factores de Transcripción/genética , Proteínas con Repetición de beta-Transducina/deficiencia , Proteínas con Repetición de beta-Transducina/genética
20.
Clin Pharmacol Ther ; 2024 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-38973127

RESUMEN

The 2022 United States Food and Drug Administration (US FDA) draft guidance on diversity plan (DP), which will be implemented through the Diversity Action Plans by December 2025, under the 21st Century Cures Act, marks a pivotal effort by the FDA to ensure that registrational studies adequately reflect the target patient populations based on diversity in demographics and baseline characteristics. This white paper represents the culminated efforts of the International Consortium of Quality and Innovation (IQ) Diversity and Inclusion (D&I) Working Group (WG) to assess the implementation of the draft FDA guidance by members of the IQ consortium in the discipline of clinical pharmacology (CP). This article describes current practices in the industry and emphasizes the tools and techniques of quantitative pharmacology that can be applied to support the inclusion of a diverse population during global drug development, to support diversity and inclusion of underrepresented patient populations, in multiregional clinical trials (MRCTs). It outlines strategic and technical recommendations to integrate demographics, including age, sex/gender, race/ethnicity, and comorbidities, in multiregional phase III registrational studies, through the application of quantitative pharmacology. Finally, this article discusses the challenges faced during global drug development, which may otherwise limit the enrollment of a broader, potentially diverse population in registrational trials. Based on the outcomes of the IQ survey that provided the current awareness of diversity planning, it is envisioned that in the future, industry efforts in the inclusion of previously underrepresented populations during global drug development will culminate in drug labels that apply to the intended patient populations at the time of new drug application or biologics license application rather than through post-marketing requirements.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA