Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Cell Biol ; 182: 265-284, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38359982

RESUMEN

Alternative lengthening of telomeres (ALT) is a telomerase-independent and recombination-based mechanism used by approximately 15% of human cancers to maintain telomere length and to sustain proliferation. ALT-positive cells display unique features that could be exploited for tailored cancer therapies. A key limitation for the development of ALT-specific treatments is the lack of an assay to detect ALT-positive cells that is easy to perform and that can be scaled up. One of the most broadly used assays for ALT detection, CCA (C-circle assay), does not provide single-cell information and it is not amenable to High-Throughput Screening (HTS). To overcome these limitations, we developed Native-FISH (N-FISH) as an alternative method to visualize ALT-specific single-stranded telomeric DNA. N-FISH produces single-cell data, can be applied to fixed tissues, does not require DNA isolation or amplification steps, and it can be miniaturized in a 384-well format. This protocol details the steps to perform N-FISH protocol both in a low- and high-throughput format to analyze ALT. While low-throughput N-FISH is useful to assay the ALT state of cell lines, we expect that the miniaturized N-FISH assay coupled with high-throughput imaging will be useful in functional genomics and chemical screens to identify novel cellular factors that regulate ALT and potential ALT therapeutic targets for cancer therapies directed against ALT-positive tumors, respectively.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Neoplasias , Humanos , Animales , ADN , Telómero/genética , Peces/genética
2.
Biochem Soc Trans ; 51(5): 1935-1946, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37767563

RESUMEN

Approximately 15% of human cancers depend on the alternative lengthening of telomeres (ALT) pathway to maintain telomeres and proliferate. Telomeres that are elongated using ALT display unique features raising the exciting prospect of tailored cancer therapies. ALT-mediated telomere elongation shares several features with recombination-based DNA repair. Strikingly, cells that use the ALT pathway display abnormal levels of replication stress at telomeres and accumulate abundant extrachromosomal telomeric DNA. In this review, we examine recent findings that shed light on the ALT mechanisms and the strategies currently available to suppress this telomere elongation mechanism.


Asunto(s)
Homeostasis del Telómero , Telómero , Humanos , Recombinación Genética
3.
Elife ; 112022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-36373674

RESUMEN

The human SMC5/6 complex is a conserved guardian of genome stability and an emerging component of antiviral responses. These disparate functions likely require distinct mechanisms of SMC5/6 regulation. In yeast, Smc5/6 is regulated by its Nse5/6 subunits, but such regulatory subunits for human SMC5/6 are poorly defined. Here, we identify a novel SMC5/6 subunit called SIMC1 that contains SUMO interacting motifs (SIMs) and an Nse5-like domain. We isolated SIMC1 from the proteomic environment of SMC5/6 within polyomavirus large T antigen (LT)-induced subnuclear compartments. SIMC1 uses its SIMs and Nse5-like domain to localize SMC5/6 to polyomavirus replication centers (PyVRCs) at SUMO-rich PML nuclear bodies. SIMC1's Nse5-like domain binds to the putative Nse6 orthologue SLF2 to form an anti-parallel helical dimer resembling the yeast Nse5/6 structure. SIMC1-SLF2 structure-based mutagenesis defines a conserved surface region containing the N-terminus of SIMC1's helical domain that regulates SMC5/6 localization to PyVRCs. Furthermore, SLF1, which recruits SMC5/6 to DNA lesions via its BRCT and ARD motifs, binds SLF2 analogously to SIMC1 and forms a separate Nse5/6-like complex. Thus, two Nse5/6-like complexes with distinct recruitment domains control human SMC5/6 localization.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , Humanos , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteómica , Compartimentos de Replicación Viral
4.
Trends Cell Biol ; 32(6): 527-536, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35063336

RESUMEN

Telomeres are distinctive structures that protect the ends of linear chromosomes and ensure genome stability. They are composed of long tracks of repetitive and G-rich DNA that is bound by shelterin, a dedicated six-subunit protein complex. In somatic cells, shelterin protects telomeres from the DNA damage response and regulates telomere length. Telomere repeats are replenished by telomerase, a specialized ribonucleoprotein composed of telomerase reverse transcriptase and an integral RNA component. Telomere protection and telomerase regulation have been primarily studied in somatic cells. However, recent evidence points out striking differences in the context of embryonic stem cells (ESCs). In this review, we discuss insights into telomere protection in ESCs versus somatic cells and summarize findings on telomerase regulation as a function of pluripotency.


Asunto(s)
Telomerasa , Células Madre Embrionarias , Inestabilidad Genómica , Humanos , Complejo Shelterina , Telomerasa/química , Telomerasa/genética , Telomerasa/metabolismo , Telómero/metabolismo
6.
Nat Commun ; 12(1): 4856, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34381034

RESUMEN

Totipotent cells have the ability to generate embryonic and extra-embryonic tissues. Interestingly, a rare population of cells with totipotent-like potential, known as 2 cell (2C)-like cells, has been identified within ESC cultures. They arise from ESC and display similar features to those found in the 2C embryo. However, the molecular determinants of 2C-like conversion have not been completely elucidated. Here, we show that the CCCTC-binding factor (CTCF) is a barrier for 2C-like reprogramming. Indeed, forced conversion to a 2C-like state by the transcription factor DUX is associated with DNA damage at a subset of CTCF binding sites. Depletion of CTCF in ESC efficiently promotes spontaneous and asynchronous conversion to a 2C-like state and is reversible upon restoration of CTCF levels. This phenotypic reprogramming is specific to pluripotent cells as neural progenitor cells do not show 2C-like conversion upon CTCF-depletion. Furthermore, we show that transcriptional activation of the ZSCAN4 cluster is necessary for successful 2C-like reprogramming. In summary, we reveal an unexpected relationship between CTCF and 2C-like reprogramming.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Reprogramación Celular , Células Madre Totipotentes/citología , Animales , Sitios de Unión , Factor de Unión a CCCTC/genética , Muerte Celular , Daño del ADN , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Células Madre Totipotentes/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Nature ; 589(7840): 110-115, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33239785

RESUMEN

In mammals, telomere protection is mediated by the essential protein TRF2, which binds chromosome ends and ensures genome integrity1,2. TRF2 depletion results in end-to-end chromosome fusions in all cell types that have been tested so far. Here we find that TRF2 is dispensable for the proliferation and survival of mouse embryonic stem (ES) cells. Trf2-/- (also known as Terf2) ES cells do not exhibit telomere fusions and can be expanded indefinitely. In response to the deletion of TRF2, ES cells exhibit a muted DNA damage response that is characterized by the recruitment of γH2AX-but not 53BP1-to telomeres. To define the mechanisms that control this unique DNA damage response in ES cells, we performed a CRISPR-Cas9-knockout screen. We found a strong dependency of TRF2-null ES cells on the telomere-associated protein POT1B and on the chromatin remodelling factor BRD2. Co-depletion of POT1B or BRD2 with TRF2 restores a canonical DNA damage response at telomeres, resulting in frequent telomere fusions. We found that TRF2 depletion in ES cells activates a totipotent-like two-cell-stage transcriptional program that includes high levels of ZSCAN4. We show that the upregulation of ZSCAN4 contributes to telomere protection in the absence of TRF2. Together, our results uncover a unique response to telomere deprotection during early development.


Asunto(s)
Células Madre Pluripotentes/metabolismo , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/deficiencia , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/citología , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Células Madre Totipotentes/citología , Células Madre Totipotentes/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
8.
EMBO Rep ; 22(2): e50803, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33369867

RESUMEN

Mutations in the nuclear trypsin-like serine protease FAM111A cause Kenny-Caffey syndrome (KCS2) with hypoparathyroidism and skeletal dysplasia or perinatally lethal osteocraniostenosis (OCS). In addition, FAM111A was identified as a restriction factor for certain host range mutants of the SV40 polyomavirus and VACV orthopoxvirus. However, because FAM111A function is poorly characterized, its roles in restricting viral replication and the etiology of KCS2 and OCS remain undefined. We find that FAM111A KCS2 and OCS patient mutants are hyperactive and cytotoxic, inducing apoptosis-like phenotypes such as disruption of nuclear structure and pore distribution, in a protease-dependent manner. Moreover, wild-type FAM111A activity causes similar nuclear phenotypes, including the loss of nuclear barrier function, when SV40 host range mutants attempt to replicate in restrictive cells. Interestingly, pan-caspase inhibitors do not block these FAM111A-induced phenotypes, implying it acts independently or upstream of caspases. In this regard, we identify nucleoporins and the associated GANP transcription/replication factor as FAM111A interactors and candidate targets. Overall, we reveal a potentially unifying mechanism through which deregulated FAM111A activity restricts viral replication and causes KCS2 and OCS.


Asunto(s)
Enfermedades del Desarrollo Óseo , Núcleo Celular/patología , Anomalías Craneofaciales , Hiperostosis Cortical Congénita , Hipoparatiroidismo , Receptores Virales , Humanos , Virus 40 de los Simios , Replicación Viral
9.
Genes Dev ; 34(23-24): 1619-1636, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33122293

RESUMEN

Mutations in the telomere-binding protein POT1 are associated with solid tumors and leukemias. POT1 alterations cause rapid telomere elongation, ATR kinase activation, telomere fragility, and accelerated tumor development. Here, we define the impact of mutant POT1 alleles through complementary genetic and proteomic approaches based on CRISPR interference and biotin-based proximity labeling, respectively. These screens reveal that replication stress is a major vulnerability in cells expressing mutant POT1, which manifests as increased telomere mitotic DNA synthesis at telomeres. Our study also unveils a role for the nuclear pore complex in resolving replication defects at telomeres. Depletion of nuclear pore complex subunits in the context of POT1 dysfunction increases DNA damage signaling, telomere fragility and sister chromatid exchanges. Furthermore, we observed telomere repositioning to the nuclear periphery driven by nuclear F-actin polymerization in cells with POT1 mutations. In conclusion, our study establishes that relocalization of dysfunctional telomeres to the nuclear periphery is critical to preserve telomere repeat integrity.


Asunto(s)
Replicación del ADN/genética , Poro Nuclear/patología , Proteínas de Unión a Telómeros/genética , Telómero/genética , Línea Celular Tumoral , Daño del ADN/genética , Humanos , Mitosis/genética , Mutación , Neoplasias/genética , Neoplasias/fisiopatología , Complejo Shelterina , Telómero/metabolismo , Proteínas de Unión a Telómeros/metabolismo
10.
PLoS Pathog ; 16(4): e1008496, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32320442

RESUMEN

Human herpesviruses 6A and 6B (HHV-6A/B) are unique among human herpesviruses in their ability to integrate their genome into host chromosomes. Viral integration occurs at the ends of chromosomes within the host telomeres. The ends of the HHV-6A/B genomes contain telomeric repeats that facilitate the integration process. Here, we report that productive infections are associated with a massive increase in telomeric sequences of viral origin. The majority of the viral telomeric signals can be detected within viral replication compartments (VRC) that contain the viral DNA processivity factor p41 and the viral immediate-early 2 (IE2) protein. Components of the shelterin protein complex present at telomeres, including TRF1 and TRF2 are also recruited to VRC during infection. Biochemical, immunofluorescence coupled with in situ hybridization and chromatin immunoprecipitation demonstrated the binding of TRF2 to the HHV-6A/B telomeric repeats. In addition, approximately 60% of the viral IE2 protein localize at cellular telomeres during infection. Transient knockdown of TRF2 resulted in greatly reduced (13%) localization of IE2 at cellular telomeres (p<0.0001). Lastly, TRF2 knockdown reduced HHV-6A/B integration frequency (p<0.05), while no effect was observed on the infection efficiency. Overall, our study identified that HHV-6A/B IE2 localizes to telomeres during infection and highlight the role of TRF2 in HHV-6A/B infection and chromosomal integration.


Asunto(s)
Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Integración Viral/genética , Línea Celular Tumoral , ADN Viral/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Infecciones por Roseolovirus/genética , Infecciones por Roseolovirus/metabolismo , Infecciones por Roseolovirus/virología , Complejo Shelterina , Telómero/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
11.
Science ; 355(6325): 638-641, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28082411

RESUMEN

Telomeres are found at the end of chromosomes and are important for chromosome stability. Here we describe a specific telomere-associated protein: TZAP (telomeric zinc finger-associated protein). TZAP binds preferentially to long telomeres that have a low concentration of shelterin complex, competing with the telomeric-repeat binding factors TRF1 and TRF2. When localized at telomeres, TZAP triggers a process known as telomere trimming, which results in the rapid deletion of telomeric repeats. On the basis of these results, we propose a model for telomere length regulation in mammalian cells: The reduced concentration of the shelterin complex at long telomeres results in TZAP binding and initiation of telomere trimming. Binding of TZAP to long telomeres represents the switch that triggers telomere trimming, setting the upper limit of telomere length.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Homeostasis del Telómero , Telómero/metabolismo , Factores de Transcripción/metabolismo , Dedos de Zinc , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Técnicas de Inactivación de Genes , Humanos , Unión Proteica , Secuencias Repetidas en Tándem , Telómero/genética , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Factores de Transcripción/genética
12.
Nat Rev Mol Cell Biol ; 17(6): 364-78, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27165790

RESUMEN

Mammalian cells have evolved specialized mechanisms to sense and repair double-strand breaks (DSBs) to maintain genomic stability. However, in certain cases, the activity of these pathways can lead to aberrant DNA repair, genomic instability and tumorigenesis. One such case is DNA repair at the natural ends of linear chromosomes, known as telomeres, which can lead to chromosome-end fusions. Here, we review data obtained over the past decade and discuss the mechanisms that protect mammalian chromosome ends from the DNA damage response. We also discuss how telomere research has helped to uncover key steps in DSB repair. Last, we summarize how dysfunctional telomeres and the ensuing genomic instability drive the progression of cancer.


Asunto(s)
Reparación del ADN , Telómero/genética , Animales , Roturas del ADN de Doble Cadena , Inestabilidad Genómica , Humanos , Transducción de Señal , Homeostasis del Telómero
13.
Nature ; 518(7538): 254-7, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25642960

RESUMEN

The alternative non-homologous end-joining (NHEJ) machinery facilitates several genomic rearrangements, some of which can lead to cellular transformation. This error-prone repair pathway is triggered upon telomere de-protection to promote the formation of deleterious chromosome end-to-end fusions. Using next-generation sequencing technology, here we show that repair by alternative NHEJ yields non-TTAGGG nucleotide insertions at fusion breakpoints of dysfunctional telomeres. Investigating the enzymatic activity responsible for the random insertions enabled us to identify polymerase theta (Polθ; encoded by Polq in mice) as a crucial alternative NHEJ factor in mammalian cells. Polq inhibition suppresses alternative NHEJ at dysfunctional telomeres, and hinders chromosomal translocations at non-telomeric loci. In addition, we found that loss of Polq in mice results in increased rates of homology-directed repair, evident by recombination of dysfunctional telomeres and accumulation of RAD51 at double-stranded breaks. Lastly, we show that depletion of Polθ has a synergistic effect on cell survival in the absence of BRCA genes, suggesting that the inhibition of this mutagenic polymerase represents a valid therapeutic avenue for tumours carrying mutations in homology-directed repair genes.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN Polimerasa Dirigida por ADN/metabolismo , Recombinación Genética , Telómero/genética , Telómero/metabolismo , Animales , Secuencia de Bases , Muerte Celular/genética , Línea Celular , Aberraciones Cromosómicas , Cromosomas de los Mamíferos/genética , ADN Polimerasa Dirigida por ADN/deficiencia , Genes BRCA1 , Genes BRCA2 , Células HeLa , Humanos , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Recombinasa Rad51/metabolismo , Recombinación Genética/genética , Reparación del ADN por Recombinación/genética , Translocación Genética/genética , ADN Polimerasa theta
14.
Cell Rep ; 7(4): 1320-32, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24813883

RESUMEN

When telomeres become critically short, DNA damage response factors are recruited at chromosome ends, initiating a cellular response to DNA damage. We performed proteomic isolation of chromatin fragments (PICh) in order to define changes in chromatin composition that occur upon onset of acute telomere dysfunction triggered by depletion of the telomere-associated factor TRF2. This unbiased purification of telomere-associated proteins in functional or dysfunctional conditions revealed the dynamic changes in chromatin composition that take place at telomeres upon DNA damage induction. On the basis of our results, we describe a critical role for the polycomb group protein Ring1b in nonhomologous end-joining (NHEJ)-mediated end-to-end chromosome fusions. We show that cells with reduced levels of Ring1b have a reduced ability to repair uncapped telomeric chromatin. Our data represent an unbiased isolation of chromatin undergoing DNA damage and are a valuable resource to map the changes in chromatin composition in response to DNA damage activation.


Asunto(s)
Cromatina/genética , Reparación del ADN por Unión de Extremidades , Complejo Represivo Polycomb 1/genética , Telómero/genética , Cromatina/aislamiento & purificación , Cromatina/metabolismo , Daño del ADN , Humanos , Complejo Represivo Polycomb 1/metabolismo , Telómero/química , Telómero/metabolismo
15.
EMBO Rep ; 15(5): 601-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24714598

RESUMEN

The post-translational modification of DNA repair and checkpoint proteins by ubiquitin and small ubiquitin-like modifier (SUMO) critically orchestrates the DNA damage response (DDR). The ubiquitin ligase RNF4 integrates signaling by SUMO and ubiquitin, through its selective recognition and ubiquitination of SUMO-modified proteins. Here, we define a key new determinant for target discrimination by RNF4, in addition to interaction with SUMO. We identify a nucleosome-targeting motif within the RNF4 RING domain that can bind DNA and thereby enables RNF4 to selectively ubiquitinate nucleosomal histones. Furthermore, RNF4 nucleosome-targeting is crucially required for the repair of TRF2-depleted dysfunctional telomeres by 53BP1-mediated non-homologous end joining.


Asunto(s)
Reparación del ADN , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestructura , Nucleosomas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/ultraestructura , Secuencias de Aminoácidos , Animales , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Cristalografía por Rayos X , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Técnicas de Inactivación de Genes , Ratones , Proteínas Nucleares/genética , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Telómero/efectos de los fármacos , Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Factores de Transcripción/genética , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas , Ubiquitinación
16.
Nat Struct Mol Biol ; 19(4): 387-94, 2012 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-22407014

RESUMEN

Telomere shortening and disruption of telomeric components are pathways that induce telomere deprotection. Here we describe another pathway, in which prolonged mitotic arrest induces damage signals at telomeres in human cells. Exposure to microtubule drugs, kinesin inhibitors, proteasome inhibitors or the disruption of proper chromosome cohesion resulted in the formation of damage foci at telomeres. Induction of mitotic telomere deprotection coincided with dissociation of TRF2 from telomeres, telomeric 3'-overhang degradation and ATM activation, and deprotection could be suppressed by TRF2 overexpression or inhibition of Aurora B kinase. Normal cells that escaped from prolonged mitotic arrest halted in the following G1 phase, whereas cells lacking p53 continued to cycle and became aneuploid. We propose a telomere-dependent mitotic-duration monitoring system that reacts to improper progression through mitosis.


Asunto(s)
Puntos de Control del Ciclo Celular , Daño del ADN , Mitosis , Telómero/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Aurora Quinasa B , Aurora Quinasas , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Fase G1/efectos de los fármacos , Humanos , Mitosis/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Moduladores de Tubulina/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba
17.
Genes Dev ; 20(19): 2648-53, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17015429

RESUMEN

We report that mouse liver cells are highly resistant to extensive telomere dysfunction. In proliferating cells, telomere dysfunction results in chromosome end fusions, a DNA damage signal, and apoptosis or senescence. To determine the consequences of telomere dysfunction in noncycling cells, we used conditional deletion of the telomeric protein TRF2 in hepatocytes. TRF2 loss resulted in telomeric accumulation of gamma-H2AX and frequent telomere fusions, indicating telomere deprotection. However, there was no induction of p53 or apoptosis, and liver function appeared unaffected. Furthermore, the loss of TRF2 did not compromise liver regeneration after partial hepatectomy. Remarkably, liver regeneration occurred without cell division involving endoreduplication and cell growth, thereby circumventing the chromosome segregation problems associated with telomere fusions. We conclude that nondividing hepatocytes can maintain and regenerate liver function despite substantial loss of telomere integrity.


Asunto(s)
Hepatocitos/metabolismo , Regeneración Hepática/genética , Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Animales , Apoptosis/genética , Apoptosis/fisiología , Proliferación Celular , Senescencia Celular/genética , Senescencia Celular/fisiología , Daño del ADN , Femenino , Hepatocitos/citología , Hibridación Fluorescente in Situ/métodos , Hígado/patología , Hígado/fisiopatología , Hígado/cirugía , Regeneración Hepática/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
Mol Cell Biol ; 25(20): 8874-86, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16199867

RESUMEN

Nucleophosmin (NPM) is a nucleolar phosphoprotein that binds the tumor suppressors p53 and p19(Arf) and is thought to be indispensable for ribogenesis, cell proliferation, and survival after DNA damage. The NPM gene is the most frequent target of genetic alterations in leukemias and lymphomas, though its role in tumorigenesis is unknown. We report here the first characterization of a mouse NPM knockout strain. Lack of NPM expression results in accumulation of DNA damage, activation of p53, widespread apoptosis, and mid-stage embryonic lethality. Fibroblasts explanted from null embryos fail to grow and rapidly acquire a senescent phenotype. Transfer of the NPM mutation into a p53-null background rescued apoptosis in vivo and fibroblast proliferation in vitro. Cells null for both p53 and NPM grow faster than control cells and are more susceptible to transformation by activated oncogenes, such as mutated Ras or overexpressed Myc. In the absence of NPM, Arf protein is excluded from nucleoli and is markedly less stable. Our data demonstrate that NPM regulates DNA integrity and, through Arf, inhibits cell proliferation and are consistent with a putative tumor-suppressive function of NPM.


Asunto(s)
ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Animales , Apoptosis , Secuencia de Bases , Nucléolo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina , ADN/genética , Daño del ADN , Estabilidad de Medicamentos , Femenino , Muerte Fetal/genética , Muerte Fetal/metabolismo , Muerte Fetal/patología , Genes p53 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Nucleofosmina , Embarazo , Proteína p53 Supresora de Tumor/metabolismo
19.
EMBO Rep ; 6(7): 661-8, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15976820

RESUMEN

Loss of the retinoblastoma protein, pRB, leads to apoptosis, and several results have suggested that this is dependent on the E2F transcription factors. However, so far, the ability of the different E2F family members to contribute to apoptosis is controversial. Here, we show that ectopic expression of E2F3 results in apoptosis in both primary mouse fibroblasts and transgenic mice. Apoptosis induced by E2F3 is associated with the accumulation of E2F1 and, strikingly, we found that E2F3-induced apoptosis is dependent on E2F1. On the basis of these results, we propose that the accumulation of crucial levels of E2F1 activity, and not total E2F activity, is essential for the induction of apoptosis in response to a deregulated pRB pathway. These results are consistent with previous findings that E2F1, but not other E2Fs, can have tumour-suppressing activities.


Asunto(s)
Apoptosis , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular , Proteínas de Unión al ADN/genética , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Factor de Transcripción E2F3 , Fibroblastos/metabolismo , Ratones , Ratones Transgénicos , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Mol Cell Biol ; 25(7): 2660-72, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15767672

RESUMEN

The retinoblastoma gene, RB1, is one of the most frequently mutated genes in human cancer. Rb heterozygous mice develop pituitary tumors with 100% incidence, and the E2F transcription factors are required for this. To assess whether deregulated E2F activity is sufficient to induce pituitary tumors, we generated transgenic mice expressing an inducible E2F3 protein in the intermediate lobe of the pituitary gland. We found that short-term deregulation of E2F activity, similar to the earliest stages of Rb loss, is able to induce abnormal proliferation of otherwise quiescent melanotrophs. However, while long-term exposure to deregulated E2F activity results in hyperplasia of the intermediate lobe, it did not lead to tumor formation. In fact, melanotrophs become insensitive to sustained E2F stimulation and enter an irreversible senescence-like state. Thus, although deregulated E2F activity results in hyperproliferation, it is not sufficient to mimic loss of Rb, sustain proliferation of melanotrophs, and ultimately induce pituitary tumors. Similarly, we found that primary cells in tissue culture become insensitive to sustained E2F3 activation and undergo premature senescence in a pRB-, p16Ink4a-, and p19Arf-dependent manner. Thus, we conclude that deregulated E2F activity is not sufficient to fully mimic loss of Rb due to the engagement of a senescence response.


Asunto(s)
Envejecimiento/fisiología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patología , Hipófisis/metabolismo , Hipófisis/patología , Factores de Transcripción/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Factores de Transcripción E2F , Factor de Transcripción E2F3 , Hiperplasia/genética , Ratones , Ratones Transgénicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Transcripción/genética , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...