Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nat Rev Mol Cell Biol ; 17(6): 364-78, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27165790

RESUMO

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.


Assuntos
Reparo do DNA , Telômero/genética , Animais , Quebras de DNA de Cadeia Dupla , Instabilidade Genômica , Humanos , Transdução de Sinais , Homeostase do Telômero
2.
Genes Dev ; 34(23-24): 1619-1636, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33122293

RESUMO

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.


Assuntos
Replicação do DNA/genética , Poro Nuclear/patologia , Proteínas de Ligação a Telômeros/genética , Telômero/genética , Linhagem Celular Tumoral , Dano ao DNA/genética , Humanos , Mitose/genética , Mutação , Neoplasias/genética , Neoplasias/fisiopatologia , Complexo Shelterina , Telômero/metabolismo , Proteínas de Ligação a Telômeros/metabolismo
3.
Nature ; 589(7840): 110-115, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33239785

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes/metabolismo , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/deficiência , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Pluripotentes/citologia , Proteína 2 de Ligação a Repetições Teloméricas/genética , Células-Tronco Totipotentes/citologia , Células-Tronco Totipotentes/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
5.
Biochem Soc Trans ; 51(5): 1935-1946, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37767563

RESUMO

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.


Assuntos
Homeostase do Telômero , Telômero , Humanos , Recombinação Genética
6.
EMBO Rep ; 22(2): e50803, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33369867

RESUMO

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.


Assuntos
Doenças do Desenvolvimento Ósseo , Núcleo Celular/patologia , Anormalidades Craniofaciais , Hiperostose Cortical Congênita , Hipoparatireoidismo , Receptores Virais , Humanos , Vírus 40 dos Símios , Replicação Viral
7.
PLoS Pathog ; 16(4): e1008496, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32320442

RESUMO

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.


Assuntos
Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Integração Viral/genética , Linhagem Celular Tumoral , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Infecções por Roseolovirus/genética , Infecções por Roseolovirus/metabolismo , Infecções por Roseolovirus/virologia , Complexo Shelterina , Telômero/genética , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/genética
8.
Nature ; 518(7538): 254-7, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25642960

RESUMO

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.


Assuntos
Cromossomos de Mamíferos/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , DNA Polimerase Dirigida por DNA/metabolismo , Recombinação Genética , Telômero/genética , Telômero/metabolismo , Animais , Sequência de Bases , Morte Celular/genética , Linhagem Celular , Aberrações Cromossômicas , Cromossomos de Mamíferos/genética , DNA Polimerase Dirigida por DNA/deficiência , Genes BRCA1 , Genes BRCA2 , Células HeLa , Humanos , Camundongos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Rad51 Recombinase/metabolismo , Recombinação Genética/genética , Reparo de DNA por Recombinação/genética , Translocação Genética/genética , DNA Polimerase teta
9.
EMBO Rep ; 15(5): 601-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24714598

RESUMO

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.


Assuntos
Reparo do DNA , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestrutura , Nucleossomos/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/ultraestrutura , Motivos de Aminoácidos , Animais , Linhagem Celular , Proteínas Cromossômicas não Histona/metabolismo , Cristalografia por Raios X , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Técnicas de Inativação de Genes , Camundongos , Proteínas Nucleares/genética , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Telômero/efeitos dos fármacos , Telômero/genética , Proteína 2 de Ligação a Repetições Teloméricas/genética , Fatores de Transcrição/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
10.
Methods Cell Biol ; 182: 265-284, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38359982

RESUMO

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.


Assuntos
Ensaios de Triagem em Larga Escala , Neoplasias , Humanos , Animais , DNA , Telômero/genética , Peixes/genética
11.
Cancer Cell ; 5(6): 513-5, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15193252

RESUMO

The molecular mechanisms and the cell-of-origin leading to retinoblastoma are not well defined. In this issue of Cancer Cell, Bremner and colleagues describe the first inheritable model of retinoblastoma, revealing that loss of the pocket proteins pRb and p107 deregulates cell cycle exit in retinal precursors. The authors show that a subset of these precursors contain an inherent resistance to apoptosis, and that while most terminally differentiate, some are likely to acquire additional mutations, leading to tumor formation. Thus, this work defines the cell-of-origin of retinoblastoma and suggests that mutations giving increased proliferative capacity are required for retinoblastoma development.


Assuntos
Morte Celular , Neuroblastoma/patologia , Alelos , Animais , Apoptose , Diferenciação Celular , Divisão Celular , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Mutação , Neuroblastoma/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/fisiologia
12.
Trends Cell Biol ; 32(6): 527-536, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35063336

RESUMO

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.


Assuntos
Telomerase , Células-Tronco Embrionárias , Instabilidade Genômica , Humanos , Complexo Shelterina , Telomerase/química , Telomerase/genética , Telomerase/metabolismo , Telômero/metabolismo
13.
Elife ; 112022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-36373674

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona , Humanos , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteômica , Compartimentos de Replicação Viral
14.
Nat Commun ; 12(1): 4856, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381034

RESUMO

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.


Assuntos
Fator de Ligação a CCCTC/metabolismo , Reprogramação Celular , Células-Tronco Totipotentes/citologia , Animais , Sítios de Ligação , Fator de Ligação a CCCTC/genética , Morte Celular , Dano ao DNA , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Células-Tronco Totipotentes/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Mol Cell Biol ; 25(7): 2660-72, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15767672

RESUMO

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.


Assuntos
Envelhecimento/fisiologia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patologia , Hipófise/metabolismo , Hipófise/patologia , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proliferação de Células , Células Cultivadas , Proteínas de Ligação a DNA/genética , Fatores de Transcrição E2F , Fator de Transcrição E2F3 , Hiperplasia/genética , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/genética
16.
Mol Cell Biol ; 25(20): 8874-86, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16199867

RESUMO

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.


Assuntos
DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Animais , Apoptose , Sequência de Bases , Nucléolo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina , DNA/genética , Dano ao DNA , Estabilidade de Medicamentos , Feminino , Morte Fetal/genética , Morte Fetal/metabolismo , Morte Fetal/patologia , Genes p53 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Nucleofosmina , Gravidez , Proteína Supressora de Tumor p53/metabolismo
17.
Mol Cell Biol ; 22(15): 5527-38, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12101245

RESUMO

We report here the characterization of a mutant mouse line with a specific gene trap event in the Mdm4 locus. Absence of Mdm4 expression results in embryonic lethality (10.5 days postcoitum [dpc]), which was rescued by transferring the Mdm4 mutation into a Trp53-null background. Mutant embryos were characterized by overall growth deficiency, anemia, improper neural tube closure, and dilation of lateral ventricles. In situ analysis demonstrated increased levels of p21(CIP1/Waf1) and lower levels of Cyclin E and proliferating cell nuclear antigen expression. Consistent with lack of 5-bromo-2'-deoxyuridine incorporation, these data suggest a block of mutant embryo cells in the G(1) phase of the cell cycle. Accordingly, Mdm4-deficient mouse embryonic fibroblasts manifested a greatly reduced proliferative capacity in culture. Moreover, extensive p53-dependent cell death was specifically detected in the developing central nervous system of the Mdm4 mutant embryos. These findings unambiguously assign a critical role for Mdm4 as a negative regulator of p53 and suggest that Mdm4 could contribute to neoplasias retaining wild-type Trp53. Finally, we provide evidence indicating that Mdm4 plays no role on cell proliferation or cell cycle control that is distinct from its ability to modulate p53 function.


Assuntos
Anormalidades Múltiplas , Embrião de Mamíferos/anormalidades , Neurônios/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Anormalidades Múltiplas/genética , Animais , Bromodesoxiuridina , Morte Celular/genética , Divisão Celular/genética , Células Cultivadas , Ciclina E/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Fibroblastos/citologia , Fase G1 , Genes Letais , Ventrículos Laterais/anormalidades , Camundongos , Camundongos Mutantes , Defeitos do Tubo Neural/genética , Neurônios/patologia , Fenótipo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2 , Rombencéfalo/metabolismo , Rombencéfalo/patologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Ubiquitina-Proteína Ligases
18.
Science ; 355(6325): 638-641, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28082411

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Homeostase do Telômero , Telômero/metabolismo , Fatores de Transcrição/metabolismo , Dedos de Zinco , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Técnicas de Inativação de Genes , Humanos , Ligação Proteica , Sequências de Repetição em Tandem , Telômero/genética , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Fatores de Transcrição/genética
19.
Cell Rep ; 7(4): 1320-32, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24813883

RESUMO

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.


Assuntos
Cromatina/genética , Reparo do DNA por Junção de Extremidades , Complexo Repressor Polycomb 1/genética , Telômero/genética , Cromatina/isolamento & purificação , Cromatina/metabolismo , Dano ao DNA , Humanos , Complexo Repressor Polycomb 1/metabolismo , Telômero/química , Telômero/metabolismo
20.
Nat Struct Mol Biol ; 19(4): 387-94, 2012 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-22407014

RESUMO

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.


Assuntos
Pontos de Checagem do Ciclo Celular , Dano ao DNA , Mitose , Telômero/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Aurora Quinase B , Aurora Quinases , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Fase G1/efeitos dos fármacos , Humanos , Mitose/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Moduladores de Tubulina/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA