Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nature ; 522(7557): 492-6, 2015 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-26108857

RESUMEN

Tumour formation is blocked by two barriers: replicative senescence and crisis. Senescence is triggered by short telomeres and is bypassed by disruption of tumour-suppressive pathways. After senescence bypass, cells undergo crisis, during which almost all of the cells in the population die. Cells that escape crisis harbour unstable genomes and other parameters of transformation. The mechanism of cell death during crisis remains unexplained. Here we show that human cells in crisis undergo spontaneous mitotic arrest, resulting in death during mitosis or in the following cell cycle. This phenotype is induced by loss of p53 function, and is suppressed by telomerase overexpression. Telomere fusions triggered mitotic arrest in p53-compromised non-crisis cells, indicating that such fusions are the underlying cause of cell death. Exacerbation of mitotic telomere deprotection by partial TRF2 (also known as TERF2) knockdown increased the ratio of cells that died during mitotic arrest and sensitized cancer cells to mitotic poisons. We propose a crisis pathway wherein chromosome fusions induce mitotic arrest, resulting in mitotic telomere deprotection and cell death, thereby eliminating precancerous cells from the population.


Asunto(s)
Puntos de Control del Ciclo Celular , Muerte Celular , Aberraciones Cromosómicas , Mitosis , Neoplasias/patología , Telómero/metabolismo , Puntos de Control del Ciclo Celular/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular , Senescencia Celular , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , Daño del ADN , Fusión Génica/genética , Inestabilidad Genómica , Humanos , Mitosis/efectos de los fármacos , Mitosis/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/deficiencia , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Mol Cell ; 51(2): 141-55, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23850488

RESUMEN

Loss of chromosome end protection through telomere erosion is a hallmark of aging and senescence. Here we developed an experimental system that mimics physiological telomere deprotection in human cells and discovered that the telomere deprotection response is functionally distinct from the genomic DNA damage response. We found that, unlike genomic breaks, deprotected telomeres that are recognized as DNA damage but remain in the fusion-resistant intermediate state activate differential ataxia telangiectasia mutated (ATM) signaling where CHK2 is not phosphorylated. Also unlike genomic breaks, we found that deprotected telomeres do not contribute to the G2/M checkpoint and are instead passed through cell division to induce p53-dependent G1 arrest in the daughter cells. Telomere deprotection is therefore an epigenetic signal passed between cell generations to ensure that replication-associated telomere-dependent growth arrest occurs in stable diploid G1 phase cells before genome instability can occur.


Asunto(s)
División Celular/fisiología , Senescencia Celular/fisiología , Daño del ADN/genética , Replicación del ADN , Fase G2/fisiología , Genoma Humano , Telómero/fisiología , Western Blotting , Puntos de Control del Ciclo Celular , Proliferación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Mitosis/fisiología , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Nihon Rinsho ; 74(9): 1485-1490, 2016 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-30557481

RESUMEN

Telomeres are vital for chromosome end protection against activation of DNA damage response. Telomere attrition leads to cell cycle arrest, which underlies cellular senescence and can restrict tissue replenishment. Although stem cells express telomerase reverse tran- scriptase, which elongates telomeric DNA, its activity is not enough to fully compensate for chronic telomere shortening. Growing lines of evidence, including telomerase knockout mouse models and human genetic diseases, suggest that a decline in the cellular renewal capacity of stem cells is a consequence of such telomere shortening. These findings highlight the critical importance of telomere protection control for human aging process, and may lead to new strategies for healthy life extension.


Asunto(s)
Senescencia Celular , Telomerasa , Telómero , Animales , Senescencia Celular/genética , Humanos , Ratones , Telómero/metabolismo
4.
Life Sci Alliance ; 7(4)2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38307626

RESUMEN

Micronuclei (MN) have been associated with the innate immune response. The abrupt rupture of MN membranes results in the accumulation of cGAS, potentially activating STING and downstream interferon-responsive genes. However, direct evidence connecting MN and cGAS activation has been lacking. We have developed the FuVis2 reporter system, which enables the visualization of the cell nucleus carrying a single sister chromatid fusion and, consequently, MN. Using this FuVis2 reporter equipped with cGAS and STING reporters, we rigorously assessed the potency of cGAS activation by MN in individual living cells. Our findings reveal that cGAS localization to membrane-ruptured MN during interphase is infrequent, with cGAS primarily capturing MN during mitosis and remaining bound to cytosolic chromatin. We found that cGAS accumulation during mitosis neither activates STING in the subsequent interphase nor triggers the interferon response. Gamma-ray irradiation activates STING independently of MN formation and cGAS localization to MN. These results suggest that cGAS accumulation in cytosolic MN is not a robust indicator of its activation and that MN are not the primary trigger of the cGAS/STING pathway.


Asunto(s)
Interferón Tipo I , Transducción de Señal , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Inmunidad Innata , Interferón Tipo I/metabolismo , Núcleo Celular/metabolismo
5.
Sci Rep ; 13(1): 645, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36635307

RESUMEN

Telomeric ends form a loop structure (T-loop) necessary for the repression of ATM kinase activation throughout the normal cell cycle. However, cells undergoing a prolonged mitotic arrest are prone to lose the T-loop, resulting in Aurora B kinase-dependent mitotic telomere deprotection, which was proposed as an anti-tumor mechanism that eliminates precancerous cells from the population. The mechanism of mitotic telomere deprotection has not been elucidated. Here, we show that WRN, a RECQ helicase family member, can suppress mitotic telomere deprotection independently of its exonuclease and helicase activities. Truncation of WRN revealed that N-terminus amino acids 168-333, a region that contains a coiled-coil motif, is sufficient to suppress mitotic telomere deprotection without affecting both mitotic Aurora B-dependent spindle checkpoint and ATM kinase activity. The suppressive activity of the WRN168-333 fragment is diminished in cells partially depleted of TRF2, while WRN is required for complete suppression of mitotic telomere deprotection by TRF2 overexpression. Finally, we found that phosphomimetic but not alanine mutations of putative Aurora B target sites in the WRN168-333 fragment abolished its suppressive effect. Our findings reveal a non-enzymatic function of WRN, which may be regulated by phosphorylation in cells undergoing mitotic arrest. We propose that WRN enhances the protective function of TRF2 to counteract the hypothetical pathway that resolves the mitotic T-loop.


Asunto(s)
Exodesoxirribonucleasas , Proteína 2 de Unión a Repeticiones Teloméricas , Helicasa del Síndrome de Werner/genética , Helicasa del Síndrome de Werner/metabolismo , Exodesoxirribonucleasas/genética , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Telómero/genética , Telómero/metabolismo
6.
Chromosoma ; 120(1): 39-46, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20680317

RESUMEN

The initiation of DNA replication and the elongation of DNA strands take place in chromatin, a huge compound DNA-protein complex. Although the factors involved in the process of DNA replication have been largely elucidated, the underlying mechanisms that determine their behavior in the context of chromatin have only recently begun to be understood. It has been known that transcription is tightly regulated by the state of chromatin compaction, which governs the accessibility of DNA to trans-acting factors. This process is influenced by several determinants of chromatin structure, including intrinsic nucleosome positioning, the nucleosome remodeling complex, histone post-translational modifiers, and histone- and DNA-binding proteins. Growing evidence indicates that this concept is also applicable to the regulation of DNA replication. In addition, recent studies have demonstrated a distinctive mode of regulation. Some non-histone chromatin-binding proteins have been shown to interact physically with replication factors, thereby facilitating their recruitment at specific chromosomal loci. This type of regulation may allow control of local replication activity without affecting other chromosomal processes.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Replicación del ADN/fisiología , Nucleosomas/metabolismo , Animales , Histonas/metabolismo , Humanos , Procesamiento Proteico-Postraduccional/fisiología
8.
Life Sci Alliance ; 3(12)2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33106324

RESUMEN

Chromosome fusion is a frequent intermediate in oncogenic chromosome rearrangements and has been proposed to cause multiple tumor-driving abnormalities. In conventional experimental systems, however, these abnormalities were often induced by randomly induced chromosome fusions involving multiple different chromosomes. It was therefore not well understood whether a single defined type of chromosome fusion, which is reminiscent of a sporadic fusion in tumor cells, has the potential to cause chromosome instabilities. Here, we developed a human cell-based sister chromatid fusion visualization system (FuVis), in which a single defined sister chromatid fusion is induced by CRISPR/Cas9 concomitantly with mCitrine expression. The fused chromosome subsequently developed extra-acentric chromosomes, including chromosome scattering, indicative of chromothripsis. Live-cell imaging and statistical modeling indicated that sister chromatid fusion generated micronuclei (MN) in the first few cell cycles and that cells with MN tend to display cell cycle abnormalities. The powerful FuVis system thus demonstrates that even a single sporadic sister chromatid fusion can induce chromosome instability and destabilize the cell cycle through MN formation.


Asunto(s)
Inestabilidad Cromosómica/genética , Análisis de la Célula Individual/métodos , Intercambio de Cromátides Hermanas/fisiología , Sistemas CRISPR-Cas/genética , Ciclo Celular/genética , División Celular/genética , Cromátides/genética , Cromátides/patología , Cromátides/fisiología , Inestabilidad Cromosómica/fisiología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Ingeniería Genética/métodos , Células HCT116 , Humanos , Microscopía Fluorescente/métodos , Neoplasias/genética , Intercambio de Cromátides Hermanas/genética
9.
Nat Commun ; 10(1): 4224, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31530811

RESUMEN

Mitotic catastrophe is a broad descriptor encompassing unclear mechanisms of cell death. Here we investigate replication stress-driven mitotic catastrophe in human cells and identify that replication stress principally induces mitotic death signalled through two independent pathways. In p53-compromised cells we find that lethal replication stress confers WAPL-dependent centromere cohesion defects that maintain spindle assembly checkpoint-dependent mitotic arrest in the same cell cycle. Mitotic arrest then drives cohesion fatigue and triggers mitotic death through a primary pathway of BAX/BAK-dependent apoptosis. Simultaneously, a secondary mitotic death pathway is engaged through non-canonical telomere deprotection, regulated by TRF2, Aurora B and ATM. Additionally, we find that suppressing mitotic death in replication stressed cells results in distinct cellular outcomes depending upon how cell death is averted. These data demonstrate how replication stress-induced mitotic catastrophe signals cell death with implications for cancer treatment and cancer genome evolution.


Asunto(s)
Apoptosis , Proteínas Portadoras/metabolismo , Replicación del ADN , Mitosis , Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Telómero/metabolismo , Muerte Celular , Línea Celular Tumoral , Humanos , Neoplasias/genética , Neoplasias/fisiopatología , Telómero/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
10.
Genes Genet Syst ; 92(3): 107-118, 2018 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-28993556

RESUMEN

The ends of eukaryotic linear chromosomes are protected from undesired enzymatic activities by a nucleoprotein complex called the telomere. Expanding evidence indicates that telomeres have central functions in human aging and tumorigenesis. While it is undoubtedly important to follow current advances in telomere biology, it is also fruitful to be well informed in seminal historical studies for a comprehensive understanding of telomere biology, and for the anticipation of future directions. With this in mind, I here summarize the early history of telomere biology and current advances in the field, mostly focusing on mammalian studies relevant to aging and cancer.


Asunto(s)
Envejecimiento/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Telomerasa/metabolismo , Homeostasis del Telómero , Telómero/metabolismo , Envejecimiento/genética , Animales , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Proteínas de Neoplasias/genética , Neoplasias/genética , Telomerasa/genética , Telómero/genética
11.
Aging Cell ; 13(5): 946-50, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24863242

RESUMEN

Replicative senescence is a fundamental tumor-suppressive mechanism triggered by telomere erosion that results in a permanent cell cycle arrest. To understand the impact of telomere shortening on gene expression, we analyzed the transcriptome of diploid human fibroblasts as they progressed toward and entered into senescence. We distinguished novel transcription regulation due to replicative senescence by comparing senescence-specific expression profiles to profiles from cells arrested by DNA damage or serum starvation. Only a small specific subset of genes was identified that was truly senescence-regulated and changes in gene expression were exacerbated from presenescent to senescent cells. The majority of gene expression regulation in replicative senescence was shown to occur due to telomere shortening, as exogenous telomerase activity reverted most of these changes.


Asunto(s)
Senescencia Celular/genética , Regulación de la Expresión Génica , Línea Celular , Daño del ADN , Fibroblastos/citología , Fibroblastos/fisiología , Genómica , Humanos , Telomerasa/metabolismo
12.
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
13.
Nat Cell Biol ; 11(3): 357-62, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19182789

RESUMEN

Heterochromatin is a structurally compacted region of chromosomes in which transcription and recombination are inactivated. DNA replication is temporally regulated in heterochromatin, but the molecular mechanism for regulation has not been elucidated. Among heterochromatin loci in Schizosaccharomyces pombe, the pericentromeric region and the silent mating-type (mat) locus replicate in early S phase, whereas the sub-telomeric region does not, suggesting complex mechanisms for regulation of replication in heterochromatic regions. Here, we show that Swi6, an S. pombe counterpart of heterochromatin protein 1 (HP1), is required for early replication of the pericentromeric region and the mat locus. Origin-loading of Sld3, which depends on Dfp1/Dbf4-dependent kinase Cdc7 (DDK), is stimulated by Swi6. An HP1-binding motif within Dfp1 is required for interaction with Swi6 in vitro and for early replication of the pericentromeric region and mat locus. Tethering of Dfp1 to the pericentromeric region and mat locus in swi6-deficient cells restores early replication of these loci. Our results show that a heterochromatic protein positively regulates initiation of replication in silenced chromatin by interacting with an essential kinase.


Asunto(s)
Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Genes del Tipo Sexual de los Hongos , Heterocromatina/metabolismo , Origen de Réplica , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Secuencias de Aminoácidos , Homólogo de la Proteína Chromobox 5 , Momento de Replicación del ADN , Modelos Biológicos , Mutación Puntual/genética , Unión Proteica , Transporte de Proteínas , Fase S , Proteínas de Schizosaccharomyces pombe/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA