Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Mol Cell ; 81(24): 5007-5024.e9, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34767771

RESUMEN

As cells enter mitosis, chromatin compacts to facilitate chromosome segregation yet remains transcribed. Transcription supercoils DNA to levels that can impede further progression of RNA polymerase II (RNAPII) unless it is removed by DNA topoisomerase 1 (TOP1). Using ChIP-seq on mitotic cells, we found that TOP1 is required for RNAPII translocation along genes. The stimulation of TOP1 activity by RNAPII during elongation allowed RNAPII clearance from genes in prometaphase and enabled chromosomal segregation. Disruption of the TOP1-RNAPII interaction impaired RNAPII spiking at promoters and triggered defects in the post-mitotic transcription program. This program includes factors necessary for cell growth, and cells with impaired TOP1-RNAPII interaction are more sensitive to inhibitors of mTOR signaling. We conclude that TOP1 is necessary for assisting transcription during mitosis with consequences for growth and gene expression long after mitosis is completed. In this sense, TOP1 ensures that cellular memory is preserved in subsequent generations.


Asunto(s)
Proliferación Celular , Ensamble y Desensamble de Cromatina , Neoplasias Colorrectales/enzimología , ADN-Topoisomerasas de Tipo I/metabolismo , Fase G1 , Mitosis , ARN Polimerasa II/metabolismo , Transcripción Genética , Proliferación Celular/efectos de los fármacos , Secuenciación de Inmunoprecipitación de Cromatina , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , ADN-Topoisomerasas de Tipo I/genética , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Inhibidores mTOR/farmacología , Mitosis/efectos de los fármacos , ARN Polimerasa II/genética
2.
Nature ; 605(7910): 545-550, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35508652

RESUMEN

In preparation for mitotic cell division, the nuclear DNA of human cells is compacted into individualized, X-shaped chromosomes1. This metamorphosis is driven mainly by the combined action of condensins and topoisomerase IIα (TOP2A)2,3, and has been observed using microscopy for over a century. Nevertheless, very little is known about the structural organization of a mitotic chromosome. Here we introduce a workflow to interrogate the organization of human chromosomes based on optical trapping and manipulation. This allows high-resolution force measurements and fluorescence visualization of native metaphase chromosomes to be conducted under tightly controlled experimental conditions. We have used this method to extensively characterize chromosome mechanics and structure. Notably, we find that under increasing mechanical load, chromosomes exhibit nonlinear stiffening behaviour, distinct from that predicted by classical polymer models4. To explain this anomalous stiffening, we introduce a hierarchical worm-like chain model that describes the chromosome as a heterogeneous assembly of nonlinear worm-like chains. Moreover, through inducible degradation of TOP2A5 specifically in mitosis, we provide evidence that TOP2A has a role in the preservation of chromosome compaction. The methods described here open the door to a wide array of investigations into the structure and dynamics of both normal and disease-associated chromosomes.


Asunto(s)
Cromosomas Humanos , Cromosomas , Cromosomas/genética , Cromosomas/metabolismo , Cromosomas Humanos/metabolismo , ADN/química , ADN-Topoisomerasas de Tipo II/genética , Humanos , Mitosis , Óptica y Fotónica
3.
Cell ; 142(2): 230-42, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20655466

RESUMEN

Human telomeres are protected from DNA damage by a nucleoprotein complex that includes the repeat-binding factor TRF2. Here, we report that TRF2 regulates the 5' exonuclease activity of its binding partner, Apollo, a member of the metallo-beta-lactamase family that is required for telomere integrity during S phase. TRF2 and Apollo also suppress damage to engineered interstitial telomere repeat tracts that were inserted far away from chromosome ends. Genetic data indicate that DNA topoisomerase 2alpha acts in the same pathway of telomere protection as TRF2 and Apollo. Moreover, TRF2, which binds preferentially to positively supercoiled DNA substrates, together with Apollo, negatively regulates the amount of TOP1, TOP2alpha, and TOP2beta at telomeres. Our data are consistent with a model in which TRF2 and Apollo relieve topological stress during telomere replication. Our work also suggests that cellular senescence may be caused by topological problems that occur during the replication of the inner portion of telomeres.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Senescencia Celular , Daño del ADN , Exodesoxirribonucleasas , Humanos , Estructura Terciaria de Proteína
6.
J Biol Chem ; 295(20): 7138-7153, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32277049

RESUMEN

The double-helical structure of genomic DNA is both elegant and functional in that it serves both to protect vulnerable DNA bases and to facilitate DNA replication and compaction. However, these design advantages come at the cost of having to evolve and maintain a cellular machinery that can manipulate a long polymeric molecule that readily becomes topologically entangled whenever it has to be opened for translation, replication, or repair. If such a machinery fails to eliminate detrimental topological entanglements, utilization of the information stored in the DNA double helix is compromised. As a consequence, the use of B-form DNA as the carrier of genetic information must have co-evolved with a means to manipulate its complex topology. This duty is performed by DNA topoisomerases, which therefore are, unsurprisingly, ubiquitous in all kingdoms of life. In this review, we focus on how DNA topoisomerases catalyze their impressive range of DNA-conjuring tricks, with a particular emphasis on DNA topoisomerase III (TOP3). Once thought to be the most unremarkable of topoisomerases, the many lives of these type IA topoisomerases are now being progressively revealed. This research interest is driven by a realization that their substrate versatility and their ability to engage in intimate collaborations with translocases and other DNA-processing enzymes are far more extensive and impressive than was thought hitherto. This, coupled with the recent associations of TOP3s with developmental and neurological pathologies in humans, is clearly making us reconsider their undeserved reputation as being unexceptional enzymes.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , ADN Forma B/metabolismo , Enfermedades del Sistema Nervioso/enzimología , ADN-Topoisomerasas de Tipo I/genética , ADN Forma B/genética , Humanos , Enfermedades del Sistema Nervioso/genética
7.
Am J Hum Genet ; 103(2): 221-231, 2018 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-30057030

RESUMEN

Bloom syndrome, caused by biallelic mutations in BLM, is characterized by prenatal-onset growth deficiency, short stature, an erythematous photosensitive malar rash, and increased cancer predisposition. Diagnostically, a hallmark feature is the presence of increased sister chromatid exchanges (SCEs) on cytogenetic testing. Here, we describe biallelic mutations in TOP3A in ten individuals with prenatal-onset growth restriction and microcephaly. TOP3A encodes topoisomerase III alpha (TopIIIα), which binds to BLM as part of the BTRR complex, and promotes dissolution of double Holliday junctions arising during homologous recombination. We also identify a homozygous truncating variant in RMI1, which encodes another component of the BTRR complex, in two individuals with microcephalic dwarfism. The TOP3A mutations substantially reduce cellular levels of TopIIIα, and consequently subjects' cells demonstrate elevated rates of SCE. Unresolved DNA recombination and/or replication intermediates persist into mitosis, leading to chromosome segregation defects and genome instability that most likely explain the growth restriction seen in these subjects and in Bloom syndrome. Clinical features of mitochondrial dysfunction are evident in several individuals with biallelic TOP3A mutations, consistent with the recently reported additional function of TopIIIα in mitochondrial DNA decatenation. In summary, our findings establish TOP3A mutations as an additional cause of prenatal-onset short stature with increased cytogenetic SCEs and implicate the decatenation activity of the BTRR complex in their pathogenesis.

8.
Nucleic Acids Res ; 46(2): 861-872, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29253195

RESUMEN

DNA topoisomerases are essential enzymes involved in all the DNA processes and among them, type IA topoisomerases emerged as a key actor in the maintenance of genome stability. The hyperthermophilic archaeon, Sulfolobus solfataricus, contains three topoisomerases IA including one classical named TopA. SsoTopA is very efficient at unlinking DNA catenanes, grouping SsoTopA into the topoisomerase III family. SsoTopA is active over a wide range of temperatures and at temperatures of up to 85°C it produces highly unwound DNA. At higher temperatures, SsoTopA unlinks the two DNA strands. Thus depending on the temperature, SsoTopA is able to either prevent or favor DNA melting. While canonical topoisomerases III require a single-stranded DNA region or a nick in one of the circles to decatenate them, we show for the first time that a type I topoisomerase, SsoTopA, is able to efficiently unlink covalently closed catenanes, with no additional partners. By using single molecule experiments we demonstrate that SsoTopA requires the presence of a short single-stranded DNA region to be efficient. The unexpected decatenation property of SsoTopA probably comes from its high ability to capture this unwound region. This points out a possible role of TopA in S. solfataricus as a decatenase in Sulfolobus.


Asunto(s)
Proteínas Arqueales/metabolismo , ADN-Topoisomerasas de Tipo I/metabolismo , ADN Encadenado/metabolismo , Sulfolobus solfataricus/enzimología , Proteínas Arqueales/genética , Secuencia de Bases , ADN-Topoisomerasas de Tipo I/genética , ADN de Archaea/química , ADN de Archaea/genética , ADN de Archaea/metabolismo , ADN Encadenado/química , ADN Encadenado/genética , ADN Concatenado/química , ADN Concatenado/genética , ADN Concatenado/metabolismo , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Calor , Cinética , Modelos Moleculares , Conformación de Ácido Nucleico , Sulfolobus solfataricus/genética
9.
Nucleic Acids Res ; 45(19): 11413-11424, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28977671

RESUMEN

PICH is a DNA translocase required for the maintenance of chromosome stability in human cells. Recent data indicate that PICH co-operates with topoisomerase IIα to suppress pathological chromosome missegregation through promoting the resolution of ultra-fine anaphase bridges (UFBs). Here, we identify the BEN domain-containing protein 3 (BEND3) as an interaction partner of PICH in human cells in mitosis. We have purified full length PICH and BEND3 and shown that they exhibit a functional biochemical interaction in vitro. We demonstrate that the PICH-BEND3 interaction occurs via a novel interface between a TPR domain in PICH and a BEN domain in BEND3, and have determined the crystal structure of this TPR-BEN complex at 2.2 Å resolution. Based on the structure, we identified amino acids important for the TPR-BEN domain interaction, and for the functional interaction of the full-length proteins. Our data reveal a proposed new function for BEND3 in association with PICH, and the first example of a specific protein-protein interaction mediated by a BEN domain.


Asunto(s)
Secuencias de Aminoácidos , ADN Helicasas/química , Dominios Proteicos , Proteínas Represoras/química , Secuencia de Aminoácidos , Sitios de Unión/genética , Cristalografía por Rayos X , ADN Helicasas/genética , ADN Helicasas/metabolismo , Células HEK293 , Células HeLa , Humanos , Mitosis/genética , Modelos Moleculares , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Homología de Secuencia de Aminoácido
10.
Nucleic Acids Res ; 43(10): 5221-35, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25901030

RESUMEN

Bloom's syndrome helicase (BLM) is a member of the RecQ family of DNA helicases, which play key roles in the maintenance of genome integrity in all organism groups. We describe crystal structures of the BLM helicase domain in complex with DNA and with an antibody fragment, as well as SAXS and domain association studies in solution. We show an unexpected nucleotide-dependent interaction of the core helicase domain with the conserved, poorly characterized HRDC domain. The BLM-DNA complex shows an unusual base-flipping mechanism with unique positioning of the DNA duplex relative to the helicase core domains. Comparison with other crystal structures of RecQ helicases permits the definition of structural transitions underlying ATP-driven helicase action, and the identification of a nucleotide-regulated tunnel that may play a role in interactions with complex DNA substrates.


Asunto(s)
RecQ Helicasas/química , Adenosina Difosfato/química , Cristalografía por Rayos X , ADN/química , Modelos Moleculares , Mutación , Conformación Proteica , Estructura Terciaria de Proteína , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Anticuerpos de Dominio Único/química , Zinc/química
12.
BMC Mol Biol ; 15: 18, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25200003

RESUMEN

BACKGROUND: Reverse gyrases are DNA topoisomerases characterized by their unique DNA positive-supercoiling activity. Sulfolobus solfataricus, like most Crenarchaeota, contains two genes each encoding a reverse gyrase. We showed previously that the two genes are differently regulated according to temperature and that the corresponding purified recombinant reverse gyrases have different enzymatic characteristics. These observations suggest a specialization of functions of the two reverse gyrases. As no mutants of the TopR genes could be obtained in Sulfolobales, we used immunodetection techniques to study the function(s) of these proteins in S. solfataricus in vivo. In particular, we investigated whether one or both reverse gyrases are required for the hyperthermophilic lifestyle. RESULTS: For the first time the two reverse gyrases of S. solfataricus have been discriminated at the protein level and their respective amounts have been determined in vivo. Actively dividing S. solfataricus cells contain only small amounts of both reverse gyrases, approximately 50 TopR1 and 125 TopR2 molecules per cell at 80°C. S. solfataricus cells are resistant at 45°C for several weeks, but there is neither cell division nor replication initiation; these processes are fully restored upon a return to 80°C. TopR1 is not found after three weeks at 45°C whereas the amount of TopR2 remains constant. Enzymatic assays in vitro indicate that TopR1 is not active at 45°C but that TopR2 exhibits highly positive DNA supercoiling activity at 45°C. CONCLUSIONS: The two reverse gyrases of S. solfataricus are differently regulated, in terms of protein abundance, in vivo at 80°C and 45°C. TopR2 is present both at high and low temperatures and is therefore presumably required whether cells are dividing or not. By contrast, TopR1 is present only at high temperature where the cell division occurs, suggesting that TopR1 is required for controlling DNA topology associated with cell division activity and/or life at high temperature. Our findings in vitro that TopR1 is able to positively supercoil DNA only at high temperature, and TopR2 is active at both temperatures are consistent with them having different functions within the cells.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , Sulfolobus solfataricus/citología , Sulfolobus solfataricus/enzimología , Secuencia de Aminoácidos , ADN-Topoisomerasas de Tipo I/análisis , ADN Superhelicoidal/metabolismo , Calor , Datos de Secuencia Molecular , Sulfolobus solfataricus/química , Sulfolobus solfataricus/fisiología
13.
J Biol Chem ; 287(36): 30282-95, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22722926

RESUMEN

RecQ family helicases and topoisomerase 3 enzymes form evolutionary conserved complexes that play essential functions in DNA replication, recombination, and repair, and in vitro, show coordinate activities on model recombination and replication intermediates. Malfunctioning of these complexes in humans is associated with genomic instability and cancer-prone syndromes. Although both RecQ-like and topoisomerase 3 enzymes are present in archaea, only a few of them have been studied, and no information about their functional interaction is available. We tested the combined activities of the RecQ-like helicase, Hel112, and the topoisomerase 3, SsTop3, from the thermophilic archaeon Sulfolobus solfataricus. Hel112 showed coordinate DNA unwinding and annealing activities, a feature shared by eukaryotic RecQ homologs, which resulted in processing of synthetic Holliday junctions and stabilization of model replication forks. SsTop3 catalyzed DNA relaxation and annealing. When assayed in combination, SsTop3 inhibited the Hel112 helicase activity on Holliday junctions and stimulated formation and stabilization of such structures. In contrast, Hel112 did not affect the SsTop3 DNA relaxation activity. RecQ-topoisomerase 3 complexes show structural similarity with the thermophile-specific enzyme reverse gyrase, which catalyzes positive supercoiling of DNA and was suggested to play a role in genome stability at high temperature. Despite such similarity and the high temperature of reaction, the SsTop3-Hel112 complex does not induce positive supercoiling and is thus likely to play different roles. We propose that the interplay between Hel112 and SsTop3 might regulate the equilibrium between recombination and anti-recombination activities at replication forks.


Asunto(s)
Proteínas Arqueales/metabolismo , Replicación del ADN/fisiología , ADN-Topoisomerasas de Tipo I/metabolismo , ADN de Archaea/biosíntesis , ADN Cruciforme/metabolismo , RecQ Helicasas/metabolismo , Sulfolobus solfataricus/enzimología , Proteínas Arqueales/genética , ADN-Topoisomerasas de Tipo I/genética , ADN de Archaea/genética , ADN Cruciforme/genética , RecQ Helicasas/genética , Sulfolobus solfataricus/genética
14.
Nat Struct Mol Biol ; 30(9): 1303-1313, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37474739

RESUMEN

SUMOylation regulates numerous cellular processes, but what represents the essential functions of this protein modification remains unclear. To address this, we performed genome-scale CRISPR-Cas9-based screens, revealing that the BLM-TOP3A-RMI1-RMI2 (BTRR)-PICH pathway, which resolves ultrafine anaphase DNA bridges (UFBs) arising from catenated DNA structures, and the poorly characterized protein NIP45/NFATC2IP become indispensable for cell proliferation when SUMOylation is inhibited. We demonstrate that NIP45 and SUMOylation orchestrate an interphase pathway for converting DNA catenanes into double-strand breaks (DSBs) that activate the G2 DNA-damage checkpoint, thereby preventing cytokinesis failure and binucleation when BTRR-PICH-dependent UFB resolution is defective. NIP45 mediates this new TOP2-independent DNA catenane resolution process via its SUMO-like domains, promoting SUMOylation of specific factors including the SLX4 multi-nuclease complex, which contributes to catenane conversion into DSBs. Our findings establish that SUMOylation exerts its essential role in cell proliferation by enabling resolution of toxic DNA catenanes via nonepistatic NIP45- and BTRR-PICH-dependent pathways to prevent mitotic failure.


Asunto(s)
Anafase , ADN Encadenado , ADN , Sumoilación
15.
Nat Commun ; 13(1): 584, 2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35102151

RESUMEN

Topoisomerase IIIα is a type 1A topoisomerase that forms a complex with RMI1 and RMI2 called TRR in human cells. TRR plays an essential role in resolving DNA replication and recombination intermediates, often alongside the helicase BLM. While the TRR catalytic cycle is known to involve a protein-mediated single-stranded (ss)DNA gate, the detailed mechanism is not fully understood. Here, we probe the catalytic steps of TRR using optical tweezers and fluorescence microscopy. We demonstrate that TRR forms an open gate in ssDNA of 8.5 ± 3.8 nm, and directly visualize binding of a second ssDNA or double-stranded (ds)DNA molecule to the open TRR-ssDNA gate, followed by catenation in each case. Strikingly, dsDNA binding increases the gate size (by ~16%), while BLM alters the mechanical flexibility of the gate. These findings reveal an unexpected plasticity of the TRR-ssDNA gate size and suggest that TRR-mediated transfer of dsDNA may be more relevant in vivo than previously believed.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , RecQ Helicasas/metabolismo , Biocatálisis , Escherichia coli/enzimología , Proteínas de Escherichia coli/metabolismo , Fluorescencia , Humanos , Magnesio/metabolismo , Especificidad por Sustrato
16.
Nat Struct Mol Biol ; 26(4): 267-274, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30936532

RESUMEN

All known eukaryotic topoisomerases are only able to relieve torsional stress in DNA. Nevertheless, it has been proposed that the introduction of positive DNA supercoiling is required for efficient sister-chromatid disjunction by Topoisomerase 2a during mitosis. Here we identify a eukaryotic enzymatic activity that introduces torsional stress into DNA. We show that the human Plk1-interacting checkpoint helicase (PICH) and Topoisomerase 3a proteins combine to create an extraordinarily high density of positive DNA supercoiling. This activity, which is analogous to that of a reverse-gyrase, is apparently driven by the ability of PICH to progressively extrude hypernegatively supercoiled DNA loops that are relaxed by Topoisomerase 3a. We propose that this positive supercoiling provides an optimal substrate for the rapid disjunction of sister centromeres by Topoisomerase 2a at the onset of anaphase in eukaryotic cells.


Asunto(s)
ADN Helicasas/metabolismo , ADN-Topoisomerasas de Tipo I/química , ADN-Topoisomerasas de Tipo I/metabolismo , ADN/química , ADN/metabolismo , Cromátides/metabolismo , ADN Helicasas/química , ADN-Topoisomerasas de Tipo II/metabolismo , ADN Superhelicoidal/química , ADN Superhelicoidal/metabolismo , Humanos
17.
J Cell Biol ; 218(12): 3943-3953, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31615875

RESUMEN

The ATR kinase is a master regulator of the cellular response to DNA replication stress. Activation of ATR relies on dual pathways involving the TopBP1 and ETAA1 proteins, both of which harbor ATR-activating domains (AADs). However, the exact contribution of the recently discovered ETAA1 pathway to ATR signaling in different contexts remains poorly understood. Here, using an unbiased CRISPR-Cas9-based genome-scale screen, we show that the ATR-stimulating function of ETAA1 becomes indispensable for cell fitness and chromosome stability when the fidelity of DNA replication is compromised. We demonstrate that the ATR-activating potential of ETAA1 is controlled by cell cycle- and replication stress-dependent phosphorylation of highly conserved residues within its AAD, and that the stimulatory impact of these modifications is required for the ability of ETAA1 to prevent mitotic chromosome abnormalities following replicative stress. Our findings suggest an important role of ETAA1 in protecting against genome instability arising from incompletely duplicated DNA via regulatory control of its ATR-stimulating potential.


Asunto(s)
Antígenos de Superficie/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Replicación del ADN , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Sistemas CRISPR-Cas , Ciclo Celular , Línea Celular Tumoral , Aberraciones Cromosómicas , Daño del ADN , Genoma Humano , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Mitosis , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica , Transducción de Señal
18.
Curr Opin Cell Biol ; 52: 112-119, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29525475

RESUMEN

The anaphase of mitosis is one of the most critical stages of the cell division cycle in that it can reveal precious information on the fate of a cell lineage. Indeed, most types of nuclear DNA segregation defects visualized during anaphase are manifestations of genomic instability and augur dramatic outcomes, such as cell death or chromosomal aberrations characteristic of cancer cells. Although chromatin bridges and lagging chromatin are always pathological (generating aneuploidy or complex genomic rearrangements), the main subject of this article, the ultrafine anaphase bridges, might, in addition to potentially driving genomic instability, play critical roles for the maintenance of chromosome structure in rapidly proliferating cells.


Asunto(s)
Anafase/genética , Inestabilidad Genómica/genética , Humanos
19.
Methods Mol Biol ; 1672: 495-508, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29043644

RESUMEN

Ultrafine anaphase bridges (UFBs) are thin DNA threads linking the separating sister chromatids in the anaphase of mitosis. UFBs are thought to form when topological DNA entanglements between two chromatids are not resolved prior to anaphase onset. In contrast to other markers of defective chromosome segregation, UFBs cannot be detected by direct staining of the DNA, but instead can be detected using immunofluorescence-based approaches. Due to the fact that they are short-lived and fragile in nature, UFBs can be challenging to detect. In this chapter, we describe methods that have been optimized for successful detection of UFBs. We also provide guidelines for the optimization of UFBs detection depending on the antibody and the cell line to be used.


Asunto(s)
Anafase/genética , Cromátides/genética , ADN , Línea Celular Tumoral , Centrómero/genética , Sitios Frágiles del Cromosoma , Segregación Cromosómica , Técnica del Anticuerpo Fluorescente , Inestabilidad Genómica , Humanos , Microscopía Fluorescente , Telómero/genética
20.
Cell Rep ; 24(12): 3274-3284, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30232008

RESUMEN

PICH is a DNA translocase necessary for the resolution of ultrafine anaphase DNA bridges and to ensure the fidelity of chromosomal segregation. Here, we report the generation of an animal model deficient for PICH that allowed us to investigate its physiological relevance. Pich KO mice lose viability during embryonic development due to a global accumulation of DNA damage. However, despite the presence of chromosomal instability, extensive p53 activation, and increased apoptosis throughout the embryo, Pich KO embryos survive until day 12.5 of embryonic development. The absence of p53 failed to improve the viability of the Pich KO embryos, suggesting that the observed developmental defects are not solely due to p53-induced apoptosis. Moreover, Pich-deficient mouse embryonic fibroblasts exhibit chromosomal instability and are resistant to RASV12/E1A-induced transformation. Overall, our data indicate that PICH is essential to preserve chromosomal integrity in rapidly proliferating cells and is therefore critical during embryonic development and tumorigenesis.


Asunto(s)
Inestabilidad Cromosómica , Desarrollo Embrionario/genética , Animales , Apoptosis , Células Cultivadas , Daño del ADN , ADN Helicasas/metabolismo , Ratones , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA