Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 269
Filtrar
1.
Mol Cell ; 84(9): 1684-1698.e9, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38593805

RESUMEN

The Bloom syndrome (BLM) helicase is critical for alternative lengthening of telomeres (ALT), a homology-directed repair (HDR)-mediated telomere maintenance mechanism that is prevalent in cancers of mesenchymal origin. The DNA substrates that BLM engages to direct telomere recombination during ALT remain unknown. Here, we determine that BLM helicase acts on lagging strand telomere intermediates that occur specifically in ALT-positive cells to assemble a replication-associated DNA damage response. Loss of ATRX was permissive for BLM localization to ALT telomeres in S and G2, commensurate with the appearance of telomere C-strand-specific single-stranded DNA (ssDNA). DNA2 nuclease deficiency increased 5'-flap formation in a BLM-dependent manner, while telomere C-strand, but not G-strand, nicks promoted ALT. These findings define the seminal events in the ALT DNA damage response, linking aberrant telomeric lagging strand DNA replication with a BLM-directed HDR mechanism that sustains telomere length in a subset of human cancers.


Asunto(s)
Daño del ADN , Replicación del ADN , RecQ Helicasas , Homeostasis del Telómero , Telómero , RecQ Helicasas/metabolismo , RecQ Helicasas/genética , Humanos , Telómero/metabolismo , Telómero/genética , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína Nuclear Ligada al Cromosoma X/metabolismo , ADN Helicasas/metabolismo , ADN Helicasas/genética , Síndrome de Bloom/genética , Síndrome de Bloom/metabolismo , Síndrome de Bloom/enzimología , Síndrome de Bloom/patología , Línea Celular Tumoral
2.
Mol Cell ; 84(4): 640-658.e10, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38266639

RESUMEN

The Bloom syndrome helicase BLM interacts with topoisomerase IIIα (TOP3A), RMI1, and RMI2 to form the BTR complex, which dissolves double Holliday junctions and DNA replication intermediates to promote sister chromatid disjunction before cell division. In its absence, structure-specific nucleases like the SMX complex (comprising SLX1-SLX4, MUS81-EME1, and XPF-ERCC1) can cleave joint DNA molecules instead, but cells deficient in both BTR and SMX are not viable. Here, we identify a negative genetic interaction between BLM loss and deficiency in the BRCA1-BARD1 tumor suppressor complex. We show that this is due to a previously overlooked role for BARD1 in recruiting SLX4 to resolve DNA intermediates left unprocessed by BLM in the preceding interphase. Consequently, cells with defective BLM and BRCA1-BARD1 accumulate catastrophic levels of chromosome breakage and micronucleation, leading to cell death. Thus, we reveal mechanistic insights into SLX4 recruitment to DNA lesions, with potential clinical implications for treating BRCA1-deficient tumors.


Asunto(s)
Proteínas de Unión al ADN , Recombinasas , Humanos , ADN/genética , Reparación del ADN , Replicación del ADN , ADN Cruciforme , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Recombinasas/genética , RecQ Helicasas/genética , RecQ Helicasas/metabolismo
3.
Annu Rev Biochem ; 83: 519-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24606147

RESUMEN

RecQ helicases are an important family of genome surveillance proteins conserved from bacteria to humans. Each of the five human RecQ helicases plays critical roles in genome maintenance and stability, and the RecQ protein family members are often referred to as guardians of the genome. The importance of these proteins in cellular homeostasis is underscored by the fact that defects in BLM, WRN, and RECQL4 are linked to distinct heritable human disease syndromes. Each human RecQ helicase has a unique set of protein-interacting partners, and these interactions dictate its specialized functions in genome maintenance, including DNA repair, recombination, replication, and transcription. Human RecQ helicases also interact with each other, and these interactions have significant impact on enzyme function. Future research goals in this field include a better understanding of the division of labor among the human RecQ helicases and learning how human RecQ helicases collaborate and cooperate to enhance genome stability.


Asunto(s)
Reparación del ADN , Replicación del ADN , RecQ Helicasas/fisiología , Recombinación Genética , ADN/química , Exodesoxirribonucleasas/química , Genoma Humano , Inestabilidad Genómica , Humanos , Modelos Moleculares , Conformación Molecular , Familia de Multigenes , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , RecQ Helicasas/química , Fase S , Helicasa del Síndrome de Werner
4.
Mol Cell ; 81(5): 1027-1042.e4, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33453166

RESUMEN

Alternative lengthening of telomeres (ALT) is mediated by break-induced replication (BIR), but how BIR is regulated at telomeres is poorly understood. Here, we show that telomeric BIR is a self-perpetuating process. By tethering PML-IV to telomeres, we induced telomere clustering in ALT-associated PML bodies (APBs) and a POLD3-dependent ATR response at telomeres, showing that BIR generates replication stress. Ablation of BLM helicase activity in APBs abolishes telomere synthesis but causes multiple chromosome bridges between telomeres, revealing a function of BLM in processing inter-telomere BIR intermediates. Interestingly, the accumulation of BLM in APBs requires its own helicase activity and POLD3, suggesting that BIR triggers a feedforward loop to further recruit BLM. Enhancing BIR induces PIAS4-mediated TRF2 SUMOylation, and PIAS4 loss deprives APBs of repair proteins and compromises ALT telomere synthesis. Thus, a BLM-driven and PIAS4-mediated feedforward loop operates in APBs to perpetuate BIR, providing a critical mechanism to extend ALT telomeres.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Retroalimentación Fisiológica , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas Inhibidoras de STAT Activados/genética , ARN Helicasas/genética , Homeostasis del Telómero , Telómero/química , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Línea Celular , Línea Celular Tumoral , ADN Polimerasa III/genética , ADN Polimerasa III/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/antagonistas & inhibidores , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Cuerpos de Inclusión Intranucleares/genética , Cuerpos de Inclusión Intranucleares/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Inhibidoras de STAT Activados/antagonistas & inhibidores , Proteínas Inhibidoras de STAT Activados/metabolismo , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Transducción de Señal , Sumoilación , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética
5.
Genes Dev ; 34(9-10): 650-662, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32217664

RESUMEN

Telomeres consist of TTAGGG repeats bound by protein complexes that serve to protect the natural end of linear chromosomes. Most cells maintain telomere repeat lengths by using the enzyme telomerase, although there are some cancer cells that use a telomerase-independent mechanism of telomere extension, termed alternative lengthening of telomeres (ALT). Cells that use ALT are characterized, in part, by the presence of specialized PML nuclear bodies called ALT-associated PML bodies (APBs). APBs localize to and cluster telomeric ends together with telomeric and DNA damage factors, which led to the proposal that these bodies act as a platform on which ALT can occur. However, the necessity of APBs and their function in the ALT pathway has remained unclear. Here, we used CRISPR/Cas9 to delete PML and APB components from ALT-positive cells to cleanly define the function of APBs in ALT. We found that PML is required for the ALT mechanism, and that this necessity stems from APBs' role in localizing the BLM-TOP3A-RMI (BTR) complex to ALT telomere ends. Strikingly, recruitment of the BTR complex to telomeres in a PML-independent manner bypasses the need for PML in the ALT pathway, suggesting that BTR localization to telomeres is sufficient to sustain ALT activity.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas de Unión al ADN/metabolismo , RecQ Helicasas/metabolismo , Homeostasis del Telómero/fisiología , Telómero/genética , Telómero/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos , Transporte de Proteínas
6.
Genes Dev ; 34(19-20): 1392-1405, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32883681

RESUMEN

TRF1 facilitates the replication of telomeric DNA in part by recruiting the BLM helicase, which can resolve G-quadruplexes on the lagging-strand template. Lagging-strand telomeres lacking TRF1 or BLM form fragile telomeres-structures that resemble common fragile sites (CFSs)-but how they are formed is not known. We report that analogous to CFSs, fragile telomeres in BLM-deficient cells involved double-strand break (DSB) formation, in this case by the SLX4/SLX1 nuclease. The DSBs were repaired by POLD3/POLD4-dependent break-induced replication (BIR), resulting in fragile telomeres containing conservatively replicated DNA. BIR also promoted fragile telomere formation in cells with FokI-induced telomeric DSBs and in alternative lengthening of telomeres (ALT) cells, which have spontaneous telomeric damage. BIR of telomeric DSBs competed with PARP1-, LIG3-, and XPF-dependent alternative nonhomologous end joining (alt-NHEJ), which did not generate fragile telomeres. Collectively, these findings indicate that fragile telomeres can arise from BIR-mediated repair of telomeric DSBs.


Asunto(s)
Sitios Frágiles del Cromosoma/genética , Roturas del ADN de Doble Cadena , Replicación del ADN , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Telómero/patología , Animales , Línea Celular , Células Cultivadas , Reparación del ADN , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Fibroblastos , Humanos , Ratones , Recombinasas/genética , Recombinasas/metabolismo
7.
Mol Cell ; 75(1): 145-153.e5, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31153714

RESUMEN

Genetic recombination in all kingdoms of life initiates when helicases and nucleases process (resect) the free DNA ends to expose single-stranded DNA (ssDNA) overhangs. Resection regulation in bacteria is programmed by a DNA sequence, but a general mechanism limiting resection in eukaryotes has remained elusive. Using single-molecule imaging of reconstituted human DNA repair factors, we identify phosphorylated RPA (pRPA) as a negative resection regulator. Bloom's syndrome (BLM) helicase together with exonuclease 1 (EXO1) and DNA2 nucleases catalyze kilobase-length DNA resection on nucleosome-coated DNA. The resulting ssDNA is rapidly bound by RPA, which further stimulates DNA resection. RPA is phosphorylated during resection as part of the DNA damage response (DDR). Remarkably, pRPA inhibits DNA resection in cellular assays and in vitro via inhibition of BLM helicase. pRPA suppresses BLM initiation at DNA ends and promotes the intrinsic helicase strand-switching activity. These findings establish that pRPA provides a feedback loop between DNA resection and the DDR.


Asunto(s)
ADN de Cadena Simple/genética , Retroalimentación Fisiológica , RecQ Helicasas/genética , Proteínas Recombinantes de Fusión/genética , Proteína de Replicación A/genética , Sitios de Unión , ADN Helicasas/genética , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , ADN de Cadena Simple/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Regulación de la Expresión Génica , Recombinación Homóloga , Humanos , Microscopía Fluorescente , Nucleosomas/química , Nucleosomas/metabolismo , Oligopéptidos/genética , Oligopéptidos/metabolismo , Fosforilación , Unión Proteica , RecQ Helicasas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Replicación A/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Imagen Individual de Molécula
8.
Mol Cell ; 76(1): 27-43.e11, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31447390

RESUMEN

Cancer cells acquire unlimited proliferative capacity by either re-expressing telomerase or inducing alternative lengthening of telomeres (ALT), which relies on telomere recombination. Here, we show that ALT recombination requires coordinate regulation of the SMX and BTR complexes to ensure the appropriate balance of resolution and dissolution activities at recombining telomeres. Critical to this control is SLX4IP, which accumulates at ALT telomeres and interacts with SLX4, XPF, and BLM. Loss of SLX4IP increases ALT-related phenotypes, which is incompatible with cell growth following concomitant loss of SLX4. Inactivation of BLM is sufficient to rescue telomere aggregation and the synthetic growth defect in this context, suggesting that SLX4IP favors SMX-dependent resolution by antagonizing promiscuous BLM activity during ALT recombination. Finally, we show that SLX4IP is inactivated in a subset of ALT-positive osteosarcomas. Collectively, our findings uncover an SLX4IP-dependent regulatory mechanism critical for telomere maintenance in ALT cancer cells.


Asunto(s)
Neoplasias Óseas/enzimología , Proteínas Portadoras/metabolismo , Osteosarcoma/enzimología , RecQ Helicasas/metabolismo , Homeostasis del Telómero , Telómero/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Proteínas Portadoras/genética , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Ratones Noqueados , Ratones SCID , Osteosarcoma/genética , Osteosarcoma/patología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , RecQ Helicasas/genética , Recombinasas/genética , Recombinasas/metabolismo , Transducción de Señal , Telómero/genética , Telómero/patología
9.
Mol Cell ; 75(4): 859-874.e4, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31351878

RESUMEN

Homologous recombination (HR) is essential for high-fidelity DNA repair during mitotic proliferation and meiosis. Yet, context-specific modifications must tailor the recombination machinery to avoid (mitosis) or enforce (meiosis) the formation of reciprocal exchanges-crossovers-between recombining chromosomes. To obtain molecular insight into how crossover control is achieved, we affinity purified 7 DNA-processing enzymes that channel HR intermediates into crossovers or noncrossovers from vegetative cells or cells undergoing meiosis. Using mass spectrometry, we provide a global characterization of their composition and reveal mitosis- and meiosis-specific modules in the interaction networks. Functional analyses of meiosis-specific interactors of MutLγ-Exo1 identified Rtk1, Caf120, and Chd1 as regulators of crossing-over. Chd1, which transiently associates with Exo1 at the prophase-to-metaphase I transition, enables the formation of MutLγ-dependent crossovers through its conserved ability to bind and displace nucleosomes. Thus, rewiring of the HR network, coupled to chromatin remodeling, promotes context-specific control of the recombination outcome.


Asunto(s)
Intercambio Genético/fisiología , Meiosis/fisiología , Mitosis/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Espectrometría de Masas , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
10.
Proc Natl Acad Sci U S A ; 121(19): e2318438121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38696464

RESUMEN

Alternative lengthening of telomeres (ALT) is a telomere maintenance mechanism mediated by break-induced replication, evident in approximately 15% of human cancers. A characteristic feature of ALT cancers is the presence of C-circles, circular single-stranded telomeric DNAs composed of C-rich sequences. Despite the fact that extrachromosomal C-rich single-stranded DNAs (ssDNAs), including C-circles, are unique to ALT cells, their generation process remains undefined. Here, we introduce a method to detect single-stranded telomeric DNA, called 4SET (Strand-Specific Southern-blot for Single-stranded Extrachromosomal Telomeres) assay. Utilizing 4SET, we are able to capture C-rich single-stranded DNAs that are near 200 to 1500 nucleotides in size. Both linear C-rich ssDNAs and C-circles are abundant in the fractions of cytoplasm and nucleoplasm, which supports the idea that linear and circular C-rich ssDNAs are generated concurrently. We also found that C-rich ssDNAs originate during Okazaki fragment processing during lagging strand DNA synthesis. The generation of C-rich ssDNA requires CST-PP (CTC1/STN1/TEN1-PRIMASE-Polymerase alpha) complex-mediated priming of the C-strand DNA synthesis and subsequent excessive strand displacement of the C-rich strand mediated by the DNA Polymerase delta and the BLM helicase. Our work proposes a model for the generation of C-rich ssDNAs and C-circles during ALT-mediated telomere elongation.


Asunto(s)
ADN de Cadena Simple , Homeostasis del Telómero , Telómero , Telómero/genética , Telómero/metabolismo , Humanos , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/genética , Replicación del ADN , ADN/genética , ADN/metabolismo , ADN Circular/genética , ADN Circular/metabolismo , Southern Blotting , ADN Polimerasa III/metabolismo , ADN Polimerasa III/genética
11.
Mol Cell ; 70(3): 395-407.e4, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727616

RESUMEN

Telomeres and telomere-binding proteins form complex secondary nucleoprotein structures that are critical for genome integrity but can present serious challenges during telomere DNA replication. It remains unclear how telomere replication stress is resolved during S phase. Here, we show that the BUB3-BUB1 complex, a component in spindle assembly checkpoint, binds to telomeres during S phase and promotes telomere DNA replication. Loss of the BUB3-BUB1 complex results in telomere replication defects, including fragile and shortened telomeres. We also demonstrate that the telomere-binding ability of BUB3 and kinase activity of BUB1 are indispensable to BUB3-BUB1 function at telomeres. TRF2 targets BUB1-BUB3 to telomeres, and BUB1 can directly phosphorylate TRF1 and promote TRF1 recruitment of BLM helicase to overcome replication stress. Our findings have uncovered previously unknown roles for the BUB3-BUB1 complex in S phase and shed light on how proteins from diverse pathways function coordinately to ensure proper telomere replication and maintenance.


Asunto(s)
Proteínas de Ciclo Celular/genética , Replicación del ADN/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas Serina-Treonina Quinasas/genética , Telómero/genética , Línea Celular , Línea Celular Tumoral , ADN Helicasas/genética , Células HEK293 , Células HeLa , Humanos , Puntos de Control de la Fase M del Ciclo Celular/genética , Fase S/genética , Huso Acromático/genética , Proteínas de Unión a Telómeros/genética
12.
Mol Cell Proteomics ; 23(3): 100728, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38296025

RESUMEN

Proteasomes are multisubunit, multicatalytic protein complexes present in eukaryotic cells that degrade misfolded, damaged, or unstructured proteins. In this study, we used an activity-guided proteomic methodology based on a fluorogenic peptide substrate to characterize the composition of proteasome complexes in WT yeast and the changes these complexes undergo upon the deletion of Pre9 (Δα3) or of Sem1 (ΔSem1). A comparison of whole-cell proteomic analysis to activity-guided proteasome profiling indicates that the amounts of proteasomal proteins and proteasome interacting proteins in the assembled active proteasomes differ significantly from their total amounts in the cell as a whole. Using this activity-guided profiling approach, we characterized the changes in the abundance of subunits of various active proteasome species in different strains, quantified the relative abundance of active proteasomes across these strains, and charted the overall distribution of different proteasome species within each strain. The distributions obtained by our mass spectrometry-based quantification were markedly higher for some proteasome species than those obtained by activity-based quantification alone, suggesting that the activity of some of these species is impaired. The impaired activity appeared mostly among 20SBlm10 proteasome species which account for 20% of the active proteasomes in WT. To identify the factors behind this impaired activity, we mapped and quantified known proteasome-interacting proteins. Our results suggested that some of the reduced activity might be due to the association of the proteasome inhibitor Fub1. Additionally, we provide novel evidence for the presence of nonmature and therefore inactive proteasomal protease subunits ß2 and ß5 in the fully assembled proteasomes.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Proteínas de Saccharomyces cerevisiae , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica , Proteínas , Péptidos/química , Espectrometría de Masas , Saccharomyces cerevisiae/metabolismo
13.
RNA ; 29(10): 1557-1574, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37460154

RESUMEN

Assemblysomes are EDTA- and RNase-resistant ribonucleoprotein (RNP) complexes of paused ribosomes with protruding nascent polypeptide chains. They have been described in yeast and human cells for the proteasome subunit Rpt1, and the disordered amino-terminal part of the nascent chain was found to be indispensable for the accumulation of the Rpt1-RNP into assemblysomes. Motivated by this, to find other assemblysome-associated RNPs we used bioinformatics to rank subunits of Saccharomyces cerevisiae protein complexes according to their amino-terminal disorder propensity. The results revealed that gene products involved in DNA repair are enriched among the top candidates. The Sgs1 DNA helicase was chosen for experimental validation. We found that indeed nascent chains of Sgs1 form EDTA-resistant RNP condensates, assemblysomes by definition. Moreover, upon exposure to UV, SGS1 mRNA shifted from assemblysomes to polysomes, suggesting that external stimuli are regulators of assemblysome dynamics. We extended our studies to human cell lines. The BLM helicase, ortholog of yeast Sgs1, was identified upon sequencing assemblysome-associated RNAs from the MCF7 human breast cancer cell line, and mRNAs encoding DNA repair proteins were overall enriched. Using the radiation-resistant A549 cell line, we observed by transmission electron microscopy that 1,6-hexanediol, an agent known to disrupt phase-separated condensates, depletes ring ribosome structures compatible with assemblysomes from the cytoplasm of cells and makes the cells more sensitive to X-ray treatment. Taken together, these findings suggest that assemblysomes may be a component of the DNA damage response from yeast to human.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Humanos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , RecQ Helicasas/genética , Ácido Edético/metabolismo , Daño del ADN , ARN/metabolismo , Ribonucleoproteínas/genética , Ribosomas/genética , Ribosomas/metabolismo
14.
Mol Cell ; 65(3): 515-526.e3, 2017 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-28065599

RESUMEN

Saccharomyces cerevisiae mating-type switching is initiated by a double-strand break (DSB) at MATa, leaving one cut end perfectly homologous to the HMLα donor, while the second end must be processed to remove a non-homologous tail before completing repair by gene conversion (GC). When homology at the matched end is ≤150 bp, efficient repair depends on the recombination enhancer, which tethers HMLα near the DSB. Thus, homology shorter than an apparent minimum efficient processing segment can be rescued by tethering the donor near the break. When homology at the second end is ≤150 bp, second-end capture becomes inefficient and repair shifts from GC to break-induced replication (BIR). But when pol32 or pif1 mutants block BIR, GC increases 3-fold, indicating that the steps blocked by these mutations are reversible. With short second-end homology, absence of the RecQ helicase Sgs1 promotes gene conversion, whereas deletion of the FANCM-related Mph1 helicase promotes BIR.


Asunto(s)
Reparación del ADN , ADN de Hongos/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , ARN Helicasas DEAD-box/metabolismo , Roturas del ADN de Doble Cadena , Replicación del ADN , Conversión Génica , Recombinación Homóloga , RecQ Helicasas/metabolismo
15.
Proc Natl Acad Sci U S A ; 119(23): e2116462119, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35658074

RESUMEN

Helicases are multifunctional motor proteins with the primary task of separating nucleic acid duplexes. These enzymes often exist in distinct oligomeric forms and play essential roles during nucleic acid metabolism. Whether there is a correlation between their oligomeric state and cellular function, and how helicases effectively perform functional switching remains enigmatic. Here, we address these questions using a combined single-molecule approach and Bloom syndrome helicase (BLM). By examining the head-on collision of two BLM-mediated DNA unwinding forks, we find that two groups of BLM, upon fork convergence, promptly oligomerize across the fork junctions and tightly bridge two independent single-stranded (ss) DNA molecules that were newly generated by the unwinding BLMs. This protein oligomerization is mediated by the helicase and RNase D C-terminal (HRDC) domain of BLM and can sustain a disruptive force of up to 300 pN. Strikingly, onsite BLM oligomerization gives rise to an immediate transition of their helicase activities, from unwinding dsDNA to translocating along ssDNA at exceedingly fast rates, thus allowing for the efficient displacement of ssDNA-binding proteins, such as RPA and RAD51. These findings uncover an activity transition pathway for helicases and help to explain how BLM plays both pro- and anti-recombination roles in the maintenance of genome stability.


Asunto(s)
ADN de Cadena Simple , RecQ Helicasas , ADN/metabolismo , ADN de Cadena Simple/genética , Recombinación Homóloga , Microscopía Confocal , Pinzas Ópticas , RecQ Helicasas/metabolismo
16.
Proc Natl Acad Sci U S A ; 119(18): e2123420119, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35452329

RESUMEN

Four-way DNA intermediates, also known as Holliday junctions (HJs), are formed during homologous recombination and DNA repair, and their resolution is necessary for proper chromosome segregation. To facilitate the biochemical analysis of HJ processing, we developed a method involving DNAzyme self-cleavage to generate 1.8-kb DNA molecules containing either single (sHJ) or double Holliday junctions (dHJs). We show that dHJ DNAs (referred to as HoJo DNAs) are dissolved by the human BLM­TopIIIα­RMI1­RMI2 complex to form two noncrossover products. However, structure-selective endonucleases (human GEN1 and SMX complex) resolve DNA containing single or double HJs to yield a mixture of crossover and noncrossover products. Finally, we demonstrate that chromatin inhibits the resolution of the double HJ by GEN or SMX while allowing BTRR-mediated dissolution.


Asunto(s)
Cromatina , ADN Cruciforme , Cromatina/genética , Cromosomas , ADN/genética , ADN Cruciforme/genética , Solubilidad
17.
J Biol Chem ; 299(2): 102802, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36529288

RESUMEN

DNA resection-the nucleolytic processing of broken DNA ends-is the first step of homologous recombination. Resection is catalyzed by the resectosome, a multienzyme complex that includes bloom syndrome helicase (BLM), DNA2 or exonuclease 1 nucleases, and additional DNA-binding proteins. Although the molecular players have been known for over a decade, how the individual proteins work together to regulate DNA resection remains unknown. Using single-molecule imaging, we characterized the roles of the MRE11-RAD50-NBS1 complex (MRN) and topoisomerase IIIa (TOP3A)-RMI1/2 during long-range DNA resection. BLM partners with TOP3A-RMI1/2 to form the BTRR (BLM-TOP3A-RMI1/2) complex (or BLM dissolvasome). We determined that TOP3A-RMI1/2 aids BLM in initiating DNA unwinding, and along with MRN, stimulates DNA2-mediated resection. Furthermore, we found that MRN promotes the association between BTRR and DNA and synchronizes BLM and DNA2 translocation to prevent BLM from pausing during resection. Together, this work provides direct observation of how MRN and DNA2 harness the BTRR complex to resect DNA efficiently and how TOP3A-RMI1/2 regulates the helicase activity of BLM to promote efficient DNA repair.


Asunto(s)
Reparación del ADN , ADN-Topoisomerasas de Tipo I , ADN , Complejos Multienzimáticos , Humanos , ADN/metabolismo , Roturas del ADN de Doble Cadena , ADN-Topoisomerasas de Tipo I/metabolismo , Complejos Multienzimáticos/metabolismo , Imagen Individual de Molécula
18.
BMC Geriatr ; 24(1): 603, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009979

RESUMEN

BACKGROUND: High-grade endometrial stromal sarcoma (HG-ESS) is a rare malignant tumor with poor prognosis. To overcome the limitations of current treatment for advanced patients, the intervention of targeted drug therapy is urgently needed. CASE PRESENTATION: A 74-year-old married woman who presented with abdominal distension and lower abdominal pain was admitted to Hebei General Hospital. After surgery, immunohistochemical staining revealed a malignant tumor which was consistent with HG-ESS. Tumor recurrence occurred 2 months after surgery. Then the patient underwent chemotherapy with two courses but responded poorly. Subsequently we observed ATM, BLM, and CDH1 co-mutations by Next Generation Sequencing (NGS). Then the patient received pamiparib, which resulted in a 10-month progression-free survival (PFS) and is now stable with the administration of sintilimab in combination with pamiparib and anlotinib. CONCLUSIONS: Due to the successful use of poly ADP-ribose polymerase inhibitor (PARPi) on HG-ESS, we suggest that the selection of effective targeted drugs combined with anti- programmed death-1 (PD-1) drug therapy based on genetic testing may become a new option for the treatment of homologous repair deficient (HR-deficient) HG-ESS.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , Cadherinas , Neoplasias Endometriales , RecQ Helicasas , Sarcoma Estromático Endometrial , Humanos , Femenino , Anciano , Neoplasias Endometriales/genética , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/patología , Sarcoma Estromático Endometrial/genética , Sarcoma Estromático Endometrial/tratamiento farmacológico , Sarcoma Estromático Endometrial/diagnóstico , Proteínas de la Ataxia Telangiectasia Mutada/genética , RecQ Helicasas/genética , Cadherinas/genética , Antígenos CD/genética , Mutación
19.
BMC Pulm Med ; 24(1): 405, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39180004

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF), an interstitial lung disease, is characterized by the exacerbation of progressive pulmonary fibrosis (PF). IPF primarily affects older individuals and can lead to respiratory failure. This study aimed to assess the effects of triiodothyronine (T3) treatment on the lung microbiome of mice with PF. METHODS: Mice were perfused with bleomycin (BLM) to establish a PF model. Using a randomized design, 40 female specific pathogen-free (SPF) C57BL6/N mice were divided into four groups: saline, saline + T3, BLM, and BLM + T3. Histological morphology was assessed through Hematoxylin and Eosin staining as well as Masson's Trichrome staining. For the identification of lung bacteria, 16S rRNA gene sequencing was employed. An Enzyme-Linked Immunosorbent Assay was used to measure total T3 (TT3), free T3 (FT3, and reverse T3 (rT3) levels in the peripheral serum. RESULTS: T3 treatment ameliorated BLM-induced lung fibrosis and structural damage. The microbiome experienced a decrease in the abundance of Proteobacteria, Bacteroides, and Actinomycetes and an increase in the abundance of Firmicutes when exposed to BLM; however, T3 treatment reversed this effect. The four groups showed no significant difference in alpha microbiome diversity (P > 0.05). Serum concentrations of TT3 and FT3 were positively correlated with microbiome abundance (P < 0.05). Administration of T3 enhanced the microbiota in PF without affecting the diversity and biological functions of the microbiome (P > 0.05). CONCLUSION: The administration of T3 demonstrated a favorable impact on the lung microbiota of mice afflicted with PF, thereby partially substantiating the potential role of T3 as a therapeutic agent in the management of PF.


Asunto(s)
Bleomicina , Modelos Animales de Enfermedad , Pulmón , Ratones Endogámicos C57BL , Microbiota , ARN Ribosómico 16S , Triyodotironina , Animales , Ratones , Triyodotironina/sangre , Triyodotironina/farmacología , Microbiota/efectos de los fármacos , Pulmón/patología , Pulmón/microbiología , Femenino , ARN Ribosómico 16S/genética , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/microbiología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/microbiología
20.
Ecotoxicol Environ Saf ; 271: 116013, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38281433

RESUMEN

The damage excessive neodymium (Nd) causes to animals and plants should not be underestimated. However, there is little research on the impact of pH and associated ions on the toxicity of Nd. Here, a biotic ligand model (BLM) was expanded to predict the effects of pH and chief anions on the toxic impact of Nd on wheat root elongation in a simulated soil solution. The results suggested that Nd3+ and NdOH2+ were the major ions causing phytotoxicity to wheat roots at pH values of 4.5-7.0. The Nd toxicity decreased as the activities of H+, Ca2+, and Mg2+ increased but not when the activities of K+ and Na+ increased. The results indicated that H+, Ca2+, and Mg2+ competed with Nd for binding sites. An extended BLM was developed to consider the effects of pH, H+, Ca2+, and Mg2+, and the following stability constants were obtained: logKNdBL = 2.51, logKNdOHBL = 3.90, logKHBL = 4.01, logKCaBL = 2.43, and logKMgBL = 2.70. The results demonstrated that the BLM could predict the Nd toxicity well while considering the competition of H+, Ca2+, Mg2+ and the toxic species Nd3+ and NdOH2+ for binding sites.


Asunto(s)
Neodimio , Contaminantes del Suelo , Neodimio/toxicidad , Triticum , Ligandos , Contaminantes del Suelo/toxicidad , Modelos Biológicos , Raíces de Plantas , Iones/farmacología , Concentración de Iones de Hidrógeno
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda