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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 78(6): 1152-1165.e8, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32516598

RESUMEN

The APEX2 gene encodes APE2, a nuclease related to APE1, the apurinic/apyrimidinic endonuclease acting in base excision repair. Loss of APE2 is lethal in cells with mutated BRCA1 or BRCA2, making APE2 a prime target for homologous recombination-defective cancers. However, because the function of APE2 in DNA repair is poorly understood, it is unclear why BRCA-deficient cells require APE2 for viability. Here we present the genetic interaction profiles of APE2, APE1, and TDP1 deficiency coupled to biochemical and structural dissection of APE2. We conclude that the main role of APE2 is to reverse blocked 3' DNA ends, problematic lesions that preclude DNA synthesis. Our work also suggests that TOP1 processing of genomic ribonucleotides is the main source of 3'-blocking lesions relevant to APEX2-BRCA1/2 synthetic lethality. The exquisite sensitivity of BRCA-deficient cells to 3' blocks indicates that they represent a tractable vulnerability in homologous recombination-deficient tumor cells.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endonucleasas/metabolismo , Enzimas Multifuncionales/metabolismo , Proteína BRCA1/genética , Proteína BRCA2/genética , Línea Celular , ADN/metabolismo , Daño del ADN , Reparación del ADN/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Endonucleasas/genética , Genes BRCA1/fisiología , Humanos , Enzimas Multifuncionales/genética , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo
2.
Genes Dev ; 33(19-20): 1293-1294, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575675

RESUMEN

Homologous recombination (HR) is an important route for repairing DNA double-strand breaks (DSBs). The early stages of HR are well understood, but later stages remain mysterious. In this issue of Genes & Development, Hustedt and colleagues (pp. 1397-1415) reveal HROB as a new player in HR required for recruitment of the MCM8-9 complex, which is paralogous to the MCM2-7 replicative helicase. HROB functions closely with MCM8-9 to promote postsynaptic DNA repair synthesis. This study sheds valuable light on late events in HR and suggests that HROB may load MCM8-9 onto HR intermediates to facilitate the DNA unwinding required for DNA repair synthesis.


Asunto(s)
Roturas del ADN de Doble Cadena , Recombinación Homóloga , Reparación del ADN , Replicación del ADN , Proteínas de Mantenimiento de Minicromosoma
3.
Mol Cell Proteomics ; : 100802, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38880245

RESUMEN

The ATR kinase protects cells against DNA damage and replication stress and represents a promising anti-cancer drug target. The ATR inhibitors (ATRi) berzosertib and gartisertib are both in clinical trials for the treatment of advanced solid tumours as monotherapy or in combination with genotoxic agents. We carried out quantitative phospho-proteomic screening for ATR biomarkers that are highly sensitive to berzosertib and gartisertib, using an optimized mass spectrometry pipeline. Screening identified a range of novel ATR-dependent phosphorylation events, which were grouped into three broad classes: i) targets whose phosphorylation is highly sensitive to ATRi and which could be the next generation of ATR biomarkers; ii) proteins with known genome maintenance roles not previously known to be regulated by ATR; iii) novel targets whose cellular roles are unclear. Class iii targets represent candidate DNA damage response proteins and, with this in mind, proteins in this class were subjected to secondary screening for recruitment to DNA damage sites. We show that one of the proteins recruited, SCAF1, interacts with RNAPII in a phospho-dependent manner and recruitment requires PARP activity and interaction with RNAPII. We also show that SCAF1 deficiency partly rescues RAD51 loading in cells lacking the BRCA1 tumour suppressor. Taken together these data reveal potential new ATR biomarkers and new genome maintenance factors.

4.
EMBO J ; 40(23): e108271, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34605059

RESUMEN

Mutations in the gene encoding the CDKL5 kinase are among the most common genetic causes of childhood epilepsy and can also give rise to the severe neurodevelopmental condition CDD (CDKL5 deficiency disorder). Despite its importance for human health, the phosphorylation targets and cellular roles of CDKL5 are poorly understood, especially in the cell nucleus. Here, we report that CDKL5 is recruited to sites of DNA damage in actively transcribed regions of the nucleus. A quantitative phosphoproteomic screen for nuclear CDKL5 substrates reveals a network of transcriptional regulators including Elongin A (ELOA), phosphorylated on a specific CDKL5 consensus motif. Recruitment of CDKL5 and ELOA to damaged DNA, and subsequent phosphorylation of ELOA, requires both active transcription and the synthesis of poly(ADP-ribose) (PAR), to which CDKL5 can bind. Critically, CDKL5 kinase activity is essential for the transcriptional silencing of genes induced by DNA double-strand breaks. Thus, CDKL5 is a DNA damage-sensing, PAR-controlled transcriptional modulator, a finding with implications for understanding the molecular basis of CDKL5-related diseases.


Asunto(s)
Roturas del ADN de Doble Cadena , Daño del ADN , Elonguina/metabolismo , Neuronas/patología , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Activación Transcripcional , Elonguina/genética , Síndromes Epilépticos/genética , Síndromes Epilépticos/metabolismo , Síndromes Epilépticos/patología , Humanos , Mutación , Neuronas/metabolismo , Fosfoproteínas/genética , Fosforilación , Poli Adenosina Difosfato Ribosa/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Espasmos Infantiles/genética , Espasmos Infantiles/metabolismo , Espasmos Infantiles/patología
5.
Cell ; 142(1): 65-76, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20603015

RESUMEN

DNA interstrand crosslinks (ICLs) are highly toxic because they block the progression of replisomes. The Fanconi Anemia (FA) proteins, encoded by genes that are mutated in FA, are important for repair of ICLs. The FA core complex catalyzes the monoubiquitination of FANCD2, and this event is essential for several steps of ICL repair. However, how monoubiquitination of FANCD2 promotes ICL repair at the molecular level is unknown. Here, we describe a highly conserved protein, KIAA1018/MTMR15/FAN1, that interacts with, and is recruited to sites of DNA damage by, the monoubiquitinated form of FANCD2. FAN1 exhibits endonuclease activity toward 5' flaps and has 5' exonuclease activity, and these activities are mediated by an ancient VRR_nuc domain. Depletion of FAN1 from human cells causes hypersensitivity to ICLs, defects in ICL repair, and genome instability. These data at least partly explain how ubiquitination of FANCD2 promotes DNA repair.


Asunto(s)
Reparación del ADN , Exodesoxirribonucleasas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis , Proteína BRCA2/metabolismo , Línea Celular , Reactivos de Enlaces Cruzados/farmacología , Daño del ADN/efectos de los fármacos , Endodesoxirribonucleasas , Endonucleasas/química , Endonucleasas/metabolismo , Exodesoxirribonucleasas/química , Humanos , Datos de Secuencia Molecular , Enzimas Multifuncionales , Estructura Terciaria de Proteína , Alineación de Secuencia , Ubiquitinación
6.
Mol Cell ; 66(5): 610-621.e4, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575657

RESUMEN

Defects in the repair of DNA interstrand crosslinks (ICLs) are associated with the genome instability syndrome Fanconi anemia (FA). Here we report that cells with mutations in RFWD3, an E3 ubiquitin ligase that interacts with and ubiquitylates replication protein A (RPA), show profound defects in ICL repair. An amino acid substitution in the WD40 repeats of RFWD3 (I639K) found in a new FA subtype abolishes interaction of RFWD3 with RPA, thereby preventing RFWD3 recruitment to sites of ICL-induced replication fork stalling. Moreover, single point mutations in the RPA32 subunit of RPA that abolish interaction with RFWD3 also inhibit ICL repair, demonstrating that RPA-mediated RFWD3 recruitment to stalled replication forks is important for ICL repair. We also report that unloading of RPA from sites of ICL induction is perturbed in RFWD3-deficient cells. These data reveal important roles for RFWD3 localization in protecting genome stability and preserving human health.


Asunto(s)
Daño del ADN , Anemia de Fanconi/enzimología , Reparación del ADN por Recombinación , Origen de Réplica , Proteína de Replicación A/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Sitios de Unión , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Anemia de Fanconi/genética , Células HeLa , Humanos , Mutación , Unión Proteica , Interferencia de ARN , Proteína de Replicación A/genética , Transfección , Ubiquitina-Proteína Ligasas/genética
7.
Genes Dev ; 30(6): 639-44, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26980188

RESUMEN

The Fan1 endonuclease is required for repair of DNA interstrand cross-links (ICLs). Mutations in human Fan1 cause karyomegalic interstitial nephritis (KIN), but it is unclear whether defective ICL repair is responsible or whether Fan1 nuclease activity is relevant. We show that Fan1 nuclease-defective (Fan1(nd/nd)) mice develop a mild form of KIN. The karyomegalic nuclei from Fan1(nd/nd) kidneys are polyploid, and fibroblasts from Fan1(nd/nd) mice become polyploid upon ICL induction, suggesting that defective ICL repair causes karyomegaly. Thus, Fan1 nuclease activity promotes ICL repair in a manner that controls ploidy, a role that we show is not shared by the Fanconi anemia pathway or the Slx4-Slx1 nuclease also involved in ICL repair.


Asunto(s)
Daño del ADN/genética , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Nefritis Intersticial/enzimología , Nefritis Intersticial/genética , Poliploidía , Animales , Células Cultivadas , Reparación del ADN/genética , Desoxirribonucleasas/metabolismo , Exodesoxirribonucleasas , Técnicas de Sustitución del Gen , Riñón/patología , Ratones , Enzimas Multifuncionales , Nefritis Intersticial/fisiopatología
8.
EMBO J ; 37(24)2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30266825

RESUMEN

Mutations in the gene encoding the protein kinase CDKL5 cause a debilitating neurodevelopmental disease termed CDKL5 disorder. The impact of these mutations on CDKL5 function is poorly understood because the substrates and cellular processes controlled by CDKL5 are unclear. Here, we describe a quantitative phosphoproteomic screening which identified MAP1S, CEP131 and DLG5-regulators of microtubule and centrosome function-as cellular substrates of CDKL5. Antibodies against MAP1S phospho-Ser900 and CEP131 phospho-Ser35 confirmed CDKL5-dependent phosphorylation of these targets in human cells. The phospho-acceptor serine residues in MAP1S, CEP131 and DLG5 lie in the motif RPXSA, although CDKL5 can tolerate residues other than Ala immediately C-terminal to the phospho-acceptor serine. We provide insight into the control of CDKL5 activity and show that pathogenic mutations in CDKL5 cause a major reduction in CDKL5 activity in vitro and in cells. These data reveal the first cellular substrates of CDKL5, which may represent important biomarkers in the diagnosis and treatment of CDKL5 disorder, and illuminate the functions of this poorly characterized kinase.


Asunto(s)
Síndromes Epilépticos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Espasmos Infantiles/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencias de Aminoácidos , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Centrosoma/metabolismo , Proteínas del Citoesqueleto , Síndromes Epilépticos/genética , Síndromes Epilépticos/patología , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Proteínas de Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Microtúbulos/metabolismo , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteómica , Espasmos Infantiles/genética , Espasmos Infantiles/patología , Proteínas Supresoras de Tumor/genética
9.
Mol Cell ; 52(2): 221-33, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24076219

RESUMEN

Holliday junctions (HJs) are X-shaped DNA structures that arise during homologous recombination, which must be removed to enable chromosome segregation. The SLX1 and MUS81-EME1 nucleases can both process HJs in vitro, and they bind in close proximity on the SLX4 scaffold, hinting at possible cooperation. However, the cellular roles of mammalian SLX1 are not yet known. Here, we use mouse genetics and structure function analysis to investigate SLX1 function. Disrupting the murine Slx1 and Slx4 genes revealed that they are essential for HJ resolution in mitotic cells. Moreover, SLX1 and MUS81-EME1 act together to resolve HJs in a manner that requires tethering to SLX4. We also show that SLX1, like MUS81-EME1, is required for repair of DNA interstrand crosslinks, but this role appears to be independent of HJ cleavage, at least in mouse cells. These findings shed light on HJ resolution in mammals and on maintenance of genome stability.


Asunto(s)
Reparación del ADN , ADN Cruciforme , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , Endonucleasas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Células Cultivadas , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/citología , Endodesoxirribonucleasas/genética , Endonucleasas/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Datos de Secuencia Molecular , Unión Proteica , Interferencia de ARN , Recombinasas/genética , Recombinasas/metabolismo , Homología de Secuencia de Aminoácido
10.
Biochem J ; 477(10): 1893-1905, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32239177

RESUMEN

Congenital dyserythropoietic anaemia (CDA) type I is a rare blood disorder characterised by moderate to severe macrocytic anaemia and hepatomegaly, with spongy heterochromatin and inter-nuclear bridges seen in bone marrow erythroblasts. The vast majority of cases of CDA type I are caused by mutations in the CDAN1 gene. The product of CDAN1 is Codanin-1, which interacts the histone chaperone ASF1 in the cytoplasm. Codanin-1 is a negative regulator of chromatin replication, sequestering ASF1 in the cytoplasm, restraining histone deposition and thereby limiting DNA replication. The remainder of CDA-I cases are caused by mutations in the C15ORF41 gene, but very little is known about the product of this gene. Here, we report that C15ORF41 forms a tight, near-stoichiometric complex with Codanin1 in human cells, interacting with the C-terminal region of Codanin-1. We present the characterisation of the C15ORF41-Codanin-1 complex in humans in cells and in vitro, and demonstrate that Codanin-1 appears to sequester C15ORF41 in the cytoplasm as previously shown for ASF1. The findings in this study have major implications for understanding the functions of C15ORF41 and Codanin-1, and the aetiology of CDA-I.


Asunto(s)
Anemia Diseritropoyética Congénita/etiología , Proteínas de Ciclo Celular/genética , Glicoproteínas , Complejos Multiproteicos , Proteínas Nucleares/genética , Anemia Diseritropoyética Congénita/genética , Proteínas de Ciclo Celular/química , Línea Celular , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Complejos Multiproteicos/química , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo
11.
Am J Med Genet A ; 182(5): 1217-1222, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32034940

RESUMEN

Pathogenic variants in the cyclin-dependent kinase-like 5 (CDKL5) gene cause the neurodevelopmental disorder, the CDKL5 deficiency disorder. Reports of individuals with pathogenic variants in CDKL5 without seizures are exceedingly rare, and in-depth analyses of their variants have been lacking. Whole-genome sequencing was performed on a 29-year-old female with mild intellectual disability who, in the absence of overt seizures, presented with multiple episodes of altered mental status over a 24-year period. Clinical history was supplemented by a parent completed questionnaire from the International CDKL5 Disorder Database. We identified a de novo heterozygous variant in CDKL5 (NM_003159.2:c.645T>A;p.Ser215Arg). In-depth computational analysis performed to predict the impact of the variant on protein structure and function demonstrated that the variant was likely pathogenic. In this light, cell-based studies showed that the S215R substitution causes a marked reduction in CDKL5 kinase activity. Similarities between our case and one previously reported case are striking. These cases, both without seizures but with apparent behavioral symptomatology, together question whether seizures are mandatory in this neurodevelopmental disorder.


Asunto(s)
Síndromes Epilépticos/genética , Discapacidad Intelectual/genética , Proteínas Serina-Treonina Quinasas/genética , Convulsiones/genética , Espasmos Infantiles/genética , Adulto , Síndromes Epilépticos/fisiopatología , Femenino , Humanos , Discapacidad Intelectual/fisiopatología , Mutación , Mutación Missense/genética , Fenotipo , Síndrome de Rett/genética , Síndrome de Rett/fisiopatología , Convulsiones/fisiopatología , Espasmos Infantiles/fisiopatología , Secuenciación Completa del Genoma
12.
EMBO J ; 34(3): 326-43, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25538220

RESUMEN

Reversible protein ubiquitylation plays important roles in various processes including DNA repair. Here, we identify the deubiquitylase USP45 as a critical DNA repair regulator. USP45 associates with ERCC1, a subunit of the DNA repair endonuclease XPF-ERCC1, via a short acidic motif outside of the USP45 catalytic domain. Wild-type USP45, but not a USP45 mutant defective in ERCC1 binding, efficiently deubiquitylates ERCC1 in vitro, and the levels of ubiquitylated ERCC1 are markedly enhanced in USP45 knockout cells. Cells lacking USP45 are hypersensitive specifically to UV irradiation and DNA interstrand cross-links, similar to cells lacking ERCC1. Furthermore, the repair of UV-induced DNA damage is markedly reduced in USP45-deficient cells. ERCC1 translocation to DNA damage-induced subnuclear foci is markedly impaired in USP45 knockout cells, possibly accounting for defective DNA repair. Finally, USP45 localises to sites of DNA damage in a manner dependent on its deubiquitylase activity, but independent of its ability to bind ERCC1-XPF. Together, these results establish USP45 as a new regulator of XPF-ERCC1 crucial for efficient DNA repair.


Asunto(s)
Núcleo Celular/metabolismo , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Endopeptidasas/metabolismo , Ubiquitinación , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/efectos de la radiación , Animales , Línea Celular Tumoral , Núcleo Celular/genética , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Endopeptidasas/genética , Humanos , Ratones , Ratones Noqueados , Rayos Ultravioleta/efectos adversos
13.
Mol Cell ; 40(4): 632-44, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21055984

RESUMEN

Budding yeast Mms22 is required for homologous recombination (HR)-mediated repair of stalled or broken DNA replication forks. Here we identify a human Mms22-like protein (MMS22L) and an MMS22L-interacting protein, NFκBIL2/TONSL. Depletion of MMS22L or TONSL from human cells causes a high level of double-strand breaks (DSBs) during DNA replication. Both proteins accumulate at stressed replication forks, and depletion of MMS22L or TONSL from cells causes hypersensitivity to agents that cause S phase-associated DSBs, such as topoisomerase (TOP) inhibitors. In this light, MMS22L and TONSL are required for the HR-mediated repair of replication fork-associated DSBs. In cells depleted of either protein, DSBs induced by the TOP1 inhibitor camptothecin are resected normally, but the loading of the RAD51 recombinase is defective. Therefore, MMS22L and TONSL are required for the maintenance of genome stability when unscheduled DSBs occur in the vicinity of DNA replication forks.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Recombinación Genética , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Supervivencia Celular , Biología Computacional , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/química , ADN Polimerasa Dirigida por ADN , Resistencia a Medicamentos , Humanos , Modelos Biológicos , Chaperonas Moleculares , Datos de Secuencia Molecular , Complejos Multienzimáticos , FN-kappa B/química , Proteínas Nucleares/química , Unión Proteica , Recombinasa Rad51/metabolismo , Fase S
14.
Mol Cell ; 35(1): 116-27, 2009 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-19595721

RESUMEN

Budding yeast Slx4 interacts with the structure-specific endonuclease Slx1 to ensure completion of ribosomal DNA replication. Slx4 also interacts with the Rad1-Rad10 endonuclease to control cleavage of 3' flaps during repair of double-strand breaks (DSBs). Here we describe the identification of human SLX4, a scaffold for DNA repair nucleases XPF-ERCC1, MUS81-EME1, and SLX1. SLX4 immunoprecipitates show SLX1-dependent nuclease activity toward Holliday junctions and MUS81-dependent activity toward other branched DNA structures. Furthermore, SLX4 enhances the nuclease activity of SLX1, MUS81, and XPF. Consistent with a role in processing recombination intermediates, cells depleted of SLX4 are hypersensitive to genotoxins that cause DSBs and show defects in the resolution of interstrand crosslink-induced DSBs. Depletion of SLX4 causes a decrease in DSB-induced homologous recombination. These data show that SLX4 is a regulator of structure-specific nucleases and that SLX4 and SLX1 are important regulators of genome stability in human cells.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Endonucleasas/metabolismo , Recombinasas/metabolismo , Western Blotting , Línea Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunoprecipitación , Unión Proteica , ARN Interferente Pequeño/genética , Recombinasas/genética , Transfección , Técnicas del Sistema de Dos Híbridos
15.
J Cell Sci ; 127(Pt 13): 2811-7, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24794496

RESUMEN

Defects in SLX4, a scaffold for DNA repair nucleases, result in Fanconi anemia (FA), due to the defective repair of inter-strand DNA crosslinks (ICLs). Some FA patients have an SLX4 deletion removing two tandem UBZ4-type ubiquitin-binding domains that are implicated in protein recruitment to sites of DNA damage. Here, we show that human SLX4 is recruited to sites of ICL induction but that the UBZ-deleted form of SLX4 in cells from FA patients is not. SLX4 recruitment does not require either the ubiquitylation of FANCD2 or the E3 ligases RNF8, RAD18 and BRCA1. We show that the first (UBZ-1) but not the second UBZ domain of SLX4 binds to ubiquitin polymers, with a preference for K63-linked chains. Furthermore, UBZ-1 is required for SLX4 recruitment to ICL sites and for efficient ICL repair in murine fibroblasts. The SLX4 UBZ-2 domain does not bind to ubiquitin in vitro or contribute to ICL repair, but it is required for the resolution of Holliday junctions in vivo. These data shed light on SLX4 recruitment, and they point to the existence of currently unidentified ubiquitylated ligands and E3 ligases that are crucial for ICL repair.


Asunto(s)
Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Recombinasas/genética , Ubiquitina/metabolismo , Animales , Sitios de Unión , ADN/genética , ADN/metabolismo , Reparación del ADN , Humanos , Ratones , Estructura Terciaria de Proteína , Recombinasas/metabolismo
16.
HPB (Oxford) ; 17(10): 927-35, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26258662

RESUMEN

OBJECTIVES: To assess and compare the accuracy and inter-observer agreement for the detection of liver lesions using Primovist magnetic resonance imaging (pMRI) and computed tomography during arterial portography (CTAP). METHODS: Patients evaluated at St George Hospital Liver Unit for colorectal liver metastases (CRCLM) underwent CTAP as part of standard staging. pMRI was added to the pre-operative assessment. Two radiologists reported CTAP and two reported pMRI. The sensitivity and specificity of CTAP and pMRI were calculated using histopathology as the gold standard. RESULTS: Complete data were available for 62 patients corresponding to 219 lesions confirmed on histopathology. Agreement on the detection of lesions between the two radiologists that reported pMRI was higher than for CTAP (Kappa = 0.80 versus 0.74). Specificity of lesion detection for pMRI was 0.88 and 0.83 for CTAP (P = 0.112). Sensitivity for pMRI was 0.83 and 0.81 for CTAP. For patients who had chemotherapy before evaluation, pMRI had a significantly higher specificity than CTAP (0.79 versus 0.63, P = 0.011). CONCLUSIONS: pMRI is less invasive, has a good inter-observer agreement, has comparable sensitivity and specificity to CTAP in the pre-chemotherapy population and demonstrates better specificity in patients assessed post-chemotherapy. pMRI is a valid alternative to CTAP in the assessment of CRCLM.


Asunto(s)
Neoplasias Colorrectales/patología , Neoplasias Hepáticas/diagnóstico , Imagen por Resonancia Magnética/métodos , Portografía/métodos , Tomografía Computarizada por Rayos X/métodos , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/terapia , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Reproducibilidad de los Resultados
18.
Nat Struct Mol Biol ; 31(3): 523-535, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38238586

RESUMEN

Histone chaperones control nucleosome density and chromatin structure. In yeast, the H3-H4 chaperone Spt2 controls histone deposition at active genes but its roles in metazoan chromatin structure and organismal physiology are not known. Here we identify the Caenorhabditis elegans ortholog of SPT2 (CeSPT-2) and show that its ability to bind histones H3-H4 is important for germline development and transgenerational epigenetic gene silencing, and that spt-2 null mutants display signatures of a global stress response. Genome-wide profiling showed that CeSPT-2 binds to a range of highly expressed genes, and we find that spt-2 mutants have increased chromatin accessibility at a subset of these loci. We also show that SPT2 influences chromatin structure and controls the levels of soluble and chromatin-bound H3.3 in human cells. Our work reveals roles for SPT2 in controlling chromatin structure and function in Metazoa.


Asunto(s)
Proteínas de Unión al ADN , Chaperonas de Histonas , Animales , Humanos , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Cromatina/metabolismo , Nucleosomas/metabolismo , Saccharomyces cerevisiae/metabolismo
19.
J Biol Chem ; 287(39): 32346-53, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22865885

RESUMEN

The SIN3A-HDAC complex deacetylates histones thereby repressing gene transcription. Here we describe family with sequence similarity 60A (FAM60A), a cell cycle-regulated protein that binds to the SIN3-HDAC complex. FAM60A expression peaks during G(1) and S phases of the cell cycle in U2OS cells, in a manner similar to the G(1) regulator cyclin D1, which is a known target of SIN3-HDAC. In this light we found that FAM60A binds to SIN3-HDAC-regulated promoters such as cyclin D1 in G(1) and S phases. Cells depleted of FAM60A show increased histone acetylation at the cyclin D1 promoter and elevated levels of cyclin D1 mRNA and protein. Furthermore, depletion of FAM60A altered the periodic association of HDAC1 with the cyclin D1 promoter, increased cyclin D1 expression at all cell cycle phases, and caused premature S phase entry. The data in this study introduce FAM60A as a novel regulator of SIN3-HDAC function and gene expression.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Fase G1/fisiología , Regulación de la Expresión Génica/fisiología , Histona Desacetilasa 1/metabolismo , Subunidades de Proteína/biosíntesis , Fase S/fisiología , Complejo Correpresor Histona Desacetilasa y Sin3/metabolismo , Acetilación , Ciclina D1/biosíntesis , Ciclina D1/genética , Proteínas de Unión al ADN/genética , Células HEK293 , Histona Desacetilasa 1/genética , Humanos , Unión Proteica , Subunidades de Proteína/genética , Complejo Correpresor Histona Desacetilasa y Sin3/genética
20.
Life Sci Alliance ; 6(7)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37188479

RESUMEN

The NEK1 kinase controls ciliogenesis, mitosis, and DNA repair, and NEK1 mutations cause human diseases including axial spondylometaphyseal dysplasia and amyotrophic lateral sclerosis. C21ORF2 mutations cause a similar pattern of human diseases, suggesting close functional links with NEK1 Here, we report that endogenous NEK1 and C21ORF2 form a tight complex in human cells. A C21ORF2 interaction domain "CID" at the C-terminus of NEK1 is necessary for its association with C21ORF2 in cells, and pathogenic mutations in this region disrupt the complex. AlphaFold modelling predicts an extended binding interface between a leucine-rich repeat domain in C21ORF2 and the NEK1-CID, and our model may explain why pathogenic mutations perturb the complex. We show that NEK1 mutations that inhibit kinase activity or weaken its association with C21ORF2 severely compromise ciliogenesis, and that C21ORF2, like NEK1 is required for homologous recombination. These data enhance our understanding of how the NEK1 kinase is regulated, and they shed light on NEK1-C21ORF2-associated diseases.


Asunto(s)
Reparación del ADN , Osteocondrodisplasias , Humanos , Mutación/genética , Quinasa 1 Relacionada con NIMA/genética , Osteocondrodisplasias/genética , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA