Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Clin Pathol ; 76(2): 73-75, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36600573

RESUMEN

The partner and localiser of BRCA2 (PALB2) gene, located on chromosome 16, functions as a tumour suppressor that plays a critical role in homologous recombination repair after DNA double-strand breaks. It encodes proteins involved in the BRCA2 and BRCA1, and RAD51 pathways. Heterozygous germline mutations in PALB2 have been implicated in the development of breast, pancreatic and ovarian cancers. Whereas biallelic mutations of PALB2 have been associated with Fanconi anaaemia. Currently, 604 distinct PALB2 variants have been discovered. However, only 140 variants are thought to be pathogenic and approximately 400 are variants of unknown significance. Further studies are needed before the presence of PLAB2 mutations can be implemented as a routine clinical biomarker.


Asunto(s)
Neoplasias de la Mama , Proteínas Supresoras de Tumor , Femenino , Humanos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Nucleares/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proteína BRCA1/genética , Reparación del ADN , Mutación , Neoplasias de la Mama/genética , Predisposición Genética a la Enfermedad
2.
Hepatology ; 77(1): 33-47, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35006619

RESUMEN

BACKGROUND AND AIMS: BRCA1 (BRCA1 DNA repair associated) and PALB2 (partner and localizer of BRCA2) interact with each other to promote homologous recombination and DNA double-strand breaks repair. The disruption of this interaction has been reported to play a role in tumorigenesis. However, its precise function in HCC remains poorly understood. APPROACH AND RESULTS: We demonstrated that mice with disrupted BRCA1-PALB2 interaction were more susceptible to HCC than wild-type mice. HCC tumors arising from these mice showed plenty of T-lymphocyte infiltration and a better response to programmed cell death 1 (PD-1) antibody treatment. Mechanistically, disruption of the BRCA1-PALB2 interaction causes persistent high level of DNA damage in HCC cells, leading to activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway in both malignant hepatocytes and M1 macrophages in the tumor microenvironment. The activated cGAS-STING pathway induces programmed cell death 1 ligand 1 expression via the STING-interferon regulatory factor 3 (IRF3)-signal transducer and activator of transcription 1 pathway, causing immunosuppression to facilitate tumorigenesis and tumor progression. Meanwhile, M1 macrophages with an activated cGAS-STING pathway could recruit T lymphocytes through the STING-IRF3 pathway, leading to T-lymphocyte infiltration in tumors. After normalizing immune responses by PD-1 antibody treatment, the infiltrating T lymphocytes attack tumor cells rapidly and effectively. CONCLUSIONS: This study reveals that persistent DNA damage caused by a defective BRCA pathway induces tumor immunosuppression and T-lymphocyte infiltration in HCC through the cGAS-STING pathway, providing insight into tumor immune microenvironment remodeling that may help improve HCC response to PD-1 antibody treatment.


Asunto(s)
Proteína BRCA1 , Carcinoma Hepatocelular , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Neoplasias Hepáticas , Animales , Ratones , Carcinogénesis , Carcinoma Hepatocelular/inmunología , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Terapia de Inmunosupresión , Neoplasias Hepáticas/inmunología , Nucleotidiltransferasas/metabolismo , Receptor de Muerte Celular Programada 1 , Linfocitos T , Microambiente Tumoral , Proteína BRCA1/metabolismo
3.
J Pathol ; 256(3): 321-334, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34846068

RESUMEN

PALB2 loss-of-function variants confer high risk of developing breast cancer. Here we present a systematic functional analysis of PALB2 splice-site variants detected in approximately 113,000 women in the large-scale sequencing project Breast Cancer After Diagnostic Gene Sequencing (BRIDGES; https://bridges-research.eu/). Eighty-two PALB2 variants at the intron-exon boundaries were analyzed with MaxEntScan. Forty-two variants were selected for the subsequent splicing functional assays. For this purpose, three splicing reporter minigenes comprising exons 1-12 were constructed. The 42 potential spliceogenic variants were introduced into the minigenes by site-directed mutagenesis and assayed in MCF-7/MDA-MB-231 cells. Splicing anomalies were observed in 35 variants, 23 of which showed no traces or minimal amounts of the expected full-length transcripts of each minigene. More than 30 different variant-induced transcripts were characterized, 23 of which were predicted to truncate the PALB2 protein. The pathogenicity of all variants was interpreted according to an in-house adaptation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG-AMP) variant classification scheme. Up to 23 variants were classified as pathogenic/likely pathogenic. Remarkably, three ±1,2 variants (c.49-2A>T, c.108+2T>C, and c.211+1G>A) were classified as variants of unknown significance, as they produced significant amounts of either in-frame transcripts of unknown impact on the PALB2 protein function or the minigene full-length transcripts. In conclusion, we have significantly contributed to the ongoing effort of identifying spliceogenic variants in the clinically relevant PALB2 cancer susceptibility gene. Moreover, we suggest some approaches to classify the findings in accordance with the ACMG-AMP rationale. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Empalme Alternativo , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Sitios de Empalme de ARN , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Bases de Datos Genéticas , Exones , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Femenino , Humanos , Células MCF-7 , Isoformas de Proteínas
4.
Genes (Basel) ; 12(12)2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34946951

RESUMEN

The tumor suppressor protein partner and localizer of BRCA2 (PALB2) orchestrates the interactions between breast cancer susceptibility proteins 1 and 2 (BRCA1, -2) that are critical for genome stability, homologous recombination (HR) and DNA repair. PALB2 mutations predispose patients to a spectrum of cancers, including breast and ovarian cancers. PALB2 localizes HR machinery to chromatin and links it with transcription through multiple DNA and protein interactions. This includes its interaction with MRG15 (Morf-related gene on chromosome 15), which is part of many transcription complexes, including the HAT-associated and the HDAC-associated complexes. This interaction is critical for PALB2 localization in actively transcribed genes, where transcription/replication conflicts lead to frequent replication stress and DNA breaks. We solved the crystal structure of the MRG15 MRG domain bound to the PALB2 peptide and investigated the effect of several PALB2 mutations, including patient-derived variants. PALB2 interacts with an extended surface of the MRG that is known to interact with other proteins. This, together with a nanomolar affinity, suggests that the binding of MRG15 partners, including PALB2, to this region is mutually exclusive. Breast cancer-related mutations of PALB2 cause only minor attenuation of the binding affinity. New data reveal the mechanism of PALB2-MRG15 binding, advancing our understanding of PALB2 function in chromosome maintenance and tumorigenesis.


Asunto(s)
Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Factores de Transcripción/metabolismo , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Cromatina , ADN/metabolismo , Daño del ADN , Reparación del ADN , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/ultraestructura , Inestabilidad Genómica , Humanos , Unión Proteica/genética , Factores de Transcripción/genética , Factores de Transcripción/ultraestructura
6.
Mol Genet Genomic Med ; 9(9): e1746, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34382369

RESUMEN

BACKGROUND: Isolated myelosarcoma of infancy is a rare presentation of acute myelogenous leukaemia (AML). Because of its rarity and early onset in infancy underlying genetic predisposition is potentially relevant in disease initiation. METHODS AND RESULTS: We report an oncologic emergency in an infant with thoracic and intraspinal aleukaemic myeloid sarcoma causing acute myelon compression and lower leg palsy. Whole-exome sequencing of the patient's germline DNA identified a rare PALB2 (OMIM 610355) variant (p.A1079S), which is located in a domain critical for the gene's proper function within the homology-directed repair pathway. In line with potential DNA damage repair defects mediated by the PALB2 deregulation, the patient's fibroblasts showed increased sensitivity towards radiation and DNA intercalating agents. CONCLUSION: Therefore, we suggest PALB2 p.A1079S as a pathogenic variant potentially contributing to the here observed patient phenotype.


Asunto(s)
Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Sarcoma Mieloide/genética , Neoplasias de la Columna Vertebral/genética , Células Cultivadas , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Fibroblastos/metabolismo , Fibroblastos/fisiología , Puntos de Control de la Fase G2 del Ciclo Celular , Mutación de Línea Germinal , Humanos , Lactante , Masculino , Sarcoma Mieloide/patología , Neoplasias de la Columna Vertebral/patología
7.
Nat Commun ; 12(1): 5016, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408138

RESUMEN

DNA damage prompts a diverse range of alterations to the chromatin landscape. The RNF168 E3 ubiquitin ligase catalyzes the mono-ubiquitination of histone H2A at lysine (K)13/15 (mUb-H2A), forming a binding module for DNA repair proteins. BRCA1 promotes homologous recombination (HR), in part, through its interaction with PALB2, and the formation of a larger BRCA1-PALB2-BRCA2-RAD51 (BRCA1-P) complex. The mechanism by which BRCA1-P is recruited to chromatin surrounding DNA breaks is unclear. In this study, we reveal that an RNF168-governed signaling pathway is responsible for localizing the BRCA1-P complex to DNA damage. Using mice harboring a Brca1CC (coiled coil) mutation that blocks the Brca1-Palb2 interaction, we uncovered an epistatic relationship between Rnf168- and Brca1CC alleles, which disrupted development, and reduced the efficiency of Palb2-Rad51 localization. Mechanistically, we show that RNF168-generated mUb-H2A recruits BARD1 through a BRCT domain ubiquitin-dependent recruitment motif (BUDR). Subsequently, BARD1-BRCA1 accumulate PALB2-RAD51 at DNA breaks via the CC domain-mediated BRCA1-PALB2 interaction. Together, these findings establish a series of molecular interactions that connect the DNA damage signaling and HR repair machinery.


Asunto(s)
Proteína BRCA1/metabolismo , Daño del ADN , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteína BRCA1/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN/genética , ADN/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Unión Proteica , Transporte de Proteínas , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
8.
Eur J Hum Genet ; 29(7): 1139-1145, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33854214

RESUMEN

Gastrointestinal stromal tumour (GIST) is a mesenchymal neoplasm arising in the gastrointestinal tract. A rare subset of GISTs are classified as wild-type GIST (wtGIST) and these are frequently associated with germline variants that affect the function of cancer predisposition genes such as the succinate dehydrogenase subunit genes (SDHA, SDHB, SDHC, SDHD) or NF1. However, despite this high heritability, familial clustering of wtGIST is extremely rare. Here, we report a mother-son diad who developed wtGIST at age 66 and 34 years, respectively. Comprehensive genetic testing revealed germline truncating variants in both SDHA (c.1534C>T (p.Arg512*)) and PALB2 (c.3113G>A (p.Trp1038*)) in both affected individuals. The mother also developed breast ductal carcinoma in-situ at age 70 years. Immunohistochemistry and molecular analysis of the wtGISTs revealed loss of SDHB expression and loss of the wild-type SDHA allele in tumour material. No allele loss was detected at PALB2 suggesting that wtGIST tumourigenesis was principally driven by succinate dehydrogenase deficiency. However, we speculate that the presence of multilocus inherited neoplasia alleles syndrome (MINAS) in this family might have contributed to the highly unusual occurrence of familial wtGIST. Systematic reporting of tumour risks and phenotypes in individuals with MINAS will facilitate the clinical interpretation of the significance of this diagnosis, which is becoming more frequent as strategies for genetic testing for hereditary cancer becomes more comprehensive.


Asunto(s)
Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Tumores del Estroma Gastrointestinal/diagnóstico , Tumores del Estroma Gastrointestinal/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Succinato Deshidrogenasa/genética , Adulto , Anciano , Alelos , Bases de Datos Genéticas , Familia , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Femenino , Genotipo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Análisis de Secuencia de ADN , Succinato Deshidrogenasa/metabolismo , Evaluación de Síntomas , Secuenciación Completa del Genoma
9.
Nature ; 591(7851): 665-670, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536619

RESUMEN

Strong connections exist between R-loops (three-stranded structures harbouring an RNA:DNA hybrid and a displaced single-strand DNA), genome instability and human disease1-5. Indeed, R-loops are favoured in relevant genomic regions as regulators of certain physiological processes through which homeostasis is typically maintained. For example, transcription termination pause sites regulated by R-loops can induce the synthesis of antisense transcripts that enable the formation of local, RNA interference (RNAi)-driven heterochromation6. Pause sites are also protected against endogenous single-stranded DNA breaks by BRCA17. Hypotheses about how DNA repair is enacted at pause sites include a role for RNA, which is emerging as a normal, albeit unexplained, regulator of genome integrity8. Here we report that a species of single-stranded, DNA-damage-associated small RNA (sdRNA) is generated by a BRCA1-RNAi protein complex. sdRNAs promote DNA repair driven by the PALB2-RAD52 complex at transcriptional termination pause sites that form R-loops and are rich in single-stranded DNA breaks. sdRNA repair operates in both quiescent (G0) and proliferating cells. Thus, sdRNA repair can occur in intact tissue and/or stem cells, and may contribute to tumour suppression mediated by BRCA1.


Asunto(s)
Proteína BRCA1/metabolismo , Reparación del ADN , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Interferencia de ARN , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Proteínas Argonautas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Factores Eucarióticos de Iniciación/metabolismo , Células HeLa , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Fase de Descanso del Ciclo Celular , Ribonucleasa III/metabolismo
10.
Cancer Cell ; 38(6): 844-856.e7, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33186520

RESUMEN

Ependymoma is the third most common pediatric tumor with posterior fossa group A (PFA) being its most aggressive subtype. Ependymomas are generally refractory to chemotherapies and thus lack any effective treatment. Here, we report that elevated expression of CXorf67 (chromosome X open reading frame 67), which frequently occurs in PFA ependymomas, suppresses homologous recombination (HR)-mediated DNA repair. Mechanistically, CXorf67 interacts with PALB2 and inhibits PALB2-BRCA2 interaction, thereby inhibiting HR repair. Concordantly, tumor cells with high CXorf67 expression levels show increased sensitivity to poly(ADP-ribose) polymerase (PARP) inhibitors, especially when combined with radiotherapy. Thus, our findings have revealed a role of CXorf67 in HR repair and suggest that combination of PARP inhibitors with radiotherapy could be an effective treatment option for PFA ependymomas.


Asunto(s)
Proteína BRCA2/metabolismo , Ependimoma/terapia , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Neoplasias Infratentoriales/terapia , Proteínas Oncogénicas/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Quimioradioterapia , Ependimoma/genética , Ependimoma/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Neoplasias Infratentoriales/genética , Neoplasias Infratentoriales/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Reparación del ADN por Recombinación/efectos de los fármacos , Reparación del ADN por Recombinación/efectos de la radiación , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Artículo en Inglés | MEDLINE | ID: mdl-32554798

RESUMEN

PALB2 (partner and localizer of BRCA2) gene encodes a protein that colocalizes with BRCA2 in nuclear foci and likely permits the stable intranuclear localization and accumulation of BRCA2PALB2 plays a critical role in maintaining genome integrity through its role in the Fanconi anemia and homologous recombination DNA repair pathways. It has a known loss-of-function disease mechanism. Biallelic PALB2 pathogenic variants have been described in autosomal recessive Fanconi anemia. Heterozygous pathogenic variants in PALB2 are associated with increased risk for female and male breast cancer and pancreatic cancer (Science 324: 217; Cancer Res 71: 2222-2229; N Engl J Med 371: 497-506). Heterozygous germline PALB2 mutations have also been observed in patients with medulloblastoma (Lancet Oncol 19: 785-798). However, PALB2-related cancer predisposition to high-grade gliomas has not been reported. Here we report a germline PALB2 pathogenic variant (c.509_510delGA, p.Arg170Ilefs*14, NM_024675.3) found in a pediatric patient with high-grade glioma. This variant was first identified by tumor sequencing using the Children's Hospital of Philadelphia (CHOP) Comprehensive Solid Tumor Panel and then confirmed to be a germline change using the CHOP Comprehensive Hereditary Cancer Panel on DNA from a blood sample of this patient. Parental studies showed that this variant was paternally inherited. Further studies are needed to illustrate if pathogenic variants in PALB2 convey increased risk to developing brain tumor. This case also highlights the potential of identifying germline mutation through tumor sequencing.


Asunto(s)
Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Glioma/genética , Niño , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Células Germinativas/metabolismo , Mutación de Línea Germinal/genética , Humanos
13.
Nucleic Acids Res ; 48(11): 6032-6052, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32427332

RESUMEN

Chromosomal double strand breaks (DSBs) can initiate several signaling events, such as ubiquitination, however the precise influence of such signaling on DSB repair outcomes remains poorly understood. With an RNA interference screen, we found that the E3 ubiquitin ligase RNF8 suppresses a deletion rearrangement mediated by canonical non-homologous end joining (C-NHEJ). We also found that RNF8 suppresses EJ without insertion/deletion mutations, which is a hallmark of C-NHEJ. Conversely, RNF8 promotes alternative EJ (ALT-EJ) events involving microhomology that is embedded from the edge of the DSB. These ALT-EJ events likely require limited end resection, whereas RNF8 is not required for single-strand annealing repair involving extensive end resection. Thus, RNF8 appears to specifically facilitate repair events requiring limited end resection, which we find is dependent on the DSB end protection factor KU. However, we also find that RNF8 is important for homology-directed repair (HDR) independently of KU, which appears linked to promoting PALB2 function. Finally, the influence of RNF8 on EJ is distinct from 53BP1 and the ALT-EJ factor, POLQ. We suggest that RNF8 mediates both ALT-EJ and HDR, but via distinct mechanisms, since only the former is dependent on KU.


Asunto(s)
Rotura Cromosómica , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Autoantígeno Ku/metabolismo , Reparación del ADN por Recombinación , Ubiquitina-Proteína Ligasas/metabolismo , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , Proteínas de Unión al ADN/química , ADN Polimerasa Dirigida por ADN/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Humanos , Mutación INDEL , Proteínas Nucleares/metabolismo , Dominios Proteicos , Interferencia de ARN , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación/genética , Eliminación de Secuencia , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ubiquitina-Proteína Ligasas/química , ADN Polimerasa theta
14.
Fam Cancer ; 19(2): 123-131, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32048105

RESUMEN

The PALB2 protein is essential to RAD51-mediated homologous recombination (HR) repair. Germline monoallelic PALB2 pathogenic variants confer significant risks for breast cancer. However, the majority of PALB2 variants remain classified as variants of unknown significance (VUS). We aim to functionally and mechanistically evaluate three novel PALB2 VUS. Patient-derived lymphoblastoid cell lines containing the VUS were analyzed for nuclear localization and foci formation of RAD51 as a measure of HR efficiency. To understand the mechanism underlying the HR deficiency, PALB2 nuclear localization was assessed using immunofluorescence studies. Among these VUS, c.3251C>T (p.Ser1084Leu) occurred in a patient with metastatic breast cancer while c.1054G>C (p.Glu352Gln) and c.1057A>G (p.Lys353Glu) were seen in patients with squamous cell carcinoma of skin and renal cell carcinoma respectively. Variant c.3251C>T was located within the WD40 domain which normally masked the nuclear export signal sequence responsible for nuclear delocalization of PALB2. Correspondingly, c.3251C>T displayed aberrant cytoplasmic localization of PALB2 which led to an impaired RAD51 nuclear localization and foci formation. On the other hand, both c.1054G>C and c.1057A>G showed intact HR functions and nuclear localization of PALB2, consistent with their locations within domains of no known function. Additionally, the prevalence of c.1054G>C was similar among healthy controls and patients with breast cancer (as seen in other studies), suggestive of its non-pathogenicity. In conclusion, our studies provided the functional evidence showing the deleterious effect of c.3251C>T, and non-deleterious effects of c.1054G>C and c.1057A>G. Using the ClinGen Pathogenicity calculator, c.3251C>T remains a VUS while c.1054G>C and c.1057A>G may be classified as likely benign variants.


Asunto(s)
Núcleo Celular/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Mutación de Línea Germinal , Mutación Missense , Neoplasias/genética , Recombinasa Rad51/metabolismo , Adulto , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Citoplasma/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Femenino , Variación Genética , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Linaje , Reparación del ADN por Recombinación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo
15.
Nat Commun ; 11(1): 819, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-32041954

RESUMEN

Loss of functional BRCA1 protein leads to defects in DNA double-strand break (DSB) repair by homologous recombination (HR) and renders cells hypersensitive to poly (ADP-ribose) polymerase (PARP) inhibitors used to treat BRCA1/2-deficient cancers. However, upon chronic treatment of BRCA1-mutant cells with PARP inhibitors, resistant clones can arise via several mechanisms, including loss of 53BP1 or its downstream co-factors. Defects in the 53BP1 axis partially restore the ability of a BRCA1-deficient cell to form RAD51 filaments at resected DSBs in a PALB2- and BRCA2-dependent manner, and thereby repair DSBs by HR. Here we show that depleting 53BP1 in BRCA1-null cells restores PALB2 accrual at resected DSBs. Moreover, we demonstrate that PALB2 DSB recruitment in BRCA1/53BP1-deficient cells is mediated by an interaction between PALB2's chromatin associated motif (ChAM) and the nucleosome acidic patch region, which in 53BP1-expressing cells is bound by 53BP1's ubiquitin-directed recruitment (UDR) domain.


Asunto(s)
Proteína BRCA1/deficiencia , Cromatina/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Recombinación Homóloga , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Secuencias de Aminoácidos , Proteína BRCA2/deficiencia , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/química , Proteína del Grupo de Complementación N de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Humanos , Nucleosomas/metabolismo
16.
Vet Comp Oncol ; 18(2): 247-255, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31518051

RESUMEN

RAD51 forms a complex with BRCA2 and plays a central role in the DNA damage response pathway that is associated with homologous recombination. The structures of RAD51 and its homologues are highly conserved from prokaryotes to higher eukaryotes. Although a large number of BRCA2 mutations have been reported, there are only a few reports on the mutations of RAD51, which have been shown in humans and dogs. However, several mutations of canine RAD51 were identified from mammary gland tumour tissues in a recent study. Some of these mutations seem to have an influence on the homo-oligomerization or interaction with "Partner and localizer of BRCA2" (PALB2). In this study, we cloned the canine PALB2 homologue and investigated the effect on its interaction with the RAD51 mutants to evaluate the alteration in the function of RAD51 mutants. The A209S and T225S mutants of RAD51 show an attenuation of the interaction between RAD51 and PALB2. These results indicate that the canine RAD51 mutations can potentially alter the homologous recombination pathways in response to DNA damage in dogs.


Asunto(s)
Enfermedades de los Perros/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Neoplasias Mamarias Animales/metabolismo , Recombinasa Rad51/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Enfermedades de los Perros/genética , Perros , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Femenino , Células HeLa , Humanos , Neoplasias Mamarias Animales/genética , Modelos Moleculares , Mutación , Conformación Proteica , Recombinasa Rad51/genética
17.
Mol Cancer Res ; 18(3): 424-435, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31685642

RESUMEN

Homologous recombination (HR) is a highly conserved pathway that can facilitate the repair of DNA double-strand breaks (DSB). Several Deubiquitinases (DUB) have been implicated as key players in DNA damage repair (DDR) through HR. Here, we report USP22, a DUB that is highly overexpressed in multiple cancer types, is necessary for HR through a direct interaction with PALB2 through its C-terminal WD40 domain. This interaction stimulates USP22 catalytic activity in vitro. Furthermore, we show USP22 is necessary for BRCA2, PALB2, and Rad51 recruitment to DSBs and this is, in part, through USP22 stabilizing BRCA2 and PALB2 levels. Taken together, our results describe a role for USP22 in DNA repair. IMPLICATIONS: This research provides new and exciting mechanistic insights into how USP22 overexpression promotes chemoresistance in lung cancer. We believe this study, and others, will help aid in developing targeted drugs toward USP22 and known binding partners for lung cancer treatment.


Asunto(s)
Roturas del ADN de Doble Cadena , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Recombinación Homóloga , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Resistencia a Antineoplásicos , Humanos , Neoplasias/genética , Transfección
18.
Sci Rep ; 9(1): 19110, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31836759

RESUMEN

Cancer cells maintain their telomeres by either re-activating telomerase or adopting the homologous recombination (HR)-based Alternative Lengthening of Telomere (ALT) pathway. Among the many prominent features of ALT cells, C-circles (CC) formation is considered to be the most specific and quantifiable biomarker of ALT. However, the molecular mechanism behind the initiation and maintenance of CC formation in ALT cells is still largely unknown. We reported previously that depletion of the FANCM complex (FANCM-FAAP24-MHF1&2) in ALT cells induced pronounced replication stress, which primarily takes place at their telomeres. Here, we characterized the changes in ALT associated phenotypes in cells deficient of the FANCM complex. We found that depletion of FAAP24 or FANCM, but not MHF1&2, induces a dramatic increase of CC formation. Most importantly, we identified multiple DNA damage response (DDR) and DNA repair pathways that stimulate the dramatic increase of CC formation in FANCM deficient cells, including the dissolvase complex (BLM-TOP3A-RMI1/2, or BTR), DNA damage checkpoint kinases (ATR and Chk1), HR proteins (BRCA2, PALB2, and Rad51), as well as proteins involved in Break-Induced Replication (BIR) (POLD1 and POLD3). In addition, FANCD2, another Fanconi Anemia (FA) protein, is also required for CC formation, likely through promoting the recruitment of BLM to the replication stressed ALT telomeres. Finally, we demonstrated that TERRA R-loops accumulate at telomeres in FANCM deficient ALT cells and downregulation of which attenuates the ALT-associated PML bodies (APBs), replication stress and CC formation. Taken together, our data suggest that FANCM prevents replisomes from stalling/collapsing at ALT telomeres by disrupting TERRA R-loops.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN , Regulación Neoplásica de la Expresión Génica , Estructuras R-Loop , Homeostasis del Telómero , Telómero/fisiología , Proteína BRCA2/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Daño del ADN , ADN Polimerasa III/metabolismo , Reparación del ADN , ADN de Cadena Simple , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Hibridación Fluorescente in Situ , Fenotipo , Recombinasa Rad51/metabolismo
19.
Nat Commun ; 10(1): 5296, 2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31757951

RESUMEN

Heterozygous carriers of germ-line loss-of-function variants in the DNA repair gene PALB2 are at a highly increased lifetime risk for developing breast cancer. While truncating variants in PALB2 are known to increase cancer risk, the interpretation of missense variants of uncertain significance (VUS) is in its infancy. Here we describe the development of a relatively fast and easy cDNA-based system for the semi high-throughput functional analysis of 48 VUS in human PALB2. By assessing the ability of PALB2 VUS to rescue the DNA repair and checkpoint defects in Palb2 knockout mouse embryonic stem (mES) cells, we identify various VUS in PALB2 that impair its function. Three VUS in the coiled-coil domain of PALB2 abrogate the interaction with BRCA1, whereas several VUS in the WD40 domain dramatically reduce protein stability. Thus, our functional assays identify damaging VUS in PALB2 that may increase cancer risk.


Asunto(s)
Neoplasias de la Mama/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Técnicas Genéticas , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Mutantes/metabolismo , Mutación Missense , Animales , ADN Complementario , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Citometría de Flujo , Predisposición Genética a la Enfermedad , Inestabilidad Genómica , Humanos , Ratones , Ratones Noqueados
20.
Elife ; 82019 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-31017574

RESUMEN

BReast Cancer Associated proteins 1 and 2 (BRCA1, -2) and Partner and Localizer of BRCA2 (PALB2) protein are tumour suppressors linked to a spectrum of malignancies, including breast cancer and Fanconi anemia. PALB2 coordinates functions of BRCA1 and BRCA2 during homology-directed repair (HDR) and interacts with several chromatin proteins. In addition to protein scaffold function, PALB2 binds DNA. The functional role of this interaction is poorly understood. We identified a major DNA-binding site of PALB2, mutations in which reduce RAD51 foci formation and the overall HDR efficiency in cells by 50%. PALB2 N-terminal DNA-binding domain (N-DBD) stimulates the function of RAD51 recombinase. Surprisingly, it possesses the strand exchange activity without RAD51. Moreover, N-DBD stimulates the inverse strand exchange and can use DNA and RNA substrates. Our data reveal a versatile DNA interaction property of PALB2 and demonstrate a critical role of PALB2 DNA binding for chromosome repair in cells.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , ARN/metabolismo , Recombinación Genética , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...