Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 178(1): 135-151.e19, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31251913

RESUMEN

Loss of BRCA1 p220 function often results in basal-like breast cancer (BLBC), but the underlying disease mechanism is largely opaque. In mammary epithelial cells (MECs), BRCA1 interacts with multiple proteins, including NUMB and HES1, to form complexes that participate in interstrand crosslink (ICL) DNA repair and MEC differentiation control. Unrepaired ICL damage results in aberrant transdifferentiation to a mesenchymal state of cultured, human basal-like MECs and to a basal/mesenchymal state in primary mouse luminal MECs. Loss of BRCA1, NUMB, or HES1 or chemically induced ICL damage in primary murine luminal MECs results in persistent DNA damage that triggers luminal to basal/mesenchymal transdifferentiation. In vivo single-cell analysis revealed a time-dependent evolution from normal luminal MECs to luminal progenitor-like tumor cells with basal/mesenchymal transdifferentiation during murine BRCA1 BLBC development. Growing DNA damage accompanied this malignant transformation.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Transdiferenciación Celular/genética , Daño del ADN/genética , Reparación del ADN/genética , Glándulas Mamarias Animales/patología , Animales , Proteína BRCA1/metabolismo , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/patología , Diferenciación Celular/genética , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Células HEK293 , Humanos , Células MCF-7 , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción HES-1/metabolismo , Transfección
2.
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
3.
Mol Cell ; 63(2): 277-292, 2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-27373334

RESUMEN

An abnormal differentiation state is common in BRCA1-deficient mammary epithelial cells, but the underlying mechanism is unclear. Here, we report a convergence between DNA repair and normal, cultured human mammary epithelial (HME) cell differentiation. Surprisingly, depleting BRCA1 or FANCD2 (Fanconi anemia [FA] proteins) or BRG1, a mSWI/SNF subunit, caused HME cells to undergo spontaneous epithelial-to-mesenchymal transition (EMT) and aberrant differentiation. This also occurred when wild-type HMEs were exposed to chemicals that generate DNA interstrand crosslinks (repaired by FA proteins), but not in response to double-strand breaks. Suppressed expression of ΔNP63 also occurred in each of these settings, an effect that links DNA damage to the aberrant differentiation outcome. Taken together with somatic breast cancer genome data, these results point to a breakdown in a BRCA/FA-mSWI/SNF-ΔNP63-mediated DNA repair and differentiation maintenance process in mammary epithelial cells that may contribute to sporadic breast cancer development.


Asunto(s)
Proteína BRCA1/metabolismo , Neoplasias de la Mama/prevención & control , Diferenciación Celular , Daño del ADN , ADN Helicasas/metabolismo , Reparación del ADN , Células Epiteliales/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Glándulas Mamarias Humanas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Acetaldehído/farmacología , Proteína BRCA1/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cisplatino/farmacología , ADN Helicasas/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Femenino , Formaldehído/farmacología , Humanos , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/patología , Mutación , Proteínas Nucleares/genética , Fenotipo , Interferencia de ARN , Transducción de Señal , Factores de Transcripción/genética , Transfección , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34607954

RESUMEN

BRCA1 germline mutations are associated with an increased risk of breast and ovarian cancer. Recent findings of others suggest that BRCA1 mutation carriers also bear an increased risk of esophageal and gastric cancer. Here, we employ a Brca1/Trp53 mouse model to show that unresolved replication stress (RS) in BRCA1 heterozygous cells drives esophageal tumorigenesis in a model of the human equivalent. This model employs 4-nitroquinoline-1-oxide (4NQO) as an RS-inducing agent. Upon drinking 4NQO-containing water, Brca1 heterozygous mice formed squamous cell carcinomas of the distal esophagus and forestomach at a much higher frequency and speed (∼90 to 120 d) than did wild-type (WT) mice, which remained largely tumor free. Their esophageal tissue, but not that of WT control mice, revealed evidence of overt RS as reflected by intracellular CHK1 phosphorylation and 53BP1 staining. These Brca1 mutant tumors also revealed higher genome mutation rates than those of control animals; the mutational signature SBS4, which is associated with tobacco-induced tumorigenesis; and a loss of Brca1 heterozygosity (LOH). This uniquely accelerated Brca1 tumor model is also relevant to human esophageal squamous cell carcinoma, an often lethal tumor.


Asunto(s)
Proteína BRCA1/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Pérdida de Heterocigocidad/genética , Proteína p53 Supresora de Tumor/genética , 4-Nitroquinolina-1-Óxido/toxicidad , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Modelos Animales de Enfermedad , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/inducido químicamente , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Mutación de Línea Germinal/genética , Heterocigoto , Humanos , Pérdida de Heterocigocidad/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
5.
Mol Cell ; 57(4): 636-647, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25699710

RESUMEN

The mechanisms contributing to transcription-associated genomic instability are both complex and incompletely understood. Although R-loops are normal transcriptional intermediates, they are also associated with genomic instability. Here, we show that BRCA1 is recruited to R-loops that form normally over a subset of transcription termination regions. There it mediates the recruitment of a specific, physiological binding partner, senataxin (SETX). Disruption of this complex led to R-loop-driven DNA damage at those loci as reflected by adjacent γ-H2AX accumulation and ssDNA breaks within the untranscribed strand of relevant R-loop structures. Genome-wide analysis revealed widespread BRCA1 binding enrichment at R-loop-rich termination regions (TRs) of actively transcribed genes. Strikingly, within some of these genes in BRCA1 null breast tumors, there are specific insertion/deletion mutations located close to R-loop-mediated BRCA1 binding sites within TRs. Thus, BRCA1/SETX complexes support a DNA repair mechanism that addresses R-loop-based DNA damage at transcriptional pause sites.


Asunto(s)
Proteína BRCA1/fisiología , Reparación del ADN , Modelos Genéticos , ARN Helicasas/fisiología , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Daño del ADN , ADN Helicasas , Células HeLa , Humanos , Enzimas Multifuncionales , ARN Helicasas/genética , ARN Helicasas/metabolismo , Terminación de la Transcripción Genética , Transcripción Genética
6.
Proc Natl Acad Sci U S A ; 117(4): 2084-2091, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31932421

RESUMEN

BRCA1 promotes error-free, homologous recombination-mediated repair (HRR) of DNA double-stranded breaks (DSBs). When excessive and uncontrolled, BRCA1 HRR activity promotes illegitimate recombination and genome disorder. We and others have observed that the BRCA1-associated protein RAP80 recruits BRCA1 to postdamage nuclear foci, and these chromatin structures then restrict the amplitude of BRCA1-driven HRR. What remains unclear is how this process is regulated. Here we report that both BRCA1 poly-ADP ribosylation (PARsylation) and the presence of BRCA1-bound RAP80 are critical for the normal interaction of BRCA1 with some of its partners (e.g., CtIP and BACH1) that are also known components of the aforementioned focal structures. Surprisingly, the simultaneous loss of RAP80 and failure therein of BRCA1 PARsylation results in the dysregulated accumulation in these foci of BRCA1 complexes. This in turn is associated with the intracellular development of a state of hyper-recombination and gross chromosomal disorder. Thus, physiological RAP80-BRCA1 complex formation and BRCA1 PARsylation contribute to the kinetics by which BRCA1 HRR-sustaining complexes normally concentrate in nuclear foci. These events likely contribute to aneuploidy suppression.


Asunto(s)
Proteína BRCA1/metabolismo , Proteínas de Unión al ADN/metabolismo , Chaperonas de Histonas/metabolismo , Reparación del ADN por Recombinación , Proteína BRCA1/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Cromosomas/genética , Cromosomas/metabolismo , Daño del ADN , Proteínas de Unión al ADN/genética , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Chaperonas de Histonas/genética , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Poli ADP Ribosilación , Unión Proteica
7.
Mol Cell ; 54(6): 932-945, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24857551

RESUMEN

Quiescence (G0) allows cycling cells to reversibly cease proliferation. A decision to enter quiescence is suspected of occurring early in G1, before the restriction point (R). Surprisingly, we have identified G2 as an interval during which inhibition of the protein phosphatase PP2A results in failure to exhibit stable quiescence. This effect is accompanied by shortening of the ensuing G1. The PP2A subcomplex required for stable G0 contains the B56γ B subunit. After PP2A inhibition in G2, aberrant overexpression of cyclin E occurs during mitosis and is responsible for overriding quiescence. Strikingly, suppression of Ras signaling re-establishes normal cyclin E levels during M and restores G0. These data point to PP2A-B56γ-driven Ras signaling modulation in G2 as essential for suppressing aberrant cyclin E expression during mitosis and thereby achieving normal G0 control. Thus, G2 is an interval during which the length and growth factor dependence of the next G1 interval are established.


Asunto(s)
Fase G1/genética , Fase G2/genética , Proteína Oncogénica p21(ras)/genética , Proteína Fosfatasa 2/genética , Fase de Descanso del Ciclo Celular/fisiología , Línea Celular , Ciclina E/biosíntesis , Humanos , Células MCF-7 , Mitosis/genética , Subunidades de Proteína/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/genética
8.
Genes Dev ; 28(17): 1957-75, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25184681

RESUMEN

BRCA1 is a breast and ovarian tumor suppressor. Given its numerous incompletely understood functions and the possibility that more exist, we performed complementary systematic screens in search of new BRCA1 protein-interacting partners. New BRCA1 functions and/or a better understanding of existing ones were sought. Among the new interacting proteins identified, genetic interactions were detected between BRCA1 and four of the interactors: TONSL, SETX, TCEANC, and TCEA2. Genetic interactions were also detected between BRCA1 and certain interactors of TONSL, including both members of the FACT complex. From these results, a new BRCA1 function in the response to transcription-associated DNA damage was detected. Specifically, new roles for BRCA1 in the restart of transcription after UV damage and in preventing or repairing damage caused by stabilized R loops were identified. These roles are likely carried out together with some of the newly identified interactors. This new function may be important in BRCA1 tumor suppression, since the expression of several interactors, including some of the above-noted transcription proteins, is repeatedly aberrant in both breast and ovarian cancers.


Asunto(s)
Proteína BRCA1/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Transcripción Genética/genética , Proteína BRCA1/genética , Línea Celular Tumoral , Células HeLa , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Rayos Ultravioleta
9.
Proc Natl Acad Sci U S A ; 115(41): E9600-E9609, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30254159

RESUMEN

BRCA1 is an established breast and ovarian tumor suppressor gene that encodes multiple protein products whose individual contributions to human cancer suppression are poorly understood. BRCA1-IRIS (also known as "IRIS"), an alternatively spliced BRCA1 product and a chromatin-bound replication and transcription regulator, is overexpressed in various primary human cancers, including breast cancer, lung cancer, acute myeloid leukemia, and certain other carcinomas. Its naturally occurring overexpression can promote the metastasis of patient-derived xenograft (PDX) cells and other human cancer cells in mouse models. The IRIS-driven metastatic mechanism results from IRIS-dependent suppression of phosphatase and tensin homolog (PTEN) transcription, which in turn perturbs the PI3K/AKT/GSK-3ß pathway leading to prolyl hydroxylase-independent HIF-1α stabilization and activation in a normoxic environment. Thus, despite the tumor-suppressing genetic origin of IRIS, its properties more closely resemble those of an oncoprotein that, when spontaneously overexpressed, can, paradoxically, drive human tumor progression.


Asunto(s)
Empalme Alternativo , Proteína BRCA1/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Animales , Proteína BRCA1/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Neoplasias/genética , Neoplasias/patología , Fosfohidrolasa PTEN/genética
10.
Genes Dev ; 27(20): 2274-91, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24142877

RESUMEN

Endogenous BRCA1 p220 expression peaks in S and G2 when it is activated, and the protein participates in certain key DNA damage responses. In contrast, its expression is markedly reduced in G0/G1. While variations in transcription represent a significant part of p220 expression control, there is at least one other relevant process. We found that a microRNA, miR-545, that is expressed throughout the cell cycle down-modulates endogenous p220 mRNA and protein abundance directly in both G0/G1 and S/G2. When miR-545 function was inhibited by a specific antagomir, endogenous p220 expression increased in G0/G1, and aberrant p220-associated DNA damage responses and de novo DNA strand breaks accumulated. Analogous results were observed upon inhibition of miR-545 function in S/G2. Both sets of antagomir effects were mimicked by infecting cells with a p220 cDNA-encoding adenoviral vector. Thus, strand breaks were a product of p220 overexpression, and their prevention by miR-545 depends on its modulation of p220 expression. Breaks were also dependent on aberrant, overexpressed p220-driven recruitment of RAD51 to either spontaneously arising or mutagen-based DNA damage sites. Hence, when its level is not physiologically maintained, endogenous p220 aberrantly directs at least one DNA repair protein, RAD51, to damage sites, where their action contributes to the development of de novo DNA damage. Thus, like its loss, a surfeit of endogenous p220 function represents a threat to genome integrity.


Asunto(s)
Proteína BRCA1/genética , Ciclo Celular/fisiología , Daño del ADN/genética , Regulación de la Expresión Génica , Proteína BRCA1/metabolismo , Sitios de Unión , Línea Celular Tumoral , Daño del ADN/efectos de la radiación , Reparación del ADN , Células HEK293 , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Unión Proteica , ARN Mensajero/metabolismo , Recombinasa Rad51/metabolismo , Rayos Ultravioleta
11.
Mol Cell ; 47(3): 396-409, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22704558

RESUMEN

Completion of DNA replication after replication stress depends on PCNA, which undergoes monoubiquitination to stimulate direct bypass of DNA lesions by specialized DNA polymerases or is polyubiquitinated to promote recombination-dependent DNA synthesis across DNA lesions by template switching mechanisms. Here we report that the ZRANB3 translocase, a SNF2 family member related to the SIOD disorder SMARCAL1 protein, is recruited by polyubiquitinated PCNA to promote fork restart following replication arrest. ZRANB3 depletion in mammalian cells results in an increased frequency of sister chromatid exchange and DNA damage sensitivity after treatment with agents that cause replication stress. Using in vitro biochemical assays, we show that recombinant ZRANB3 remodels DNA structures mimicking stalled replication forks and disassembles recombination intermediates. We therefore propose that ZRANB3 maintains genomic stability at stalled or collapsed replication forks by facilitating fork restart and limiting inappropriate recombination that could occur during template switching events.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN/fisiología , Inestabilidad Genómica/fisiología , Poliubiquitina/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estrés Fisiológico/genética , Secuencia de Aminoácidos , Línea Celular Tumoral , Daño del ADN/fisiología , ADN Helicasas/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Datos de Secuencia Molecular , Osteosarcoma , Unión Proteica/fisiología , Recombinación Genética/fisiología , Intercambio de Cromátides Hermanas/fisiología , Ubiquitinación/fisiología
12.
Mol Cell ; 44(2): 235-51, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21963239

RESUMEN

BRCA1 contributes to the response to UV irradiation. Utilizing its BRCT motifs, it is recruited during S/G2 to UV-damaged sites in a DNA replication-dependent but nucleotide excision repair (NER)-independent manner. More specifically, at UV-stalled replication forks, it promotes photoproduct excision, suppression of translesion synthesis, and the localization and activation of replication factor C complex (RFC) subunits. The last function, in turn, triggers post-UV checkpoint activation and postreplicative repair. These BRCA1 functions differ from those required for DSBR.


Asunto(s)
Proteína BRCA1/metabolismo , Daño del ADN , Rayos Ultravioleta , Proteína BRCA1/genética , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Replicación del ADN , Humanos , Proteína de Replicación C/genética , Proteína de Replicación C/metabolismo
13.
Proc Natl Acad Sci U S A ; 113(48): E7701-E7709, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27849576

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron dysfunction disease that leads to paralysis and death. There is currently no established molecular pathogenesis pathway. Multiple proteins involved in RNA processing are linked to ALS, including FUS and TDP43, and we propose a disease mechanism in which loss of function of at least one of these proteins leads to an accumulation of transcription-associated DNA damage contributing to motor neuron cell death and progressive neurological symptoms. In support of this hypothesis, we find that FUS or TDP43 depletion leads to increased sensitivity to a transcription-arresting agent due to increased DNA damage. Thus, these proteins normally contribute to the prevention or repair of transcription-associated DNA damage. In addition, both FUS and TDP43 colocalize with active RNA polymerase II at sites of DNA damage along with the DNA damage repair protein, BRCA1, and FUS and TDP43 participate in the prevention or repair of R loop-associated DNA damage, a manifestation of aberrant transcription and/or RNA processing. Gaining a better understanding of the role(s) that FUS and TDP43 play in transcription-associated DNA damage could shed light on the mechanisms underlying ALS pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/fisiología , Proteína FUS de Unión a ARN/fisiología , Línea Celular , Humanos , Neuronas Motoras/metabolismo , Transporte de Proteínas , Transcripción Genética
14.
Genes Dev ; 25(7): 685-700, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21406551

RESUMEN

In response to DNA double-strand breaks (DSBs), BRCA1 forms biochemically distinct complexes with certain other DNA damage response proteins. These structures, some of which are required for homologous recombination (HR)-type DSB repair, concentrate at distinct nuclear foci that demarcate sites of genome breakage. Polyubiquitin binding by one of these structures, the RAP80/BRCA1 complex, is required for efficient BRCA1 focal recruitment, but the relationship of this process to the execution of HR has been unclear. We found that this complex actively suppresses otherwise exaggerated, BRCA1-driven HR. By controlling the kinetics by which other BRCA1-interacting proteins that promote HR concentrate together with BRCA1 in nuclear foci, RAP80/BRCA1 complexes suppress excessive DSB end processing, HR-type DSB repair, and overt chromosomal instability. Since chromosomal instability emerges when BRCA1 HR function is either unbridled or absent, active tuning of BRCA1 activity, executed in nuclear foci, is important to genome integrity maintenance.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteínas Portadoras/metabolismo , Reparación del ADN , Proteínas Nucleares/metabolismo , Radiación Ionizante , Recombinación Genética , Proteínas Portadoras/genética , Línea Celular Tumoral , Inestabilidad Cromosómica , Cromosomas/química , Cromosomas/genética , Cromosomas/metabolismo , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , Células HEK293 , Células HeLa , Chaperonas de Histonas , Humanos , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética
15.
Proc Natl Acad Sci U S A ; 112(1): 232-7, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25535366

RESUMEN

High-grade serous ovarian carcinoma (HGSOC) is the most common and aggressive form of epithelial ovarian cancer, for which few targeted therapies exist. To search for new therapeutic target proteins, we performed an in vivo shRNA screen using an established human HGSOC cell line growing either subcutaneously or intraperitoneally in immunocompromised mice. We identified genes previously implicated in ovarian cancer such as AURKA1, ERBB3, CDK2, and mTOR, as well as several novel candidates including BRD4, VRK1, and GALK2. We confirmed, using both genetic and pharmacologic approaches, that the activity of BRD4, an epigenetic transcription modulator, is necessary for proliferation/survival of both an established human ovarian cancer cell line (OVCAR8) and a subset of primary serous ovarian cancer cell strains (DFs). Among the DFs tested, the strains sensitive to BRD4 inhibition revealed elevated expression of either MYCN or c-MYC, with MYCN expression correlating closely with JQ1 sensitivity. Accordingly, primary human xenografts derived from high-MYCN or c-MYC strains exhibited sensitivity to BRD4 inhibition. These data suggest that BRD4 inhibition represents a new therapeutic approach for MYC-overexpressing HGSOCs.


Asunto(s)
Pruebas Genéticas , Terapia Molecular Dirigida , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/terapia , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Factores de Transcripción/metabolismo , Animales , Carcinoma Epitelial de Ovario , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Estudios de Asociación Genética , Humanos , Ratones , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nature ; 474(7350): 230-4, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21654808

RESUMEN

Cyclin D1 is a component of the core cell cycle machinery. Abnormally high levels of cyclin D1 are detected in many human cancer types. To elucidate the molecular functions of cyclin D1 in human cancers, we performed a proteomic screen for cyclin D1 protein partners in several types of human tumours. Analyses of cyclin D1 interactors revealed a network of DNA repair proteins, including RAD51, a recombinase that drives the homologous recombination process. We found that cyclin D1 directly binds RAD51, and that cyclin D1-RAD51 interaction is induced by radiation. Like RAD51, cyclin D1 is recruited to DNA damage sites in a BRCA2-dependent fashion. Reduction of cyclin D1 levels in human cancer cells impaired recruitment of RAD51 to damaged DNA, impeded the homologous recombination-mediated DNA repair, and increased sensitivity of cells to radiation in vitro and in vivo. This effect was seen in cancer cells lacking the retinoblastoma protein, which do not require D-cyclins for proliferation. These findings reveal an unexpected function of a core cell cycle protein in DNA repair and suggest that targeting cyclin D1 may be beneficial also in retinoblastoma-negative cancers which are currently thought to be unaffected by cyclin D1 inhibition.


Asunto(s)
Ciclina D1/metabolismo , Reparación del ADN , Neoplasias/metabolismo , Mapeo de Interacción de Proteínas , Recombinasa Rad51/metabolismo , Animales , Línea Celular Tumoral , Ensayo Cometa , Ciclina D1/deficiencia , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Células HeLa , Humanos , Ratones , Neoplasias/genética , Neoplasias/patología , Unión Proteica/efectos de la radiación , Radiación Ionizante , Recombinación Genética/genética , Proteína de Retinoblastoma/deficiencia
17.
J Virol ; 89(5): 2857-65, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25540383

RESUMEN

UNLABELLED: Many of the small DNA tumor viruses encode transforming proteins that function by targeting critical cellular pathways involved in cell proliferation and survival. In this study, we have examined whether some of the functions of the polyomavirus small T antigens (ST) are shared by the E6 and E7 oncoproteins of two oncogenic papillomaviruses. Using three different assays, we have found that E7 can provide some simian virus 40 (SV40) or murine polyomavirus (PyV) ST functions. Both human papillomavirus 16 (HPV16) and bovine papillomavirus (BPV1) E7 proteins are capable of partially substituting for SV40 ST in a transformation assay that also includes SV40 large T antigen, the catalytic subunit of cellular telomerase, and oncogenic Ras. Like SV40 ST, HPV16 E7 has the ability to override a quiescence block induced by mitogen deprivation. Like PyV ST, it also has the ability to inhibit myoblast differentiation. At least two of these activities are dependent upon the interaction of HPV16 E7 with retinoblastoma protein family members. For small T antigens, interaction with PP2A is needed for each of these functions. Even though there is no strong evidence that E6 or E7 share the ability of small T to interact with PP2A, E7 provides these functions related to cellular transformation. IMPORTANCE: DNA tumor viruses have provided major insights into how cancers develop. Some viruses, like the human papillomaviruses, can cause cancer directly. Both the papillomaviruses and the polyomaviruses have served as tools for understanding pathways that are often perturbed in cancer. Here, we have compared the functions of transforming proteins from several DNA tumor viruses, including two papillomaviruses and two polyomaviruses. We tested the papillomavirus E6 and E7 oncoproteins in three functional assays and found that E7 can provide some or all of the functions of the SV40 small T antigen, another well-characterized oncoprotein, in two of these assays. In a third assay, papillomavirus E7 has the same effect as the murine polyomavirus small T protein. In summary, we report several new functions for the papillomavirus E7 proteins, which will contribute new insights into the roles of viruses in cancer and the cellular pathways they perturb in carcinogenesis.


Asunto(s)
Antígenos Transformadores de Poliomavirus/metabolismo , Transformación Celular Viral , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Represoras/metabolismo , Prueba de Complementación Genética , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/fisiología , Humanos , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/fisiología , Virus 40 de los Simios/genética , Virus 40 de los Simios/fisiología
18.
Nat Rev Cancer ; 7(8): 628-33, 2007 08.
Artículo en Inglés | MEDLINE | ID: mdl-17611545

RESUMEN

Interest has recently reawakened in whether loss of the heterochromatic X chromosome (Barr body) is prevalent in certain breast and ovarian cancers, and new insights into the mechanisms involved have emerged. Mitotic segregation errors commonly explain the loss of the inactive X chromosome (Xi), but compromise of Xi heterochromatin in some cancers may signal broader deficits of nuclear heterochromatin. The debated link between BRCA1 and Xi might reflect a general relationship between BRCA1 and heterochromatin, which could connect BRCA1 to both epigenetic and genetic instability. We suggest that heterochromatic instability is a common but largely unexplored mechanism, leading to widespread genomic misregulation and the evolution of some cancers.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias Ováricas/patología , Cromatina Sexual/patología , Neoplasias de la Mama/metabolismo , Cromosomas Humanos X , Femenino , Genes BRCA1 , Heterocromatina/metabolismo , Humanos , Neoplasias Ováricas/metabolismo , Cromatina Sexual/metabolismo
19.
Proc Natl Acad Sci U S A ; 110(21): 8632-7, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23657012

RESUMEN

Germ-line mutations in PALB2 lead to a familial predisposition to breast and pancreatic cancer or to Fanconi Anemia subtype N. PALB2 performs its tumor suppressor role, at least in part, by supporting homologous recombination-type double strand break repair (HR-DSBR) through physical interactions with BRCA1, BRCA2, and RAD51. To further understand the mechanisms underlying PALB2-mediated DNA repair and tumor suppression functions, we targeted Palb2 in the mouse. Palb2-deficient murine ES cells recapitulated DNA damage defects caused by PALB2 depletion in human cells, and germ-line deletion of Palb2 led to early embryonic lethality. Somatic deletion of Palb2 driven by K14-Cre led to mammary tumor formation with long latency. Codeletion of both Palb2 and Tumor protein 53 (Trp53) accelerated mammary tumor formation. Like BRCA1 and BRCA2 mutant breast cancers, these tumors were defective in RAD51 focus formation, reflecting a defect in Palb2 HR-DSBR function, a strongly suspected contributor to Brca1, Brca2, and Palb2 mammary tumor development. However, unlike the case of Brca1-mutant cells, Trp53bp1 deletion failed to rescue the genomic instability of Palb2- or Brca2-mutant primary lymphocytes. Therefore, Palb2-driven DNA damage control is, in part, distinct from that executed by Brca1 and more similar to that of Brca2. The mechanisms underlying Palb2 mammary tumor suppression functions can now be explored genetically in vivo.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Síndromes Neoplásicos Hereditarios/metabolismo , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Modelos Animales de Enfermedad , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Eliminación de Gen , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Mutantes , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/patología , Proteínas Nucleares/genética , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética
20.
Nat Genet ; 39(2): 159-61, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17200672

RESUMEN

The Fanconi anemia and BRCA networks are considered interconnected, as BRCA2 gene defects have been discovered in individuals with Fanconi anemia subtype D1. Here we show that a defect in the BRCA2-interacting protein PALB2 is associated with Fanconi anemia in an individual with a new subtype. PALB2-deficient cells showed hypersensitivity to cross-linking agents and lacked chromatin-bound BRCA2; these defects were corrected upon ectopic expression of PALB2 or by spontaneous reversion.


Asunto(s)
Proteína BRCA2/fisiología , Neoplasias de la Mama/genética , Anemia de Fanconi/genética , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Predisposición Genética a la Enfermedad , Humanos , Mutación , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA