Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Nucleic Acids Res ; 52(5): 2340-2354, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38180818

RESUMEN

DNA replication stress-induced fork arrest represents a significant threat to genomic integrity. One major mechanism of replication restart involves repriming downstream of the arrested fork by PRIMPOL, leaving behind a single-stranded DNA (ssDNA) gap. Accumulation of nascent strand ssDNA gaps has emerged as a possible determinant of the cellular hypersensitivity to genotoxic agents in certain genetic backgrounds such as BRCA deficiency, but how gaps are converted into cytotoxic structures is still unclear. Here, we investigate the processing of PRIMPOL-dependent ssDNA gaps upon replication stress induced by hydroxyurea and cisplatin. We show that gaps generated in PRIMPOL-overexpressing cells are expanded in the 3'-5' direction by the MRE11 exonuclease, and in the 5'-3' direction by the EXO1 exonuclease. This bidirectional exonucleolytic gap expansion ultimately promotes their conversion into DSBs. We moreover identify the de-ubiquitinating enzyme USP1 as a critical regulator of PRIMPOL-generated ssDNA gaps. USP1 promotes gap accumulation during S-phase, and their expansion by the MRE11 and EXO1 nucleases. This activity of USP1 is linked to its role in de-ubiquitinating PCNA, suggesting that PCNA ubiquitination prevents gap accumulation during replication. Finally, we show that USP1 depletion suppresses DSB formation in PRIMPOL-overexpressing cells, highlighting an unexpected role for USP1 in promoting genomic instability under these conditions.


Asunto(s)
Replicación del ADN , ADN Polimerasa Dirigida por ADN , Proteasas Ubiquitina-Específicas , ADN/genética , Daño del ADN , ADN de Cadena Simple/genética , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Humanos , Proteasas Ubiquitina-Específicas/metabolismo
2.
Mol Cell ; 65(3): 380-392, 2017 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-28157503

RESUMEN

Proliferating cell nuclear antigen (PCNA) lies at the center of the faithful duplication of eukaryotic genomes. With its distinctive doughnut-shaped molecular structure, PCNA was originally studied for its role in stimulating DNA polymerases. However, we now know that PCNA does much more than promote processive DNA synthesis. Because of the complexity of the events involved, cellular DNA replication poses major threats to genomic integrity. Whatever predicament lies ahead for the replication fork, PCNA is there to orchestrate the events necessary to handle it. Through its many protein interactions and various post-translational modifications, PCNA has far-reaching impacts on a myriad of cellular functions.


Asunto(s)
Replicación del ADN , Antígeno Nuclear de Célula en Proliferación/química , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , ADN Polimerasa Dirigida por ADN/metabolismo , Inestabilidad Genómica , Humanos , Modelos Moleculares , Conformación Proteica
3.
Cell ; 139(7): 1222-4, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20064367

RESUMEN

Left unrepaired, DNA interstrand crosslinks represent impassable hurdles for DNA replication, and their removal is a complicated stepwise process involving a variety of enzymes. In a recent paper in Science, Knipscheer et al. (2009) demonstrate that the Fanconi Anemia protein FANCD2 promotes multiple steps of the crosslink repair process.


Asunto(s)
Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Animales , Humanos
4.
Nucleic Acids Res ; 49(13): 7224-7238, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-33978751

RESUMEN

A universal response to replication stress is replication fork reversal, where the nascent complementary DNA strands are annealed to form a protective four-way junction allowing forks to avert DNA damage while replication stress is resolved. However, reversed forks are in turn susceptible to nucleolytic digestion of the regressed nascent DNA arms and rely on dedicated mechanisms to protect their integrity. The most well studied fork protection mechanism involves the BRCA pathway and its ability to catalyze RAD51 nucleofilament formation on the reversed arms of stalled replication forks. Importantly, the inability to prevent the degradation of reversed forks has emerged as a hallmark of BRCA deficiency and underlies genome instability and chemosensitivity in BRCA-deficient cells. In the past decade, multiple factors underlying fork stability have been discovered. These factors either cooperate with the BRCA pathway, operate independently from it to augment fork stability in its absence, or act as enablers of fork degradation. In this review, we examine these novel determinants of fork stability, explore the emergent conceptual underpinnings underlying fork protection, as well as the impact of fork protection on cellular viability and cancer therapy.


Asunto(s)
Replicación del ADN , Supervivencia Celular , Cromatina/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Recombinasa Rad51/metabolismo
5.
Nucleic Acids Res ; 49(22): 12855-12869, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34871431

RESUMEN

Understanding chemoresistance mechanisms in BRCA-deficient cells will allow for identification of biomarkers for predicting tumor response to therapy, as well as the design of novel therapeutic approaches targeting this chemoresistance. Here, we show that the protein MED12, a component of the Mediator transcription regulation complex, plays an unexpected role in regulating chemosensitivity in BRCA-deficient cells. We found that loss of MED12 confers resistance to cisplatin and PARP inhibitors in both BRCA1- and BRCA2-deficient cells, which is associated with restoration of both homologous recombination and replication fork stability. Surprisingly, MED12-controlled chemosensitivity does not involve a function of the Mediator complex, but instead reflects a distinct role of MED12 in suppression of the TGFß pathway. Importantly, we show that ectopic activation of the TGFß pathway is enough to overcome the fork protection and DNA repair defects of BRCA-mutant cells, resulting in chemoresistance. Our work identifies the MED12-TGFß module as an important regulator of genomic stability and chemosensitivity in BRCA-deficient cells.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Replicación del ADN/genética , Resistencia a Antineoplásicos/genética , Complejo Mediador/genética , Factor de Crecimiento Transformador beta/genética , Antineoplásicos/farmacología , Proteína BRCA1/deficiencia , Proteína BRCA1/metabolismo , Proteína BRCA2/deficiencia , Proteína BRCA2/metabolismo , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cisplatino/farmacología , ADN/química , ADN/genética , ADN/metabolismo , Reparación del ADN , Células HeLa , Humanos , Complejo Mediador/metabolismo , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Interferencia de ARN , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo
6.
PLoS Genet ; 16(11): e1009176, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33137164

RESUMEN

The ataxia telangiectasia and Rad3-related (ATR) protein kinase is a key regulator of the cellular response to DNA damage. Due to increased amount of replication stress, cancer cells heavily rely on ATR to complete DNA replication and cell cycle progression. Thus, ATR inhibition is an emerging target in cancer therapy, with multiple ATR inhibitors currently undergoing clinical trials. Here, we describe dual genome-wide CRISPR knockout and CRISPR activation screens employed to comprehensively identify genes that regulate the cellular resistance to ATR inhibitors. Specifically, we investigated two different ATR inhibitors, namely VE822 and AZD6738, in both HeLa and MCF10A cells. We identified and validated multiple genes that alter the resistance to ATR inhibitors. Importantly, we show that the mechanisms of resistance employed by these genes are varied, and include restoring DNA replication fork progression, and prevention of ATR inhibitor-induced apoptosis. In particular, we describe a role for MED12-mediated inhibition of the TGFß signaling pathway in regulating replication fork stability and cellular survival upon ATR inhibition. Our dual genome-wide screen findings pave the way for personalized medicine by identifying potential biomarkers for ATR inhibitor resistance.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Resistencia a Antineoplásicos/genética , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Biomarcadores de Tumor/metabolismo , Sistemas CRISPR-Cas/genética , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Ensayos de Selección de Medicamentos Antitumorales , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Indoles , Complejo Mediador/genética , Complejo Mediador/metabolismo , Morfolinas , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sulfonamidas , Sulfóxidos/farmacología , Sulfóxidos/uso terapéutico , Factor de Crecimiento Transformador beta/metabolismo
7.
Am J Hum Genet ; 104(1): 179-185, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30595371

RESUMEN

Accumulation of unfolded proteins in the endoplasmic reticulum (ER) initiates a stress response mechanism to clear out the unfolded proteins by either facilitating their re-folding or inducing their degradation. When this fails, an apoptotic cascade is initiated so that the affected cell is eliminated. IRE1α is a critical sensor of the unfolded-protein response, essential for initiating the apoptotic signaling. Here, we report an infantile neurodegenerative disorder associated with enhanced activation of IRE1α and increased apoptosis. Three unrelated affected individuals with congenital microcephaly, infantile epileptic encephalopathy, and profound developmental delay were found to carry heterozygous variants (c.932T>C [p.Leu311Ser] or c.935T>C [p.Leu312Pro]) in RNF13, which codes for an IRE1α-interacting protein. Structural modeling predicted that the variants, located on the surface of the protein, would not alter overall protein folding. Accordingly, the abundance of RNF13 and IRE1α was not altered in affected individuals' cells. However, both IRE1α-mediated stress signaling and stress-induced apoptosis were increased in affected individuals' cells. These results indicate that the RNF13 variants confer gain of function to the encoded protein and thereby lead to altered signaling of the ER stress response associated with severe neurodegeneration in infancy.


Asunto(s)
Ceguera/congénito , Ceguera/genética , Insuficiencia de Crecimiento/genética , Mutación con Ganancia de Función , Heterocigoto , Microcefalia/genética , Espasmos Infantiles/genética , Ubiquitina-Proteína Ligasas/genética , Secuencia de Aminoácidos , Apoptosis , Niño , Preescolar , Discapacidades del Desarrollo/genética , Estrés del Retículo Endoplásmico , Humanos , Lactante , Masculino , Modelos Moleculares , Espasmos Infantiles/congénito , Ubiquitina-Proteína Ligasas/química , Respuesta de Proteína Desplegada
8.
Nucleic Acids Res ; 48(13): 7252-7264, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32542389

RESUMEN

The DNA damage response is essential to maintain genomic stability, suppress replication stress, and protect against carcinogenesis. The ATR-CHK1 pathway is an essential component of this response, which regulates cell cycle progression in the face of replication stress. PARP14 is an ADP-ribosyltransferase with multiple roles in transcription, signaling, and DNA repair. To understand the biological functions of PARP14, we catalogued the genetic components that impact cellular viability upon loss of PARP14 by performing an unbiased, comprehensive, genome-wide CRISPR knockout genetic screen in PARP14-deficient cells. We uncovered the ATR-CHK1 pathway as essential for viability of PARP14-deficient cells, and identified regulation of DNA replication dynamics as an important mechanistic contributor to the synthetic lethality observed. Our work shows that PARP14 is an important modulator of the response to ATR-CHK1 pathway inhibitors.


Asunto(s)
Replicación del ADN , Poli(ADP-Ribosa) Polimerasas/metabolismo , Mutaciones Letales Sintéticas , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Humanos , Poli(ADP-Ribosa) Polimerasas/genética
9.
Nucleic Acids Res ; 48(16): 9161-9180, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32797166

RESUMEN

FANCJ, a DNA helicase and interacting partner of the tumor suppressor BRCA1, is crucial for the repair of DNA interstrand crosslinks (ICL), a highly toxic lesion that leads to chromosomal instability and perturbs normal transcription. In diploid cells, FANCJ is believed to operate in homologous recombination (HR) repair of DNA double-strand breaks (DSB); however, its precise role and molecular mechanism is poorly understood. Moreover, compensatory mechanisms of ICL resistance when FANCJ is deficient have not been explored. In this work, we conducted a siRNA screen to identify genes of the DNA damage response/DNA repair regime that when acutely depleted sensitize FANCJ CRISPR knockout cells to a low concentration of the DNA cross-linking agent mitomycin C (MMC). One of the top hits from the screen was RAP80, a protein that recruits repair machinery to broken DNA ends and regulates DNA end-processing. Concomitant loss of FANCJ and RAP80 not only accentuates DNA damage levels in human cells but also adversely affects the cell cycle checkpoint, resulting in profound chromosomal instability. Genetic complementation experiments demonstrated that both FANCJ's catalytic activity and interaction with BRCA1 are important for ICL resistance when RAP80 is deficient. The elevated RPA and RAD51 foci in cells co-deficient of FANCJ and RAP80 exposed to MMC are attributed to single-stranded DNA created by Mre11 and CtIP nucleases. Altogether, our cell-based findings together with biochemical studies suggest a critical function of FANCJ to suppress incompletely processed and toxic joint DNA molecules during repair of ICL-induced DNA damage.


Asunto(s)
Proteína BRCA1/genética , Proteínas de Unión al ADN/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Inestabilidad Genómica/genética , Chaperonas de Histonas/genética , ARN Helicasas/genética , Recombinasa Rad51/genética , Inestabilidad Cromosómica/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/deficiencia , Técnicas de Inactivación de Genes , Células HeLa , Chaperonas de Histonas/deficiencia , Humanos , Mitomicina/farmacología , Reparación del ADN por Recombinación/genética
10.
J Biol Chem ; 294(27): 10619-10627, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31138652

RESUMEN

DNA-protein cross-links can interfere with chromatin architecture, block DNA replication and transcription, and interfere with DNA repair. Here we synthesized a DNA 23-mer containing a site-specific DNA-peptide cross-link (DpC) by cross-linking an 11-mer peptide to the DNA epigenetic mark 5-formylcytosine in synthetic DNA and used it to generate a DpC-containing plasmid construct. Upon replication of the DpC-containing plasmid in HEK 293T cells, approximately 9% of progeny plasmids contained targeted mutations and 5% semitargeted mutations. Targeted mutations included C→T transitions and C deletions, whereas semitargeted mutations included several base substitutions and deletions near the DpC lesion. To identify DNA polymerases involved in DpC bypass, we comparatively studied translesion synthesis (TLS) efficiency and mutagenesis of the DpC in a series of cell lines with TLS polymerase knockouts or knockdowns. Knockdown of either hPol ι or hPol ζ reduced the mutation frequency by nearly 50%. However, the most significant reduction in mutation frequency (50%-70%) was observed upon simultaneous knockout of hPol η and hPol κ with knockdown of hPol ζ, suggesting that these TLS polymerases play a critical role in error-prone DpC bypass. Because TLS efficiency of the DpC construct was not significantly affected in TLS polymerase-deficient cells, we examined a possible role of replicative DNA polymerases in their bypass and determined that hPol δ and hPol ϵ can accurately bypass the DpC. We conclude that both replicative and TLS polymerases can bypass this DpC lesion in human cells but that mutations are induced mainly by TLS polymerases.


Asunto(s)
Citosina/análogos & derivados , Replicación del ADN , ADN/química , Péptidos/química , Citosina/química , ADN/metabolismo , Aductos de ADN/química , Aductos de ADN/metabolismo , ADN Polimerasa II/metabolismo , ADN Polimerasa III/metabolismo , Cartilla de ADN/metabolismo , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Mutación , Péptidos/metabolismo
11.
Am J Hum Genet ; 101(2): 267-273, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28777933

RESUMEN

Ribosomal RNA (rRNA) is transcribed from rDNA by RNA polymerase I (Pol I) to produce the 45S precursor of the 28S, 5.8S, and 18S rRNA components of the ribosome. Two transcription factors have been defined for Pol I in mammals, the selectivity factor SL1, and the upstream binding transcription factor (UBF), which interacts with the upstream control element to facilitate the assembly of the transcription initiation complex including SL1 and Pol I. In seven unrelated affected individuals, all suffering from developmental regression starting at 2.5-7 years, we identified a heterozygous variant, c.628G>A in UBTF, encoding p.Glu210Lys in UBF, which occurred de novo in all cases. While the levels of UBF, Ser388 phosphorylated UBF, and other Pol I-related components (POLR1E, TAF1A, and TAF1C) remained unchanged in cells of an affected individual, the variant conferred gain of function to UBF, manifesting by markedly increased UBF binding to the rDNA promoter and to the 5'- external transcribed spacer. This was associated with significantly increased 18S expression, and enlarged nucleoli which were reduced in number per cell. The data link neurodegeneration in childhood with altered rDNA chromatin status and rRNA metabolism.


Asunto(s)
Encefalopatías/genética , Nucléolo Celular/patología , Enfermedades Neurodegenerativas/genética , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , ARN Ribosómico 18S/biosíntesis , Adolescente , Adulto , Atrofia/genética , Encéfalo/patología , Encefalopatías/patología , Niño , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Humanos , Masculino , Enfermedades Neurodegenerativas/patología , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Adulto Joven
12.
Mol Cell ; 45(1): 75-86, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22153967

RESUMEN

Inappropriate homologous recombination (HR) causes genomic instability and cancer. In yeast, the UvrD family helicase Srs2 is recruited to sites of DNA replication by SUMO-modified PCNA, where it acts to restrict HR by disassembling toxic RAD51 nucleofilaments. How human cells control recombination at replication forks is unknown. Here, we report that the protein PARI, containing a UvrD-like helicase domain, is a PCNA-interacting partner required for preservation of genome stability in human and DT40 chicken cells. Using cell-based and biochemical assays, we show that PARI restricts unscheduled recombination by interfering with the formation of RAD51-DNA HR structures. Finally, we show that PARI knockdown suppresses the genomic instability of Fanconi Anemia/BRCA pathway-deficient cells. Thus, we propose that PARI is a long sought-after factor that suppresses inappropriate recombination events at mammalian replication forks.


Asunto(s)
Proteínas Portadoras/fisiología , Recombinación Homóloga/fisiología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Pollos/genética , Reparación del ADN , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Inestabilidad Genómica , Células HEK293 , Células HeLa , Humanos , Recombinasa Rad51/metabolismo
13.
Nucleic Acids Res ; 46(17): 8908-8916, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30032250

RESUMEN

During carcinogenesis, cells are exposed to increased replication stress due to replication fork arrest at sites of DNA lesions and difficult to replicate genomic regions. Efficient fork restart and DNA repair are important for cancer cell proliferation. We previously showed that the ADP-ribosyltransferase PARP10 interacts with the replication protein proliferating cell nuclear antigen and promotes lesion bypass by recruiting specialized, non-replicative DNA polymerases. Here, we show that PARP10 is overexpressed in a large proportion of human tumors. To understand the role of PARP10 in cellular transformation, we inactivated PARP10 in HeLa cancer cells by CRISPR/Cas9-mediated gene knockout, and overexpressed it in non-transformed RPE-1 cells. We found that PARP10 promotes cellular proliferation, and its overexpression alleviates cellular sensitivity to replication stress and fosters the restart of stalled replication forks. Importantly, mouse xenograft studies showed that loss of PARP10 reduces the tumorigenesis activity of HeLa cells, while its overexpression results in tumor formation by non-transformed RPE-1 cells. Our findings indicate that PARP10 promotes cellular transformation, potentially by alleviating replication stress and suggest that targeting PARP10 may represent a novel therapeutic approach.


Asunto(s)
Carcinogénesis/genética , Proteínas de Neoplasias/fisiología , Poli(ADP-Ribosa) Polimerasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Sistemas CRISPR-Cas , División Celular , Línea Celular Transformada , Daño del ADN , Replicación del ADN , Femenino , Técnicas de Inactivación de Genes , Células HeLa , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Proteínas de Neoplasias/deficiencia , Poli(ADP-Ribosa) Polimerasas/deficiencia , Proteínas Proto-Oncogénicas/deficiencia , Epitelio Pigmentado de la Retina/citología , Regulación hacia Arriba
14.
Nucleic Acids Res ; 46(17): 8898-8907, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30032296

RESUMEN

BRCA proteins are essential for homologous recombination (HR) DNA repair, and their germline or somatic inactivation is frequently observed in human tumors. Understanding the molecular mechanisms underlying the response of BRCA-deficient tumors to chemotherapy is paramount for developing improved personalized cancer therapies. While PARP inhibitors have been recently approved for treatment of BRCA-mutant breast and ovarian cancers, not all patients respond to this therapy, and resistance to these novel drugs remains a major clinical problem. Several mechanisms of chemoresistance in BRCA2-deficient cells have been identified. Rather than restoring normal recombination, these mechanisms result in stabilization of stalled replication forks, which can be subjected to degradation in BRCA2-mutated cells. Here, we show that the transcriptional repressor E2F7 modulates the chemosensitivity of BRCA2-deficient cells. We found that BRCA2-deficient cells are less sensitive to PARP inhibitor and cisplatin treatment after E2F7 depletion. Moreover, we show that the mechanism underlying this activity involves increased expression of RAD51, a target for E2F7-mediated transcriptional repression, which enhances both HR DNA repair, and replication fork stability in BRCA2-deficient cells. Our work describes a new mechanism of therapy resistance in BRCA2-deficient cells, and identifies E2F7 as a putative biomarker for tumor response to PARP inhibitor therapy.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA2/deficiencia , Resistencia a Antineoplásicos/fisiología , Factor de Transcripción E2F7/fisiología , Inhibidores Enzimáticos/farmacología , Proteínas de Neoplasias/fisiología , Sistemas CRISPR-Cas , Línea Celular Tumoral , Replicación del ADN/efectos de los fármacos , Replicación del ADN/fisiología , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Factor de Transcripción E2F7/deficiencia , Técnicas de Inactivación de Genes , Genes BRCA2 , Humanos , Proteínas de Neoplasias/deficiencia , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasas , Recombinasa Rad51/biosíntesis , Recombinasa Rad51/genética , Reparación del ADN por Recombinación/efectos de los fármacos , Reparación del ADN por Recombinación/fisiología
15.
Pharmacology ; 105(1-2): 19-27, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31645049

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is a common and lethal cancer of the central nervous system. This cancer is difficult to treat because most anticancer therapeutics do not readily penetrate into the brain due to the tight control at the cerebrovascular barrier. Numerous studies have suggested that dopamine D2 receptor (D2R) antagonists, such as first generation antipsychotics, may have anticancer efficacy in vivo and in vitro. The role of the D2R itself in the anticancer effects is unclear, but there is evidence suggesting that D2R activation promotes stem-like and spheroid forming behaviors in GBM. OBJECTIVES: We aimed to observe the role of the dopamine D2R and its modulators (at selective concentrations) in spheroid formation and stemness of GBM cell line, U87MG, to clarify the validity of the D2R as a therapeutic target for cancer therapy. METHODS: Spheroid formation assays and Western blotting of the glioblastoma cell line, U87MG, were used to observe responses to treatment with the D2R agonists sumanirole, ropinirole, and 4-propyl-9-hydroxynaphthoxazine (PHNO); and the D2R antagonists thioridazine, pimozide, haloperidol, and remoxipride. Extreme limiting dilution analysis was done to determine the impact of sumanirole and remoxipride treatment on sphere-forming cell frequency. Proliferation was also measured by crystal violet staining. Stable lentiviral transduction of DRD2 or shDRD2 was used to validate the role of the D2R in assay behaviors. RESULTS: D2R antagonists thioridazine, pimozide, haloperidol, and remoxipride decrease spheroid formation behaviors at a selective 100 nmol/L concentration, while D2R agonists PHNO, sumanirole, and ropinirole increase the formation of spheroids. Similarly, 100 nmol/L remoxipride decreased sphere-forming cell frequency. These results were recapitulated with genetic overexpression and knockdown of the D2R, and combination experiments indicate that the D2R is required for the effects of the pharmacological modulators. Furthermore, spheroid proliferation and invasive capacity increased under treatment with 100 nmol/L sumanirole and decreased under treatment with 100 nmol/L thioridazine. Expression levels of the stemness markers Nestin and Sox2, as well as those of differentiation marker glial fibrillary acidic protein, were not altered by 100 nmol/L thioridazine or sumanirole for 72 h or continuous treatment with these compounds for 7 days during a spheroid formation assay. CONCLUSIONS: Signaling activity of the dopamine D2R may be involved in the spheroid formation phenotype in the context of the U87MG cell line. However, this modulation may not be due to alterations in stemness marker expression, but due to other factors that may contribute to spheroid formation, such as cell-cell adhesion or EGFR signaling.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptores de Dopamina D2/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Antagonistas de los Receptores de Dopamina D2/farmacología , Glioblastoma/genética , Glioblastoma/patología , Humanos , Fenotipo , ARN Interferente Pequeño/genética , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/genética , Esferoides Celulares
16.
Mol Carcinog ; 58(10): 1770-1782, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31219654

RESUMEN

Epithelial ovarian cancer (EOC) has one of the highest death to incidence ratios among all cancers. High grade serous ovarian carcinoma (HGSOC) is the most common and deadliest EOC histotype due to the lack of therapeutic options following debulking surgery and platinum/taxane-based chemotherapies. For recurrent chemosensitive HGSOC, poly(ADP)-ribose polymerase inhibitors (PARPi; olaparib, rucaparib, or niraparib) represent an emerging treatment strategy. While PARPi are most effective in homologous recombination DNA repair-deficient (HRD) HGSOCs, recent studies have observed a significant benefit in non-HRD HGSOCs. However, all HGSOC patients are likely to acquire resistance. Therefore, there is an urgent clinical need to understand PARPi resistance and to introduce novel combinatorial therapies to manage PARPi resistance and extend HGSOC disease-free intervals. In a panel of HGSOC cell lines, we established matched olaparib sensitive and resistant cells. Transcriptome analysis of the matched olaparib-sensitive vs -resistant cells revealed activation of the Wnt signaling pathway and consequently increased TCF transcriptional activity in PARPi-resistant cells. Forced activation of canonical Wnt signaling in several PARPi-sensitive cells via WNT3A reduced olaparib and rucaparib sensitivity. PARPi resistant cells were sensitive to inhibition of Wnt signaling using the FDA-approved compound, pyrvinium pamoate, which has been shown to promote downregulation of ß-catenin. In both an HGSOC cell line and a patient-derived xenograft model, we observed that combining pyrvinium pamoate with olaparib resulted in a significant decrease in tumor burden. This study demonstrates that Wnt signaling can mediate PARPi resistance in HGSOC and provides a clinical rationale for combining PARP and Wnt inhibitors.


Asunto(s)
Neoplasias Ováricas/tratamiento farmacológico , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/genética , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Indazoles/farmacología , Indoles/farmacología , Ratones , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Piperidinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos
17.
Mol Cell ; 42(2): 141-3, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21504827

RESUMEN

In this issue of Molecular Cell, Lin et al. (2011) describe how HLTF and SHPRH, the human homologs of yeast Rad5, can discriminate between MMS-induced versus UV-induced DNA damage. The results have important implications for the suppression of damage-specific mutagenesis and for the maintenance of genomic stability.

18.
Genes Dev ; 25(17): 1847-58, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21896657

RESUMEN

The USP1/UAF1 complex deubiquitinates the Fanconi anemia protein FANCD2, thereby promoting homologous recombination and DNA cross-link repair. How USP1/UAF1 is targeted to the FANCD2/FANCI heterodimer has remained unknown. Here we show that UAF1 contains a tandem repeat of SUMO-like domains in its C terminus (SLD1 and SLD2). SLD2 binds directly to a SUMO-like domain-interacting motif (SIM) on FANCI. Deletion of the SLD2 sequence of UAF1 or mutation of the SIM on FANCI disrupts UAF1/FANCI binding and inhibits FANCD2 deubiquitination and DNA repair. The USP1/UAF1 complex also deubiquitinates PCNA-Ub, and deubiquitination requires the PCNA-binding protein hELG1. The SLD2 sequence of UAF1 binds to a SIM on hELG1, thus targeting the USP1/UAF1 complex to its PCNA-Ub substrate. We propose that the regulated targeting of USP1/UAF1 to its DNA repair substrates, FANCD2-Ub and PCNA-Ub, by SLD-SIM interactions coordinates homologous recombination and translesion DNA synthesis.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Regulación de la Expresión Génica , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Arabidopsis , Línea Celular , Pollos , Proteínas de Unión al ADN/metabolismo , Endopeptidasas/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/química , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Recombinación Genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/química , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Secuencias Repetidas en Tándem/genética , Ubiquitina/metabolismo , Proteasas Ubiquitina-Específicas
19.
EMBO Rep ; 17(6): 874-86, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27146073

RESUMEN

Defects in DNA replication, DNA damage response, and DNA repair compromise genomic stability and promote cancer development. In particular, unrepaired DNA lesions can arrest the progression of the DNA replication machinery during S-phase, causing replication stress, mutations, and DNA breaks. HUWE1 is a HECT-type ubiquitin ligase that targets proteins involved in cell fate, survival, and differentiation. Here, we report that HUWE1 is essential for genomic stability, by promoting replication of damaged DNA We show that HUWE1-knockout cells are unable to mitigate replication stress, resulting in replication defects and DNA breakage. Importantly, we find that this novel role of HUWE1 requires its interaction with the replication factor PCNA, a master regulator of replication fork restart, at stalled replication forks. Finally, we provide evidence that HUWE1 mono-ubiquitinates H2AX to promote signaling at stalled forks. Altogether, our work identifies HUWE1 as a novel regulator of the replication stress response.


Asunto(s)
Replicación del ADN , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estrés Fisiológico , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular , Daño del ADN , Reparación del ADN , Técnicas de Inactivación de Genes , Inestabilidad Genómica , Histonas/metabolismo , Humanos , Fenotipo , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Supresoras de Tumor , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
20.
Annu Rev Genet ; 43: 223-49, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19686080

RESUMEN

Fanconi Anemia (FA) is an inherited genomic instability disorder, caused by mutations in genes regulating replication-dependent removal of interstrand DNA crosslinks. The Fanconi Anemia pathway is thought to coordinate a complex mechanism that enlists elements of three classic DNA repair pathways, namely homologous recombination, nucleotide excision repair, and mutagenic translesion synthesis, in response to genotoxic insults. To this end, the Fanconi Anemia pathway employs a unique nuclear protein complex that ubiquitinates FANCD2 and FANCI, leading to formation of DNA repair structures. Lack of obvious enzymatic activities among most FA members has made it challenging to unravel its precise modus operandi. Here we review the current understanding of how the Fanconi Anemia pathway components participate in DNA repair and discuss the mechanisms that regulate this pathway to ensure timely, efficient, and correct restoration of chromosomal integrity.


Asunto(s)
Reparación del ADN , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Redes y Vías Metabólicas , Animales , Genoma , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA