Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Rep Med ; 5(3): 101471, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38508142

RESUMEN

Drug-tolerant persisters (DTPs) are a rare subpopulation of cells within a tumor that can survive therapy through nongenetic adaptive mechanisms to develop relapse and repopulate the tumor following drug withdrawal. Using a cancer cell line with an engineered suicide switch to kill proliferating cells, we perform both genetic screens and compound screens to identify the inhibition of bromodomain and extraterminal domain (BET) proteins as a selective vulnerability of DTPs. BET inhibitors are especially detrimental to DTPs that have reentered the cell cycle (DTEPs) in a broad spectrum of cancer types. Mechanistically, BET inhibition induces lethal levels of ROS through the suppression of redox-regulating genes highly expressed in DTPs, including GPX2, ALDH3A1, and MGST1. In vivo BET inhibitor treatment delays tumor relapse in both melanoma and lung cancer. Our study suggests that combining standard of care therapy with BET inhibitors to eliminate residual persister cells is a promising therapeutic strategy.


Asunto(s)
Neoplasias Pulmonares , Recurrencia Local de Neoplasia , Humanos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética
2.
Cancer Discov ; 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38533987

RESUMEN

Cancer homeostasis depends on a balance between activated oncogenic pathways driving tumorigenesis and engagement of stress-response programs that counteract the inherent toxicity of such aberrant signaling. While inhibition of oncogenic signaling pathways has been explored extensively, there is increasing evidence that overactivation of the same pathways can also disrupt cancer homeostasis and cause lethality. We show here that inhibition of Protein Phosphatase 2A (PP2A) hyperactivates multiple oncogenic pathways and engages stress responses in colon cancer cells. Genetic and compound screens identify combined inhibition of PP2A and WEE1 as synergistic in multiple cancer models by collapsing DNA replication and triggering premature mitosis followed by cell death. This combination also suppressed the growth of patient-derived tumors in vivo. Remarkably, acquired resistance to this drug combination suppressed the ability of colon cancer cells to form tumors in vivo. Our data suggest that paradoxical activation of oncogenic signaling can result in tumor suppressive resistance.

3.
EMBO J ; 43(6): 1015-1042, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38360994

RESUMEN

Targeting poly(ADP-ribose) glycohydrolase (PARG) is currently explored as a therapeutic approach to treat various cancer types, but we have a poor understanding of the specific genetic vulnerabilities that would make cancer cells susceptible to such a tailored therapy. Moreover, the identification of such vulnerabilities is of interest for targeting BRCA2;p53-deficient tumors that have acquired resistance to poly(ADP-ribose) polymerase inhibitors (PARPi) through loss of PARG expression. Here, by performing whole-genome CRISPR/Cas9 drop-out screens, we identify various genes involved in DNA repair to be essential for the survival of PARG;BRCA2;p53-deficient cells. In particular, our findings reveal EXO1 and FEN1 as major synthetic lethal interactors of PARG loss. We provide evidence for compromised replication fork progression, DNA single-strand break repair, and Okazaki fragment processing in PARG;BRCA2;p53-deficient cells, alterations that exacerbate the effects of EXO1/FEN1 inhibition and become lethal in this context. Since this sensitivity is dependent on BRCA2 defects, we propose to target EXO1/FEN1 in PARPi-resistant tumors that have lost PARG activity. Moreover, EXO1/FEN1 targeting may be a useful strategy for enhancing the effect of PARG inhibitors in homologous recombination-deficient tumors.


Asunto(s)
Neoplasias , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Reparación del ADN , Daño del ADN , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Endonucleasas de ADN Solapado/genética , Endonucleasas de ADN Solapado/metabolismo , Endonucleasas de ADN Solapado/uso terapéutico , Exodesoxirribonucleasas/genética , Enzimas Reparadoras del ADN/genética
4.
Cell Rep ; 42(10): 113293, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37858468

RESUMEN

Although distinct epithelial cell types have been distinguished in glandular tissues such as the mammary gland, the extent of heterogeneity within each cell type and the degree of endocrine control of this diversity across development are incompletely understood. By combining mass cytometry and cyclic immunofluorescence, we define a rich array of murine mammary epithelial cell subtypes associated with puberty, the estrous cycle, and sex. These subtypes are differentially proliferative and spatially segregate distinctly in adult versus pubescent glands. Further, we identify systematic suppression of lineage programs at the protein and RNA levels as a common feature of mammary epithelial expansion during puberty, the estrous cycle, and gestation and uncover a pervasive enrichment of ribosomal protein genes in luminal cells elicited specifically during progesterone-dominant expansionary periods. Collectively, these data expand our knowledge of murine mammary epithelial heterogeneity and connect endocrine-driven epithelial expansion with lineage suppression.


Asunto(s)
Señales (Psicología) , Glándulas Mamarias Animales , Ratones , Animales , Glándulas Mamarias Animales/metabolismo , ARN/metabolismo , Proliferación Celular , Análisis Espacial , Células Epiteliales/metabolismo
5.
Breast Cancer Res ; 25(1): 51, 2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-37147730

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) is a subtype of breast cancer with limited treatment options and poor clinical prognosis. Inhibitors of transcriptional CDKs are currently under thorough investigation for application in the treatment of multiple cancer types, including breast cancer. These studies have raised interest in combining these inhibitors, including CDK12/13 inhibitor THZ531, with a variety of other anti-cancer agents. However, the full scope of these potential synergistic interactions of transcriptional CDK inhibitors with kinase inhibitors has not been systematically investigated. Moreover, the mechanisms behind these previously described synergistic interactions remain largely elusive. METHODS: Kinase inhibitor combination screenings were performed to identify kinase inhibitors that synergize with CDK7 inhibitor THZ1 and CDK12/13 inhibitor THZ531 in TNBC cell lines. CRISPR-Cas9 knockout screening and transcriptomic evaluation of resistant versus sensitive cell lines were performed to identify genes critical for THZ531 resistance. RNA sequencing analysis after treatment with individual and combined synergistic treatments was performed to gain further insights into the mechanism of this synergy. Kinase inhibitor screening in combination with visualization of ABCG2-substrate pheophorbide A was used to identify kinase inhibitors that inhibit ABCG2. Multiple transcriptional CDK inhibitors were evaluated to extend the significance of the found mechanism to other transcriptional CDK inhibitors. RESULTS: We show that a very high number of tyrosine kinase inhibitors synergize with the CDK12/13 inhibitor THZ531. Yet, we identified the multidrug transporter ABCG2 as key determinant of THZ531 resistance in TNBC cells. Mechanistically, we demonstrate that most synergistic kinase inhibitors block ABCG2 function, thereby sensitizing cells to transcriptional CDK inhibitors, including THZ531. Accordingly, these kinase inhibitors potentiate the effects of THZ531, disrupting gene expression and increasing intronic polyadenylation. CONCLUSION: Overall, this study demonstrates the critical role of ABCG2 in limiting the efficacy of transcriptional CDK inhibitors and identifies multiple kinase inhibitors that disrupt ABCG2 transporter function and thereby synergize with these CDK inhibitors. These findings therefore further facilitate the development of new (combination) therapies targeting transcriptional CDKs and highlight the importance of evaluating the role of ABC transporters in synergistic drug-drug interactions in general.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas Ciclina-Dependientes/genética , Pirimidinas/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas de Neoplasias
6.
Oncogene ; 41(1): 112-124, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34703030

RESUMEN

Intratumoral heterogeneity has been described for various tumor types and models of human cancer, and can have profound effects on tumor progression and drug resistance. This study describes an in-depth analysis of molecular and functional heterogeneity among subclonal populations (SCPs) derived from a single triple-negative breast cancer cell line, including copy number analysis, whole-exome and RNA sequencing, proteome analysis, and barcode analysis of clonal dynamics, as well as functional assays. The SCPs were found to have multiple unique genetic alterations and displayed significant variation in anchorage independent growth and tumor forming ability. Analyses of clonal dynamics in SCP mixtures using DNA barcode technology revealed selection for distinct clonal populations in different in vitro and in vivo environmental contexts, demonstrating that in vitro propagation of cancer cell lines using different culture conditions can contribute to the establishment of unique strains. These analyses also revealed strong enrichment of a single SCP during the development of xenograft tumors in immune-compromised mice. This SCP displayed attenuated interferon signaling in vivo and reduced sensitivity to the antiproliferative effects of type I interferons. Reduction in interferon signaling was found to provide a selective advantage within the xenograft microenvironment specifically. In concordance with the previously described role of interferon signaling as tumor suppressor, these findings suggest that similar selective pressures may be operative in human cancer and patient-derived xenograft models.


Asunto(s)
Heterogeneidad Genética , Neoplasias de la Mama Triple Negativas/genética , Microambiente Tumoral/genética , Animales , Humanos , Ratones , Mutación , Neoplasias de la Mama Triple Negativas/patología
7.
Nat Commun ; 11(1): 5799, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33199705

RESUMEN

The extent and importance of functional heterogeneity and crosstalk between tumor cells is poorly understood. Here, we describe the generation of clonal populations from a patient-derived ovarian clear cell carcinoma model which forms malignant ascites and solid peritoneal tumors upon intraperitoneal transplantation in mice. The clonal populations are engineered with secreted Gaussia luciferase to monitor tumor growth dynamics and tagged with a unique DNA barcode to track their fate in multiclonal mixtures during tumor progression. Only one clone, CL31, grows robustly, generating exclusively malignant ascites. However, multiclonal mixtures form large solid peritoneal metastases, populated almost entirely by CL31, suggesting that transient cooperative interclonal interactions are sufficient to promote metastasis of CL31. CL31 uniquely harbors ERBB2 amplification, and its acquired metastatic activity in clonal mixtures is dependent on transient exposure to amphiregulin, which is exclusively secreted by non-tumorigenic clones. Amphiregulin enhances CL31 mesothelial clearance, a prerequisite for metastasis. These findings demonstrate that transient, ostensibly innocuous tumor subpopulations can promote metastases via "hit-and-run" commensal interactions.


Asunto(s)
Comunicación Celular , Células Clonales/patología , Metástasis de la Neoplasia/patología , Anfirregulina/metabolismo , Animales , Ascitis/patología , Carcinogénesis/patología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular , Separación Celular , Estudios de Cohortes , Variaciones en el Número de Copia de ADN/genética , Epitelio/patología , Femenino , Amplificación de Genes , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Ligandos , Ratones SCID , Modelos Biológicos , Neoplasias Peritoneales/secundario , Fenotipo , Receptor ErbB-2/genética , Factores de Tiempo
8.
Mol Cell ; 80(5): 828-844.e6, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33128871

RESUMEN

Cancer-associated mutations that stabilize NRF2, an oxidant defense transcription factor, are predicted to promote tumor development. Here, utilizing 3D cancer spheroid models coupled with CRISPR-Cas9 screens, we investigate the molecular pathogenesis mediated by NRF2 hyperactivation. NRF2 hyperactivation was necessary for proliferation and survival in lung tumor spheroids. Antioxidant treatment rescued survival but not proliferation, suggesting the presence of distinct mechanisms. CRISPR screens revealed that spheroids are differentially dependent on the mammalian target of rapamycin (mTOR) for proliferation and the lipid peroxidase GPX4 for protection from ferroptosis of inner, matrix-deprived cells. Ferroptosis inhibitors blocked death from NRF2 downregulation, demonstrating a critical role of NRF2 in protecting matrix-deprived cells from ferroptosis. Interestingly, proteomics analyses show global enrichment of selenoproteins, including GPX4, by NRF2 downregulation, and targeting NRF2 and GPX4 killed spheroids overall. These results illustrate the value of spheroid culture in revealing environmental or spatial differential dependencies on NRF2 and reveal exploitable vulnerabilities of NRF2-hyperactivated tumors.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Cultivo de Célula , Proliferación Celular , Ferroptosis , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Esferoides Celulares/metabolismo , Células A549 , Humanos , Factor 2 Relacionado con NF-E2/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Esferoides Celulares/patología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
9.
Nat Commun ; 11(1): 1711, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32249764

RESUMEN

Recently, organoid technology has been used to generate a large repository of breast cancer organoids. Here we present an extensive evaluation of the ability of organoid culture technology to preserve complex stem/progenitor and differentiated cell types via long-term propagation of normal human mammary tissues. Basal/stem and luminal progenitor cells can differentiate in culture to generate mature basal and luminal cell types, including ER+ cells that have been challenging to maintain in culture. Cells associated with increased cancer risk can also be propagated. Single-cell analyses of matched organoid cultures and native tissues by mass cytometry for 38 markers provide a higher resolution representation of the multiple mammary epithelial cell types in the organoids, and demonstrate that protein expression patterns of the tissue of origin can be preserved in culture. These studies indicate that organoid cultures provide a valuable platform for studies of mammary differentiation, transformation, and breast cancer risk.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Linaje de la Célula , Glándulas Mamarias Humanas/citología , Organoides/citología , Organoides/metabolismo , Células Madre/citología , Adulto , Proteína BRCA1/genética , Neoplasias de la Mama , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Linaje de la Célula/genética , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Femenino , Humanos , Glándulas Mamarias Humanas/química , Glándulas Mamarias Humanas/metabolismo , Persona de Mediana Edad , Organoides/química , Análisis de la Célula Individual , Células Madre/química , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Adulto Joven , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
10.
J Biol Chem ; 287(19): 15533-43, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22433856

RESUMEN

Growth hormone receptor (GHR) endocytosis is a highly regulated process that depends on the binding and activity of the multimeric ubiquitin ligase, SCF(ßTrCP) (Skp Cullin F-box). Despite a specific interaction between ß-transducin repeat-containing protein (ßTrCP) and the GHR, and a strict requirement for ubiquitination activity, the receptor is not an obligatory target for SCF(ßTrCP)-directed Lys(48) polyubiquitination. We now show that also Lys(63)-linked ubiquitin chain formation is required for GHR endocytosis. We identified both the ubiquitin-conjugating enzyme Ubc13 and the ubiquitin ligase COOH terminus of Hsp70 interacting protein (CHIP) as being connected to this process. Ubc13 activity and its interaction with CHIP precede endocytosis of GHR. In addition to ßTrCP, CHIP interacts specifically with the cytosolic tails of the dimeric GHR, identifying both Ubc13 and CHIP as novel factors in the regulation of cell surface availability of GHR.


Asunto(s)
Endocitosis , Receptores de Somatotropina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Western Blotting , Línea Celular Tumoral , Humanos , Lisina/metabolismo , Microscopía Fluorescente , Unión Proteica , Multimerización de Proteína , Interferencia de ARN , Receptores de Somatotropina/química , Receptores de Somatotropina/genética , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas con Repetición de beta-Transducina/genética , Proteínas con Repetición de beta-Transducina/metabolismo
11.
J Cell Mol Med ; 16(9): 2140-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22212761

RESUMEN

The nuclear factor κB (NF-κB) signalling pathway controls important cellular events such as cell proliferation, differentiation, apoptosis and immune responses. Pathway activation occurs rapidly upon TNFα stimulation and is highly dependent on ubiquitination events. Using cytoplasmic to nuclear translocation of the NF-κB transcription factor family member p65 as a read-out, we screened a synthetic siRNA library targeting enzymes involved in ubiquitin conjugation and de-conjugation for modifiers of regulatory ubiquitination events in NF-κB signalling. We identified F-box protein only 7 (FBXO7), a component of Skp, Cullin, F-box (SCF)-ubiquitin ligase complexes, as a negative regulator of NF-κB signalling. F-box protein only 7 binds to, and mediates ubiquitin conjugation to cIAP1 and TRAF2, resulting in decreased RIP1 ubiquitination and lowered NF-κB signalling activity.


Asunto(s)
Proteínas F-Box/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas F-Box/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , FN-kappa B/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitinación
12.
Cell Cycle ; 10(3): 380-6, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21239890

RESUMEN

Cisplatin is a widely used chemotherapeutic agent to treat a variety of solid tumors. The cytotoxic mode of action of cisplatin is mediated by inducing conformational changes in DNA including intra- and inter-strand crosslink adducts. Recognition of these adducts results in the activation of the DNA damage response resulting in cell cycle arrest, repair, and potentially, apoptosis. Despite the clinical efficacy of cisplatin, many tumors are either intrinsically resistant or acquire resistance during treatment. The identification of cisplatin drug response modulators can help us understand these resistance mechanisms, provide biomarkers for treatment strategies, or provide drug targets for combination therapy. Here we discuss functional genetic screens, including one performed by us, set up to identify genes whose inhibition results in increased sensitivity to cisplatin. In summary, the validated genes identified in these screens mainly operate in DNA damage response including nucleotide excision repair, translesion synthesis, and homologous recombination.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Aductos de ADN/efectos de los fármacos , Reparación del ADN , Humanos , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética
13.
Future Oncol ; 6(11): 1789-802, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21142664

RESUMEN

In cancer research the quest continues to identify the Achilles' heel of cancer. The ideal cancer drug targets are those that are essential in tumor cells but not in normal cells. Such targets are defined as cancer-specific vulnerabilities or as synthetic lethal interactions with cancer-specific genetic lesions. The search for synthetic lethal interactions focuses on proteins that are frequently mutated but elude pharmacological inhibition, for example, RAS, or proteins that are lost in cancer cells and by definition cannot be targeted, such as the tumor suppressor genes p53, APC and RB. These genetic interactions could yield alternative, effective targets for cancer treatment. However, it remains very difficult to predict or extrapolate these synthetic lethal interactions based on existing knowledge. With the discovery of RNAi, unbiased large-scale functional genomic screens for the identification of such targets have become possible potentially leading to major advances in the treatment of cancers. In this review we will discuss the biological basis of synthetic lethal interactions in relation to existing targeted therapeutics, lessons taught by targeted therapeutics already used in the clinic and the implementation of RNAi as tool to identify such synthetic lethal interactions.


Asunto(s)
Terapia Molecular Dirigida , Neoplasias/terapia , Animales , Ensayos de Selección de Medicamentos Antitumorales/métodos , Genes ras/fisiología , Pruebas Genéticas , Humanos , Neoplasias/genética , Interferencia de ARN
14.
Mol Cancer Res ; 8(4): 592-603, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20332216

RESUMEN

The transforming growth factor beta (TGFbeta) pathway orchestrates an extensive transcriptional program that is important for many processes in the cell. For example, TGFbeta regulates cell cycle, migration, and epithelial-to-mesenchymal transition. The TGFbeta pathway has a dual role in cancer: it is involved in early-stage tumor suppression but also contributes to tumor progression by promoting invasion. To identify the novel genes involved in TGFbeta pathway signaling, we have performed a functional genetic loss-of-function screen. We screened a small interfering RNA library targeting 700 kinases and kinase-related genes in a TGFbeta-responsive reporter assay. Several genes were identified that upon knockdown could repress the reporter signal; among these are the two cellular receptors for TGFbeta. In addition to these two known components of the TGFbeta pathway, several genes were identified that were previously not linked to the TGFbeta signaling. Knockdown of one of these genes, the IRAK2 kinase, resulted not only in an impaired TGFbeta target gene response but also in a reduction of the nuclear accumulation and phosphorylation of SMAD2. In addition, suppression of interleukin-1R-associated kinase 2 expression led to a partial override of a TGFbeta-induced cell cycle arrest. Our data show that interleukin-1R-associated kinase 2 is a novel and critical component of TGFbeta signaling.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Quinasas Asociadas a Receptores de Interleucina-1/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Transporte Activo de Núcleo Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Genes cdc/fisiología , Pruebas Genéticas , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Invasividad Neoplásica/genética , ARN Interferente Pequeño/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo
15.
Mol Cell ; 28(5): 798-809, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-18082605

RESUMEN

Monoubiquitination of FANCD2 and PCNA promotes DNA repair. It causes chromatin accumulation of FANCD2 and facilitates PCNA's recruitment of translesion polymerases to stalled replication. USP1, a protease that removes monoubiquitin from FANCD2 and PCNA, was thought to reverse the DNA damage response of these substrates. We disrupted USP1 in chicken cells to dissect its role in a stable genetic system. USP1 ablation increases FANCD2 and PCNA monoubiquitination but unexpectedly results in DNA crosslinker sensitivity. This defective DNA repair is associated with constitutively chromatin-bound, monoubiquitinated FANCD2. In contrast, persistent PCNA monoubiquitination has negligible impact on DNA repair or mutagenesis. USP1 was previously shown to autocleave after DNA damage. In DT40, USP1 autocleavage is not stimulated by DNA damage, and expressing a noncleavable mutant in the USP1 knockout strain partially rescues crosslinker sensitivity. We conclude that efficient DNA crosslink repair requires FANCD2 deubiquitination, whereas FANCD2 monoubiquitination is not dependent on USP1 autocleavage.


Asunto(s)
Reactivos de Enlaces Cruzados/farmacología , Reparación del ADN/fisiología , Endopeptidasas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinación , Animales , Apoptosis , Western Blotting , Ciclo Celular , Pollos , Cromatina/metabolismo , Cisplatino/farmacología , Daño del ADN/efectos de los fármacos , Daño del ADN/fisiología , Reparación del ADN/efectos de los fármacos , Endopeptidasas/genética , Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Regulación de la Expresión Génica , Marcación de Gen , Mitomicina/farmacología , Mutagénesis Sitio-Dirigida , Mutación , Antígeno Nuclear de Célula en Proliferación/genética , Procesamiento Proteico-Postraduccional , Fracciones Subcelulares , Ubiquitina/metabolismo , Proteasas Ubiquitina-Específicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...