Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Cell ; 77(6): 1206-1221.e7, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-31980388

RESUMEN

Alternative polyadenylation (APA) contributes to transcriptome complexity by generating mRNA isoforms with varying 3' UTR lengths. APA leading to 3' UTR shortening (3' US) is a common feature of most cancer cells; however, the molecular mechanisms are not understood. Here, we describe a widespread mechanism promoting 3' US in cancer through ubiquitination of the mRNA 3' end processing complex protein, PCF11, by the cancer-specific MAGE-A11-HUWE1 ubiquitin ligase. MAGE-A11 is normally expressed only in the male germline but is frequently re-activated in cancers. MAGE-A11 is necessary for cancer cell viability and is sufficient to drive tumorigenesis. Screening for targets of MAGE-A11 revealed that it ubiquitinates PCF11, resulting in loss of CFIm25 from the mRNA 3' end processing complex. This leads to APA of many transcripts affecting core oncogenic and tumor suppressors, including cyclin D2 and PTEN. These findings provide insights into the molecular mechanisms driving APA in cancer and suggest therapeutic strategies.


Asunto(s)
Regiones no Traducidas 3'/genética , Antígenos de Neoplasias/metabolismo , Neoplasias Pulmonares/patología , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/patología , ARN Mensajero/metabolismo , Ubiquitina/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Animales , Antígenos de Neoplasias/genética , Apoptosis , Biomarcadores de Tumor , Carcinogénesis , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Neoplasias/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Poliadenilación , Empalme del ARN , ARN Mensajero/genética , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto , Factores de Escisión y Poliadenilación de ARNm/genética
2.
Mol Cell ; 69(1): 113-125.e6, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29225034

RESUMEN

The cytosolic iron-sulfur (Fe-S) cluster assembly (CIA) pathway functions to incorporate inorganic Fe-S cofactors into a variety of proteins, including several DNA repair enzymes. However, the mechanisms regulating the CIA pathway are unknown. We describe here that the MAGE-F1-NSE1 E3 ubiquitin ligase regulates the CIA pathway through ubiquitination and degradation of the CIA-targeting protein MMS19. Overexpression or knockout of MAGE-F1 altered Fe-S incorporation into MMS19-dependent DNA repair enzymes, DNA repair capacity, sensitivity to DNA-damaging agents, and iron homeostasis. Intriguingly, MAGE-F1 has undergone adaptive pseudogenization in select mammalian lineages. In contrast, MAGE-F1 is highly amplified in multiple human cancer types and amplified tumors have increased mutational burden. Thus, flux through the CIA pathway can be regulated by degradation of the substrate-specifying MMS19 protein and its downregulation is a common feature in cancer and is evolutionarily controlled.


Asunto(s)
Proteínas Portadoras/metabolismo , Reparación del ADN/genética , Hierro/química , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Azufre/química , Factores de Transcripción/metabolismo , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Daño del ADN/genética , Células HEK293 , Células HeLa , Humanos , Proteínas Hierro-Azufre/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Ubiquitinación
3.
PLoS Biol ; 19(6): e3001281, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34077419

RESUMEN

Nutrient-responsive protein kinases control the balance between anabolic growth and catabolic processes such as autophagy. Aberrant regulation of these kinases is a major cause of human disease. We report here that the vertebrate nonreceptor tyrosine kinase Src-related kinase lacking C-terminal regulatory tyrosine and N-terminal myristylation sites (SRMS) inhibits autophagy and promotes growth in a nutrient-responsive manner. Under nutrient-replete conditions, SRMS phosphorylates the PHLPP scaffold FK506-binding protein 51 (FKBP51), disrupts the FKBP51-PHLPP complex, and promotes FKBP51 degradation through the ubiquitin-proteasome pathway. This prevents PHLPP-mediated dephosphorylation of AKT, causing sustained AKT activation that promotes growth and inhibits autophagy. SRMS is amplified and overexpressed in human cancers where it drives unrestrained AKT signaling in a kinase-dependent manner. SRMS kinase inhibition activates autophagy, inhibits cancer growth, and can be accomplished using the FDA-approved tyrosine kinase inhibitor ibrutinib. This illuminates SRMS as a targetable vulnerability in human cancers and as a new target for pharmacological induction of autophagy in vertebrates.


Asunto(s)
Autofagia , Neoplasias/metabolismo , Neoplasias/patología , Proteínas de Unión a Tacrolimus/metabolismo , Familia-src Quinasas/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia/efectos de los fármacos , Beclina-1/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Ratones , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Piperidinas/farmacología , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores
4.
Chem Soc Rev ; 51(14): 5740-5756, 2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35587208

RESUMEN

Targeted protein degradation (TPD) strategies have revolutionized how scientists tackle challenging protein targets deemed undruggable with traditional small molecule inhibitors. Many promising campaigns to inhibit proteins have failed due to factors surrounding inhibition selectivity and targeting of compounds to specific tissues and cell types. One of the major improvements that PROTAC (proteolysis targeting chimera) and molecular glue technology can exert is highly selective control of target inhibition. Multiple studies have shown that PROTACs can gain selectivity for their protein targets beyond that of their parent ligands via optimization of linker length and stabilization of ternary complexes. Due to the bifunctional nature of PROTACs, the tissue selective nature of E3 ligases can be exploited to uncover novel targeting mechanisms. In this review, we provide critical analysis of the recent progress towards making selective PROTAC molecules and new PROTAC technologies that will continue to push the boundaries of achieving selectivity. These efforts have wide implications in the future of treating disease as they will broaden the possible targets that can be addressed by small molecules, like undruggable proteins or broadly active targets that would benefit from degradation in specific tissue types.


Asunto(s)
Proteolisis , Ubiquitina-Proteína Ligasas , Ligandos , Ubiquitina-Proteína Ligasas/metabolismo
5.
Mol Cell ; 54(4): 626-38, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24768535

RESUMEN

In response to DNA damage, PCNA is mono-ubiquitinated and triggers translesion DNA synthesis (TLS) by recruiting polymerase-η. However, it remained unknown how error-prone TLS is turned off after DNA lesion bypass to prevent mutagenesis. Here we showed that ISG15 modification (ISGylation) of PCNA plays a key role in TLS termination. Upon UV irradiation, EFP, an ISG15 E3 ligase, bound to mono-ubiquitinated PCNA and promoted its ISGylation. ISGylated PCNA then tethered USP10 for deubiquitination and in turn the release of polymerase-η from PCNA. Eventually, PCNA was deISGylated by UBP43 for reloading of replicative DNA polymerases and resuming normal DNA replication. However, ISGylation-defective Lys-to-Arg mutations in PCNA or knockdown of any of ISG15, EFP, or USP10 led to persistent recruitment of mono-ubiquitinated PCNA and polymerase-η to nuclear foci, causing an increase in mutation frequency. These findings establish a crucial role of PCNA ISGylation in termination of error-prone TLS for preventing excessive mutagenesis.


Asunto(s)
Citocinas/metabolismo , Daño del ADN , Replicación del ADN , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinas/metabolismo , Arginina/metabolismo , Sitios de Unión/genética , Citocinas/genética , ADN Polimerasa II/metabolismo , Reparación del ADN , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Lisina/metabolismo , Mutagénesis , Tasa de Mutación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Ubiquitinas/genética
6.
Angew Chem Int Ed Engl ; 60(51): 26663-26670, 2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34614283

RESUMEN

Targeting cereblon (CRBN) is currently one of the most frequently reported proteolysis-targeting chimera (PROTAC) approaches, owing to favorable drug-like properties of CRBN ligands, immunomodulatory imide drugs (IMiDs). However, IMiDs are known to be inherently unstable, readily undergoing hydrolysis in body fluids. Here we show that IMiDs and IMiD-based PROTACs rapidly hydrolyze in commonly utilized cell media, which significantly affects their cell efficacy. We designed novel CRBN binders, phenyl glutarimide (PG) analogues, and showed that they retained affinity for CRBN with high ligand efficiency (LE >0.48) and displayed improved chemical stability. Our efforts led to the discovery of PG PROTAC 4 c (SJ995973), a uniquely potent degrader of bromodomain and extra-terminal (BET) proteins that inhibited the viability of human acute myeloid leukemia MV4-11 cells at low picomolar concentrations (IC50 =3 pM; BRD4 DC50 =0.87 nM). These findings strongly support the utility of PG derivatives in the design of CRBN-directed PROTACs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Piperidonas/química , Ubiquitina-Proteína Ligasas/química , Humanos , Hidrólisis , Proteolisis
7.
J Neurosci ; 33(31): 12728-38, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23904609

RESUMEN

Katanin is a heterodimeric enzyme that severs and disassembles microtubules. While the p60 subunit has the enzyme activity, the p80 subunit regulates the p60 activity. The microtubule-severing activity of katanin plays an essential role in axonal growth. However, the mechanisms by which neuronal cells regulate the expression of katanin-p60 remains unknown. Here we showed that USP47 and C terminus of Hsp70-interacting protein (CHIP) antagonistically regulate the stability of katanin-p60 and thereby axonal growth. USP47 was identified as a katanin-p60-specific deubiquitinating enzyme for its stabilization. We also identified CHIP as a ubiquitin E3 ligase that promotes proteasome-mediated degradation of katanin-p60. Moreover, USP47 promoted axonal growth of cultured rat hippocampal neurons, whereas CHIP inhibited it. Significantly, treatment with basic fibroblast growth factor (bFGF), an inducer of axonal growth, increased the levels of USP47 and katanin-p60, but not CHIP. Consistently, bFGF treatment resulted in a marked decrease in the level of ubiquitinated katanin-p60 and thereby in the promotion of axonal growth. On the other hand, the level of USP47, but not CHIP, decreased concurrently with that of katanin-p60 as axons reached their target cells. These results indicate that USP47 plays a crucial role in the control of axonal growth during neuronal development by antagonizing CHIP-mediated katanin-p60 degradation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Axones/fisiología , Proteínas del Choque Térmico HSP47/metabolismo , Neuronas/citología , Ubiquitina-Proteína Ligasas/metabolismo , Adenosina Trifosfatasas/genética , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas del Choque Térmico HSP47/genética , Hipocampo/citología , Humanos , Inmunoprecipitación , Katanina , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Transfección , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética
8.
Biochem Biophys Res Commun ; 455(3-4): 153-8, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25450678

RESUMEN

c-Cbl, a RING-type ubiquitin E3 ligase, down-regulates receptor tyrosine kinases, including EGF receptor, and inhibits cell proliferation. Moreover, c-Cbl mutations are frequently found in patients with myeloid neoplasm. Therefore, c-Cbl is known as a tumor suppressor. αPix is expressed only in highly proliferative and mobile cells, including immune cells, and up-regulated in certain invasive tumors, such as glioblastoma multiforme. Here, we showed that c-Cbl serves as an ubiquitin E3 ligase for proteasome-mediated degradation of αPix, but not ßPix. Remarkably, the rat C6 and human A172 glioma cells were unable to express c-Cbl, which leads to a dramatic accumulation of αPix. Depletion of αPix by shRNA markedly reduced the ability of the glioma cells to migrate and invade, whereas complementation of shRNA-insensitive αPix promoted it. These results indicate that c-Cbl negatively regulates αPix-mediated cell migration and invasion and the lack of c-Cbl in the C6 and A172 glioma cells is responsible for their malignant behavior.


Asunto(s)
Leucemia Mieloide/genética , Mutación , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Receptores ErbB/metabolismo , Prueba de Complementación Genética , Glioma/metabolismo , Células HEK293 , Humanos , Leucemia Mieloide/metabolismo , Invasividad Neoplásica , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba
9.
Nat Commun ; 11(1): 4931, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33004795

RESUMEN

Testis-restricted melanoma antigen (MAGE) proteins are frequently hijacked in cancer and play a critical role in tumorigenesis. MAGEs assemble with E3 ubiquitin ligases and function as substrate adaptors that direct the ubiquitination of novel targets, including key tumor suppressors. However, how MAGEs recognize their targets is unknown and has impeded the development of MAGE-directed therapeutics. Here, we report the structural basis for substrate recognition by MAGE ubiquitin ligases. Biochemical analysis of the degron motif recognized by MAGE-A11 and the crystal structure of MAGE-A11 bound to the PCF11 substrate uncovered a conserved substrate binding cleft (SBC) in MAGEs. Mutation of the SBC disrupted substrate recognition by MAGEs and blocked MAGE-A11 oncogenic activity. A chemical screen for inhibitors of MAGE-A11:substrate interaction identified 4-Aminoquinolines as potent inhibitors of MAGE-A11 that show selective cytotoxicity. These findings provide important insights into the large family of MAGE ubiquitin ligases and identify approaches for developing cancer-specific therapeutics.


Asunto(s)
Antígenos de Neoplasias/ultraestructura , Proteínas de Neoplasias/ultraestructura , Neoplasias/tratamiento farmacológico , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Secuencias de Aminoácidos , Aminoquinolinas/farmacología , Aminoquinolinas/uso terapéutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinogénesis/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Células HEK293 , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Mutagénesis , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , Prueba de Estudio Conceptual , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Dominios Proteicos/genética , Mapeo de Interacción de Proteínas , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/genética , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética
10.
Asian J Surg ; 42(10): 914-921, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30833157

RESUMEN

BACKGROUND/OBJECTIVE: The prognosis of hormone receptor-positive and HER2-negative breast cancer is better than that of other subtypes. Current guidelines recommend chemotherapy for N1 breast cancer patients. However, this has the possibility to be over-treatment. METHODS: This was a retrospective study of 18,549 patients who were surgically treated for invasive breast cancer, at a single center in South Korea, between January 1993 and December 2012. N1 stage breast cancer patients who were hormone receptor-positive and HER2-negative were enrolled, and propensity score matching was performed to compare patients treated with anti-hormonal therapy alone (N = 83) and those treated with chemotherapy followed by anti-hormonal therapy (N = 85). RESULTS: In survival analysis, the survival parameters of the endocrine therapy-only group and the chemotherapy with endocrine therapy group were respectively 96.1% and 94.0% for 5-year recurrence free survival (RFS), 89.6% and 94.0% for 10-year RFS, 97.4% and 94.0% for 5-year distant metastasis-free survival (DMFS), 93.2% and 94.0% for 10-year DMFS, 98.7% and 98.8% for 10-year breast cancer-specific survival (BCSS), and 98.7% and 98.8% for 10-year overall survival (OS). There were no significant differences in RFS (p = 0.871), DMFS (p = 0.491), BCSS (p = 0.569) and OS (p = 0.731) between the two groups. CONCLUSION: Several patients with clinicopathologic features like hormone receptor positivity and HER2 negativity can avoid chemotherapy even with lymph node metastasis. Future studies with a long-term follow-up and a larger number of patients are required for validating our results.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Quimioterapia Adyuvante , Metástasis Linfática , Receptor ErbB-2 , Adulto , Anciano , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Femenino , Humanos , Mastectomía , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Puntaje de Propensión , Estudios Retrospectivos , Tasa de Supervivencia , Factores de Tiempo , Resultado del Tratamiento
11.
Nat Commun ; 7: 12513, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27545325

RESUMEN

p53 plays a pivotal role in tumour suppression under stresses, such as DNA damage. ISG15 has been implicated in the control of tumorigenesis. Intriguingly, the expression of ISG15, UBE1L and UBCH8 is induced by DNA-damaging agents, such as ultraviolet and doxorubicin, which are known to induce p53. Here, we show that the genes encoding ISG15, UBE1L, UBCH8 and EFP, have the p53-responsive elements and their expression is induced in a p53-dependent fashion under DNA damage conditions. Furthermore, DNA damage induces ISG15 conjugation to p53 and this modification markedly enhances the binding of p53 to the promoters of its target genes (for example, CDKN1 and BAX) as well as of its own gene by promoting phosphorylation and acetylation, leading to suppression of cell growth and tumorigenesis. These findings establish a novel feedback circuit between p53 and ISG15-conjugating system for positive regulation of the tumour suppressive function of p53 under DNA damage conditions.


Asunto(s)
Carcinogénesis/patología , Citocinas/genética , Daño del ADN/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinas/genética , Acetilación , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinas/metabolismo , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Doxorrubicina/farmacología , Humanos , Fosforilación , Regiones Promotoras Genéticas/genética , Enzimas Activadoras de Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinas/metabolismo , Rayos Ultravioleta/efectos adversos , Proteína X Asociada a bcl-2/metabolismo
12.
Neoplasia ; 17(6): 518-24, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26152360

RESUMEN

c-Cbl, a RING-type ubiquitin E3 ligase, downregulates various receptor tyrosine kinases (e.g., epidermal growth factor receptor (EGFR)), leading to inhibition of cell proliferation. Moreover, patients with myeloid neoplasm frequently harbor c-Cbl mutations, implicating the role of c-Cbl as a tumor suppressor. Recently, we have shown that c-Cbl downregulates αPix-mediated cell migration and invasion, and the lack of c-Cbl in the rat C6 and human A172 glioma cells is responsible for their malignant behavior. Here, we showed that c-Cbl exon skipping occurs in the glioma cells and the brain tissues from glioblastoma patients lacking c-Cbl. This exon skipping resulted in generation of two types of c-Cbl isoforms: type I lacking exon-9 and type II lacking exon-9 and exon-10. However, the c-Cbl isoforms in the cells and tissues could not be detected as they were rapidly degraded by proteasome. Consequently, C6 and A172 cells showed sustained EGFR activation. However, no splice site mutation was found in the region from exon-7 to exon-11 of the c-Cbl gene in C6 cells and a glioblastoma tissue lacking c-Cbl. In addition, c-Cbl exon skipping could be induced when cells transfected with a c-Cbl mini-gene were grown to high density or under hypoxic stress. These results suggest that unknown alternations (e.g., mutation) of splicing machinery in C6 and A172 cells and the glioblastoma brain tissues are responsible for the deleterious exon skipping. Collectively, these findings indicate that the c-Cbl exon skipping contributes to human glioma and its malignant behavior.


Asunto(s)
Exones , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/patología , Hipoxia/fisiopatología , Proteínas Proto-Oncogénicas c-cbl/genética , Animales , Western Blotting , Receptores ErbB/genética , Humanos , Inmunoprecipitación , Mutación/genética , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Elementos Reguladores de la Transcripción , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
13.
Nat Commun ; 5: 5483, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25406032

RESUMEN

DBC1 is a major inhibitor of SIRT1, which plays critical roles in the control of diverse cellular processes, including stress response and energy metabolism. Therefore, the DBC1-SIRT1 interaction should finely be regulated. Here we report that DBC1 modification by Small Ubiquitin-like Modifier 2/3 (SUMO 2/3), but not by SUMO1, is crucial for p53 transactivation under genotoxic stress. Whereas etoposide treatment reduced the interaction of DBC1 with SENP1, it promoted that with PIAS3, resulting in an increase in DBC1 sumoylation. Remarkably, the switching from SENP1 to PIAS3 for DBC1 binding was achieved by ATM/ATR-mediated phosphorylation of DBC1. Furthermore, DBC1 sumoylation caused an increase in the DBC1-SIRT1 interaction, leading to the release of p53 from SIRT1 for transcriptional activation. Consistently, SENP1 knockdown promoted etoposide-induced apoptosis, whereas knockdown of PIAS3 or SUMO2/3 and overexpression of sumoylation-deficient DBC1 mutant inhibited it. These results establish the role of DBC1 sumoylation in the promotion of p53-mediated apoptosis in response to genotoxic stress.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Daño del ADN , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Humanos , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Sumoilación , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Ubiquitinas/genética
14.
J Gastric Cancer ; 13(4): 226-31, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24511418

RESUMEN

PURPOSE: The role of metastasectomy has been debatable and unclear in the treatment for patients with metastatic gastric cancer. Therefore, this study was designed to evaluate the role of metastasectomy on the overall survival of these patients. MATERIALS AND METHODS: In 2,406 patients who underwent gastrectomy for gastric cancer between 1998 and 2010, 188 (7.8%) patients had their first surgery for metastatic gastric cancer. To minimize the bias of systemic chemotherapy, 99 patients who received postoperative chemotherapy (fewer than 2 cycles) were excluded. The primary gastrectomy or metastasectomy had not been enforced in the following cases. Patients with far advanced peritoneal dissemination, multiple liver and lung metastasis (more than 2), and a poor general condition (Eastern Cooperative Oncology Group>2) were excluded. Based on the metastasectomy, the patients were classified into two groups, gastrectomy with metastasectomy and gastrectomy only group. RESULTS: There was no significant difference between both groups in clinicopathological characteristics except for the mean age (P=0.047). The univariate analysis for overall survival show statistical significances in metastasectomy (P=0.026), distal gastrectomy (P=0.047), and combined resection of another organ (P=0.047) group. With a multivariate analysis, metastasectomy was a significant factor in patient survival after surgery (odds ratio 1.679; P=0.034). CONCLUSIONS: Based on our results, we assume that a detailed strategy for surgery is needed to improve the overall survival of patients with metastatic gastric cancer. Therefore, we suggest that a metastasectomy can help prolong overall survival in some patients with metastatic gastric cancer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA