Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
BMC Genom Data ; 24(1): 56, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37749495

RESUMEN

BACKGROUND: Recently accumulated evidence indicates a potential association between COVID-19 and elevated susceptibility to cancer, including male genital cancer. However, the causal nature of this relationship remains unclear. METHODS: In this Mendelian randomization (MR) study, we investigated the potential causal relationship between COVID-19 and male genital cancer using genetic variants as instrumental variables. We utilized summary statistics from two large-scale genome-wide association studies of COVID-19 hospitalized Vs. controls, as well as data from a population-based male genital cancer database based on European ancestry. We applied stringent quality control measures to select instrumental variables, including checking for linkage disequilibrium, removing low-quality variants, and assessing the strength of the instruments using the F-statistic. We conducted the MR  analysis using the inverse-variance weighted method and several sensitivity analyses (including MR Egger and Weighted Median MR analysis) to test the robustness of our results. RESULTS: Our MR analysis revealed no causal associations between COVID-19 hospitalization and the incidence of male genital cancer. In the inverse-variance weighted analysis, no causal associations were observed between patients with COVID-19 hospitalization and the incidence of male genital cancer (odds ratio = 1.000 and 95% confidence interval = 0.998-1.001, p = 0.668). The estimated causal effect was consistent across all sensitivity analyses (including the Weighted Median, the MR Egger analysis, and the MR PROSSO analysis). The leave-one-out analysis showed that there was no any sing Single-nucleotide polymorphism significantly influencing our results. CONCLUSIONS: Our study provides evidence that there is no causal association between COVID-19 hospitalization and male genital cancer.


Asunto(s)
COVID-19 , Neoplasias de los Genitales Masculinos , Humanos , Masculino , Análisis de la Aleatorización Mendeliana , Estudio de Asociación del Genoma Completo , COVID-19/epidemiología , COVID-19/genética , Genitales Masculinos
2.
Eat Weight Disord ; 28(1): 11, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36790552

RESUMEN

OBJECTIVES: To determine the causal association between genetically predicted obesity and the risk of hip osteoarthritis. METHODS: We performed two-sample Mendelian randomization (MR) analysis to analyze the association between body mass index (BMI) and hip osteoarthritis using pooled-level genome-wide association study (GWAS) data. The inverse variance weighted (IVW), MR‒Egger, and weighted median methods were used to estimate the causal association. In addition, we applied the MR Steiger filtering method, MR robust adjusted profile score (MR.RAPS) methods, and the MR Pleiotropy RESidual Sum and Outlier (MR-PRESSO) global test to examine and address potential horizontal pleiotropy. RESULTS: We found a causal relationship between genetically predicted BMI and the risk of hip osteoarthritis by the IVW method [OR = 1.45, 95% confidence interval (CI) = 1.04-2.00, P = 0.02]. In the sensitivity analysis, the results of the MR‒Egger and weighted median methods revealed similar estimations but with a wide CI with lower precision. The funnel plot, MR-Egger intercept, and MR-PRESSO all indicated the absence of a directional pleiotropic effect. In addition, no heterogeneity was observed in the present analysis. Therefore, the result of IVW is most suitable and reliable for the present MR analysis. CONCLUSION: There is a causal relationship between obesity and a higher risk of hip osteoarthritis, suggesting that weight management may be an intervention for the prevention and management of hip osteoarthritis. LEVEL OF EVIDENCE: Bioinformatics, Basic science.


Asunto(s)
Estudio de Asociación del Genoma Completo , Osteoartritis de la Cadera , Humanos , Osteoartritis de la Cadera/genética , Índice de Masa Corporal , Nonoxinol , Obesidad/complicaciones , Obesidad/genética , Polimorfismo de Nucleótido Simple
3.
Front Oncol ; 12: 911466, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36237339

RESUMEN

Rationale: Fructose-1,6-bisphosphatase (FBP1) is a tumor suppressor and a key enzyme negatively regulating Warburg effect in cancer. However, regulation of FBP1 protein expression and its exact role in prostate cancer (PCa) is largely unclear. Phosphatase and tensin homolog (PTEN) is one of the most frequently deleted tumor suppressor genes in human PCa. However, the role of PTEN loss in aberrant Warburg effect in cancer remains poorly understood. Methods: Expression of PTEN and FBP1 was analyzed in several PCa cell lines and prostate tumor tissues in mice. Western blot (WB) and RT-PCR approaches were used to examine how PTEN regulates FBP1 expression. Co-immunoprecipitation (co-IP) and in vivo ubiquitination assays were used to define the regulatory mechanisms. A PCa xenograft model was employed to determine the impact of PTEN regulation of FBP1 on PCa growth in vivo. Result: We demonstrated that in a manner dependent of PI3K/AKT signal pathway PTEN regulated FBP1 expression in various PCa cell lines and tumors in mice. We confirmed that this regulation took place at the protein level and was mediated by SKP2 E3 ubiquitin ligase. Mechanistically, we showed that serine 271 phosphorylation of FBP1 by cyclin-dependent kinases (CDKs) was essential for SKP2-mediated degradation of FBP1 protein induced by PTEN loss. Most importantly, we further showed that loss of PTEN expression enhanced Warburg effect and PCa growth in mice in a manner dependent, at least partially on FBP1 protein degradation. Conclusions: Our results reveal a novel tumor-suppressive feature of PTEN in restraining FBP1 degradation and the Warburg effect. These results also suggest that prohibiting FBP1 protein degradation could be a viable therapeutic strategy for PTEN-deficient PCa.

4.
Nat Commun ; 12(1): 5716, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34588438

RESUMEN

Mutations in SPOP E3 ligase gene are reportedly associated with genome-wide DNA hypermethylation in prostate cancer (PCa) although the underlying mechanisms remain elusive. Here, we demonstrate that SPOP binds and promotes polyubiquitination and degradation of histone methyltransferase and DNMT interactor GLP. SPOP mutation induces stabilization of GLP and its partner protein G9a and aberrant upregulation of global DNA hypermethylation in cultured PCa cells and primary PCa specimens. Genome-wide DNA methylome analysis shows that a subset of tumor suppressor genes (TSGs) including FOXO3, GATA5, and NDRG1, are hypermethylated and downregulated in SPOP-mutated PCa cells. DNA methylation inhibitor 5-azacytidine effectively reverses expression of the TSGs examined, inhibits SPOP-mutated PCa cell growth in vitro and in mice, and enhances docetaxel anti-cancer efficacy. Our findings reveal the GLP/G9a-DNMT module as a mediator of DNA hypermethylation in SPOP-mutated PCa. They suggest that SPOP mutation could be a biomarker for effective treatment of PCa with DNA methylation inhibitor alone or in combination with taxane chemotherapeutics.


Asunto(s)
Metilación de ADN/genética , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas Nucleares/genética , Neoplasias de la Próstata/genética , Proteínas Represoras/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Azacitidina/farmacología , Azacitidina/uso terapéutico , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Docetaxel/farmacología , Docetaxel/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor , Humanos , Masculino , Ratones , Mutación , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteínas Represoras/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Adv Sci (Weinh) ; 8(20): e2102555, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34397171

RESUMEN

DNA-binding proteins, including transcription factors (TFs), play essential roles in various cellular processes and pathogenesis of diseases, deeming to be potential therapeutic targets. However, these proteins are generally considered undruggable as they lack an enzymatic catalytic site or a ligand-binding pocket. Proteolysis-targeting chimera (PROTAC) technology has been developed by engineering a bifunctional molecule chimera to bring a protein of interest (POI) to the proximity of an E3 ubiquitin ligase, thus inducing the ubiquitination of POI and further degradation through the proteasome pathway. Here, the development of oligonucleotide-based PROTAC (O'PROTACs), a class of noncanonical PROTACs in which a TF-recognizing double-stranded oligonucleotide is incorporated as a binding moiety of POI is reported. It is demonstrated that O'PROTACs of lymphoid enhancer-binding factor 1 (LEF1) and ETS-related gene (ERG), two highly cancer-related transcription factors, successfully promote degradation of these proteins, impede their transcriptional activity, and inhibit cancer cell growth in vitro and in vivo. The programmable nature of O'PROTACs indicates that this approach is also applicable to destruct other TFs. O'PROTACs not only can serve as a research tool but also can be harnessed as a therapeutic arsenal to target DNA binding proteins for effective treatment of diseases such as cancer.


Asunto(s)
Proteínas de Unión al ADN/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Neoplasias/genética , Oligonucleótidos/genética , Humanos , Neoplasias/terapia , Oligonucleótidos/farmacología , Complejo de la Endopetidasa Proteasomal/genética , Proteolisis/efectos de los fármacos , Regulador Transcripcional ERG/genética , Ubiquitina-Proteína Ligasas/genética
6.
Sci Adv ; 7(25)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34144977

RESUMEN

53BP1 activates nonhomologous end joining (NHEJ) and inhibits homologous recombination (HR) repair of DNA double-strand breaks (DSBs). Dissociation of 53BP1 from DSBs and consequent activation of HR, a less error-prone pathway than NHEJ, helps maintain genome integrity during DNA replication; however, the underlying mechanisms are not fully understood. Here, we demonstrate that E3 ubiquitin ligase SPOP promotes HR during S phase of the cell cycle by excluding 53BP1 from DSBs. In response to DNA damage, ATM kinase-catalyzed phosphorylation of SPOP causes a conformational change in SPOP, revealed by x-ray crystal structures, that stabilizes its interaction with 53BP1. 53BP1-bound SPOP induces polyubiquitination of 53BP1, eliciting 53BP1 extraction from chromatin by a valosin-containing protein/p97 segregase complex. Our work shows that SPOP facilitates HR repair over NHEJ during DNA replication by contributing to 53BP1 removal from chromatin. Cancer-derived SPOP mutations block SPOP interaction with 53BP1, inducing HR defects and chromosomal instability.


Asunto(s)
Cromatina , Roturas del ADN de Doble Cadena , Cromatina/genética , Reparación del ADN por Unión de Extremidades , Replicación del ADN , Proteínas Nucleares , Reparación del ADN por Recombinación , Proteínas Represoras , Proteína 1 de Unión al Supresor Tumoral P53
7.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33593912

RESUMEN

Both gene repressor (Polycomb-dependent) and activator (Polycomb-independent) functions of the Polycomb protein enhancer of zeste homolog 2 (EZH2) are implicated in cancer progression. EZH2 protein can be phosphorylated at various residues, such as threonine 487 (T487), by CDK1 kinase, and such phosphorylation acts as a Polycomb repressive complex 2 (PRC2) suppression "code" to mediate the gene repressor-to-activator switch of EZH2 functions. Here we demonstrate that the histone reader protein ZMYND8 is overexpressed in human clear cell renal cell carcinoma (ccRCC). ZMYND8 binds to EZH2, and their interaction is largely enhanced by CDK1 phosphorylation of EZH2 at T487. ZMYND8 depletion not only enhances Polycomb-dependent function of EZH2 in hypoxia-exposed breast cancer cells or von Hippel-Lindau (VHL)-deficient ccRCC cells, but also suppresses the FOXM1 transcription program. We further show that ZMYND8 is required for EZH2-FOXM1 interaction and is important for FOXM1-dependent matrix metalloproteinase (MMP) gene expression and EZH2-mediated migration and invasion of VHL-deficient ccRCC cells. Our results identify a previously uncharacterized role of the chromatin reader ZMYND8 in recognizing the PRC2-inhibitory phosphorylation "code" essential for the Polycomb-dependent to -independent switch of EZH2 functions. They also reveal an oncogenic pathway driving cell migration and invasion in hypoxia-inducible factor-activated (hypoxia or VHL-deficient) cancer.


Asunto(s)
Carcinoma de Células Renales/patología , Movimiento Celular , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Hipoxia/fisiopatología , Neoplasias Renales/patología , Complejo Represivo Polycomb 2/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Proliferación Celular , Cromatina/genética , Cromatina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Fosforilación , Complejo Represivo Polycomb 2/genética , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética
8.
Cell Rep ; 34(7): 108744, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33596421

RESUMEN

Acquisition of resistance to phosphatidylinositol 3-kinase (PI3K)/AKT-targeted monotherapy implies the existence of common resistance mechanisms independent of cancer type. Here, we demonstrate that PI3K/AKT inhibitors cause glycolytic crisis, acetyl-coenzyme A (CoA) shortage, and a global decrease in histone acetylation. In addition, PI3K/AKT inhibitors induce drug resistance by selectively augmenting histone H3 lysine 27 acetylation (H3K27ac) and binding of CBP/p300 and BRD4 proteins at a subset of growth factor and receptor (GF/R) gene loci. BRD4 occupation at these loci and drug-resistant cell growth are vulnerable to both bromodomain and histone deacetylase (HDAC) inhibitors. Little or no occupation of HDAC proteins at the GF/R gene loci underscores the paradox that cells respond equivalently to the two classes of inhibitors with opposite modes of action. Targeting this unique acetyl-histone-related vulnerability offers two clinically viable strategies to overcome PI3K/AKT inhibitor resistance in different cancers.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Neoplasias/tratamiento farmacológico , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Células HCT116 , Humanos , Masculino , Ratones , Ratones SCID , Neoplasias/enzimología , Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Cell ; 79(6): 1008-1023.e4, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32871104

RESUMEN

TMPRSS2-ERG gene fusion occurs in approximately 50% of cases of prostate cancer (PCa), and the fusion product is a key driver of prostate oncogenesis. However, how to leverage cellular signaling to ablate TMPRSS2-ERG oncoprotein for PCa treatment remains elusive. Here, we demonstrate that DNA damage induces proteasomal degradation of wild-type ERG and TMPRSS2-ERG oncoprotein through ERG threonine-187 and tyrosine-190 phosphorylation mediated by GSK3ß and WEE1, respectively. The dual phosphorylation triggers ERG recognition and degradation by the E3 ubiquitin ligase FBW7 in a manner independent of a canonical degron. DNA damage-induced TMPRSS2-ERG degradation was abolished by cancer-associated PTEN deletion or GSK3ß inactivation. Blockade of DNA damage-induced TMPRSS2-ERG oncoprotein degradation causes chemotherapy-resistant growth of fusion-positive PCa cells in culture and in mice. Our findings uncover a previously unrecognized TMPRSS2-ERG protein destruction mechanism and demonstrate that intact PTEN and GSK3ß signaling are essential for effective targeting of ERG protein by genotoxic therapeutics in fusion-positive PCa.


Asunto(s)
Proteínas de Ciclo Celular/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Proteínas de Fusión Oncogénica/genética , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/genética , Proteínas Tirosina Quinasas/genética , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Quimioterapia , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Xenoinjertos , Humanos , Masculino , Ratones , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
Gut Microbes ; 11(6): 1567-1589, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32588736

RESUMEN

Helicobacter pylori (H. pylori) infection is the strongest known risk factor for the development of gastric cancer. DNA damage response (DDR) and autophagy play key roles in tumorigenic transformation. However, it remains unclear how H. pylori modulate DDR and autophagy in gastric carcinogenesis. Here we report that H. pylori infection promotes DNA damage via suppression of Rad51 expression through inhibition of autophagy and accumulation of p62 in gastric carcinogenesis. We find that H. pylori activated DNA damage pathway in concert with downregulation of repair protein Rad51 in gastric cells, C57BL/6 mice and Mongolian gerbils. In addition, autophagy was increased early and then decreased gradually during the duration of H. pylori infection in vitro in a CagA-dependent manner. Moreover, loss of autophagy led to promotion of DNA damage in H. pylori-infected cells. Furthermore, knockdown of autophagic substrate p62 upregulated Rad51 expression, and p62 promoted Rad51 ubiquitination via the direct interaction of its UBA domain. Finally, H. pylori infection was associated with elevated levels of p62 in gastric intestinal metaplasia and decreased levels of Rad51 in dysplasia compared to their H. pylori- counterparts. Our findings provide a novel mechanism into the linkage of H. pylori infection, autophagy, DNA damage and gastric tumorigenesis.


Asunto(s)
Autofagia , Daño del ADN , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/fisiología , Recombinasa Rad51/metabolismo , Neoplasias Gástricas/metabolismo , Adulto , Anciano , Animales , Carcinogénesis , Femenino , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/fisiopatología , Helicobacter pylori/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Recombinasa Rad51/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/fisiopatología , Ubiquitinación
11.
EMBO Mol Med ; 11(11): e10659, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31559706

RESUMEN

CULLIN3-based E3 ubiquitin ligase substrate-binding adaptor gene SPOP is frequently mutated in prostate cancer (PCa). PCa harboring SPOP hotspot mutants (e.g., F133V) are resistant to BET inhibitors because of aberrant elevation of BET proteins. Here, we identified a previously unrecognized mutation Q165P at the edge of SPOP MATH domain in primary and metastatic PCa of a patient. The Q165P mutation causes structural changes in the MATH domain and impairs SPOP dimerization and substrate degradation. Different from F133V hotspot mutant tumors, Q165P mutant patient-derived xenografts (PDXs) and organoids were modestly sensitive to the BET inhibitor JQ1. Accordingly, protein levels of AR, BRD4 and downstream effectors such as RAC1 and phosphorylated AKT were not robustly elevated in Q165P mutant cells as in F133V mutant cells. However, NEO2734, a novel dual inhibitor of BET and CBP/p300, is active in both hotspot mutant (F133V) and non-hotspot mutant (Q165P) PCa cells in vitro and in vivo. These data provide a strong rationale to clinically investigate the anti-cancer efficacy of NEO2734 in SPOP-mutated PCa patients.


Asunto(s)
Antineoplásicos/farmacología , Proteína p300 Asociada a E1A/metabolismo , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/metabolismo , Neoplasias de la Próstata/patología , Proteínas Represoras/metabolismo , Sialoglicoproteínas/metabolismo , Factores de Transcripción/metabolismo , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Modelos Animales de Enfermedad , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Xenoinjertos , Humanos , Masculino , Proteínas Mutantes/genética , Mutación Missense , Trasplante de Neoplasias , Proteínas Nucleares/genética , Fragmentos de Péptidos/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Represoras/genética , Sialoglicoproteínas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores
12.
EMBO J ; 38(5)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30723117

RESUMEN

In light of the increasing number of identified cancer-driven gain-of-function (GOF) mutants of p53, it is important to define a common mechanism to systematically target several mutants, rather than developing strategies tailored to inhibit each mutant individually. Here, using RNA immunoprecipitation-sequencing (RIP-seq), we identified the Polycomb-group histone methyltransferase EZH2 as a p53 mRNA-binding protein. EZH2 bound to an internal ribosome entry site (IRES) in the 5'UTR of p53 mRNA and enhanced p53 protein translation in a methyltransferase-independent manner. EZH2 augmented p53 GOF mutant-mediated cancer growth and metastasis by increasing protein levels of mutant p53. EZH2 overexpression was associated with worsened outcome selectively in patients with p53-mutated cancer. Depletion of EZH2 by antisense oligonucleotides inhibited p53 GOF mutant-mediated cancer growth. Our findings reveal a non-methyltransferase function of EZH2 that controls protein translation of p53 GOF mutants, inhibition of which causes synthetic lethality in cancer cells expressing p53 GOF mutants.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Mutación con Ganancia de Función , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/patología , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2/genética , Humanos , Sitios Internos de Entrada al Ribosoma , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Estabilidad Proteica , ARN Mensajero/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
BMC Anesthesiol ; 18(1): 138, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30285627

RESUMEN

BACKGROUND: The goal of this study was to evaluate the accuracy and interchangeability between continuous cardiac output (CO) measured by electrical velocimetry (COEv) and continuous cardiac output obtained using the pulmonary thermodilution method (COPAC) during living donor liver transplantation (LDLT). METHOD: Twenty-three patients were enrolled in this prospective observational study. CO was recorded by both two methods and compared at nine specific time points. The data were analyzed using correlation coefficients, Bland-Altman analysis for the percentage errors, and the concordance rate for trend analysis using a four-quadrant plot. RESULTS: In total, 207 paired datasets were recorded during LDLT. CO data were in the range of 2.8-12.7 L/min measured by PAC and 3.4-14.9 L/min derived from the EV machine. The correction coefficient between COPAC and COEv was 0.415 with p < 0.01. The 95% limitation agreement was - 5.9 to 3.4 L/min and the percentage error was 60%. The concordance rate was 56.5%. CONCLUSIONS: The Aesculon™ monitor is not yet interchangeable with continuous thermodilution CO monitoring during LDLT. TRIAL REGISTRATION: The study was approved by the Institutional Review Board of Chang Gung Medical Foundation in Taiwan (registration number: 201600264B0 ).


Asunto(s)
Gasto Cardíaco/fisiología , Trasplante de Hígado/métodos , Pulmón/fisiología , Monitoreo Intraoperatorio/métodos , Reología/métodos , Adulto , Anciano , Femenino , Humanos , Trasplante de Hígado/efectos adversos , Trasplante de Hígado/normas , Masculino , Persona de Mediana Edad , Monitoreo Intraoperatorio/normas , Estudios Prospectivos , Reología/normas , Termodilución/métodos , Termodilución/normas
14.
Mol Cell ; 71(4): 592-605.e4, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-30057199

RESUMEN

The bromodomain and extra-terminal domain (BET) protein BRD4 is emerging as a promising anticancer therapeutic target. However, resistance to BET inhibitors often occurs, and it has been linked to aberrant degradation of BRD4 protein in cancer. Here, we demonstrate that the deubiquitinase DUB3 binds to BRD4 and promotes its deubiquitination and stabilization. Expression of DUB3 is transcriptionally repressed by the NCOR2-HDAC10 complex. The NCOR2 gene is frequently deleted in castration-resistant prostate cancer patient specimens, and loss of NCOR2 induces elevation of DUB3 and BRD4 proteins in cancer cells. DUB3-proficient prostate cancer cells are resistant to the BET inhibitor JQ1 in vitro and in mice, but this effect is diminished by DUB3 inhibitory agents such as CDK4/6 inhibitor in a RB-independent manner. Our findings identify a previously unrecognized mechanism causing BRD4 upregulation and drug resistance, suggesting that DUB3 is a viable therapeutic target to overcome BET inhibitor resistance in cancer.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/genética , Endopeptidasas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Factores de Transcripción/genética , Animales , Antineoplásicos/farmacología , Azepinas/farmacología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Endopeptidasas/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Masculino , Ratones , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Co-Represor 2 de Receptor Nuclear/deficiencia , Co-Represor 2 de Receptor Nuclear/genética , Piperazinas/farmacología , Próstata/efectos de los fármacos , Próstata/enzimología , Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata Resistentes a la Castración/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteolisis , Piridinas/farmacología , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Transcripción Genética , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
EMBO Mol Med ; 10(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29523594

RESUMEN

AKT-mTOR and androgen receptor (AR) signaling pathways are aberrantly activated in prostate cancer due to frequent PTEN deletions or SPOP mutations. A clinical barrier is that targeting one of them often activates the other. Here, we demonstrate that HDAC3 augments AKT phosphorylation in prostate cancer cells and its overexpression correlates with AKT phosphorylation in patient samples. HDAC3 facilitates lysine-63-chain polyubiquitination and phosphorylation of AKT, and this effect is mediated by AKT deacetylation at lysine 14 and 20 residues and HDAC3 interaction with the scaffold protein APPL1. Conditional homozygous deletion of Hdac3 suppresses prostate tumorigenesis and progression by concomitant blockade of AKT and AR signaling in the Pten knockout mouse model. Pharmacological inhibition of HDAC3 using a selective HDAC3 inhibitor RGFP966 inhibits growth of both PTEN-deficient and SPOP-mutated prostate cancer cells in culture, patient-derived organoids and xenografts in mice. Our study identifies HDAC3 as a common upstream activator of AKT and AR signaling and reveals that dual inhibition of AKT and AR pathways is achievable by single-agent targeting of HDAC3 in prostate cancer.


Asunto(s)
Proteínas Nucleares/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Acrilamidas/farmacología , Animales , Western Blotting , Genotipo , Células HEK293 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Inmunoprecipitación , Masculino , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Fosfohidrolasa PTEN/genética , Fenilendiaminas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Receptores Androgénicos/genética , Proteínas Represoras/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Complejos de Ubiquitina-Proteína Ligasa
16.
Oxid Med Cell Longev ; 2018: 8938207, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30671176

RESUMEN

Aging is a major factor that contributes to neurological impairment and neuropathological changes, such as inflammation, oxidative stress, neuronal apoptosis, and synaptic dysfunction. Flavonoids act as protective antioxidant and anti-inflammatory agents against various age-related neurodegenerative diseases. Here, we investigated the protective effect and mechanisms of the flavonoid-rich ethanol extract from the leaves of Diospyros kaki (FELDK) in the cortex and hippocampus of D-galactose- (gal-) aged mice. Our results showed that FELDK treatment restored memory impairment in mice as determined by the Y-maze and Morris water maze tests. FELDK decreased oxidative stress levels via inhibiting reactive oxygen species (ROS) and malondialdehyde (MDA) production and elevating antioxidative enzymes. FELDK also alleviated D-gal-induced neuroinflammation via suppressing the expression of advanced glycation end products (AGEs) and receptor for AGEs (RAGE) and activating microgliosis and astrocytosis, nuclear factor kappa B (NF-κB) nuclear translocation, and downstream inflammatory mediators. Moreover, FELDK inhibited the phosphatidylinositol 3-kinase (PI3K)/Akt and C-jun N-terminal kinase (JNK) apoptotic signaling pathways and ameliorated the impairment of synapse-related proteins. Hence, these results indicate that FELDK exerts neuroprotective effects on D-gal-induced brain aging. Thus, FELDK may be a potential therapeutic strategy for preventing and treating age-related neurodegenerative diseases such as Alzheimer's disease.


Asunto(s)
Envejecimiento/metabolismo , Diospyros/química , Flavonoides/farmacología , Galactosa/efectos adversos , Hipocampo/metabolismo , Enfermedades Neurodegenerativas , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Hojas de la Planta/química , Envejecimiento/patología , Animales , Etanol/química , Flavonoides/química , Galactosa/farmacología , Hipocampo/patología , Masculino , Ratones , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología , Extractos Vegetales/química
17.
Cancer Res ; 77(23): 6524-6537, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28986382

RESUMEN

E26 transformation-specific transcription factor ERG is aberrantly overexpressed in approximately 50% of all human prostate cancer due to TMPRSS2-ERG gene rearrangements. However, mice with prostate-specific transgenic expression of prostate cancer-associated ERG alone fail to develop prostate cancer, highlighting that ERG requires other lesions to drive prostate tumorigenesis. Forkhead box (FOXO) transcription factor FOXO1 is a tumor suppressor that is frequently inactivated in human prostate cancer. Here, we demonstrate that FOXO1, but not other FOXO proteins (FOXO3 and FOXO4), binds and inhibits the transcriptional activity of prostate cancer-associated ERG independently of FOXO1 transcriptional activity. Knockdown of endogenous FOXO1 increased invasion of TMPRSS2-ERG fusion-positive VCaP cells, an effect completely abolished by ERG knockdown. Patient specimen analysis demonstrated that FOXO1 and ERG protein expression inversely correlated in a subset of human prostate cancer. Although human ERG transgene expression or homozygous deletion of Foxo1 alone in the mouse prostate failed to promote tumorigenesis, concomitant ERG transgene expression and Foxo1 deletion resulted in upregulation of ERG target genes, increased cell proliferation, and formation of high-grade prostatic intraepithelial neoplasia. Overall, we provide biochemical and genetic evidence that aberrantly activated ERG cooperates with FOXO1 deficiency to promote prostate tumorigenesis and cell invasion. Our findings enhance understanding of prostate cancer etiology and suggest that the FOXO1-ERG signaling axis can be a potential target for treatment of prostate cancer. Cancer Res; 77(23); 6524-37. ©2017 AACR.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteína Forkhead Box O1/genética , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/genética , Serina Endopeptidasas/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Invasividad Neoplásica/genética , Próstata/patología , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/patología , Interferencia de ARN , ARN Interferente Pequeño/genética , Serina Endopeptidasas/biosíntesis , Transcripción Genética/genética , Regulador Transcripcional ERG/biosíntesis , Regulador Transcripcional ERG/genética
18.
Nat Med ; 23(9): 1055-1062, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28805822

RESUMEN

Bromodomain and extraterminal domain (BET) protein inhibitors are emerging as promising anticancer therapies. The gene encoding the E3 ubiquitin ligase substrate-binding adaptor speckle-type POZ protein (SPOP) is the most frequently mutated in primary prostate cancer. Here we demonstrate that wild-type SPOP binds to and induces ubiquitination and proteasomal degradation of BET proteins (BRD2, BRD3 and BRD4) by recognizing a degron motif common among them. In contrast, prostate cancer-associated SPOP mutants show impaired binding to BET proteins, resulting in decreased proteasomal degradation and accumulation of these proteins in prostate cancer cell lines and patient specimens and causing resistance to BET inhibitors. Transcriptome and BRD4 cistrome analyses reveal enhanced expression of the GTPase RAC1 and cholesterol-biosynthesis-associated genes together with activation of AKT-mTORC1 signaling as a consequence of BRD4 stabilization. Our data show that resistance to BET inhibitors in SPOP-mutant prostate cancer can be overcome by combination with AKT inhibitors and further support the evaluation of SPOP mutations as biomarkers to guide BET-inhibitor-oriented therapy in patients with prostate cancer.


Asunto(s)
Azepinas/farmacología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Proteínas Nucleares/genética , Neoplasias de la Próstata/genética , Proteínas Represoras/genética , Triazoles/farmacología , Proteína de Unión al GTP rac1/genética , Azepinas/uso terapéutico , Western Blotting , Proteínas de Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular , Perfilación de la Expresión Génica , Humanos , Inmunoprecipitación , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Terapia Molecular Dirigida , Complejos Multiproteicos/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Mutación , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Triazoles/uso terapéutico , Proteína de Unión al GTP rac1/metabolismo
19.
J Biol Chem ; 292(15): 6269-6280, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28242758

RESUMEN

Pancreatic cancer is one of the most lethal cancer types. Enhancer of zeste homolog 2 (EZH2) is an oncogenic protein overexpressed in pancreatic cancer, and EZH2 could be a potential therapeutic target for the treatment of pancreatic cancer. Although significant progress has been made toward understanding the function and deregulation of EZH2 in cancer cells, the posttranslational regulation of EZH2 in cancer cells is still unclear. F-box and WD repeat domain-containing 7 (FBW7) acts as a tumor suppressor by targeting multiple oncoprotein substrates for ubiquitination and degradation. Here we demonstrate that EZH2 is a bona fide substrate of FBW7 in pancreatic cancer cells. We provide evidence that the activated CDK5 kinase is involved in the EZH2 phosphorylation that is required for FBW7-mediated degradation. We further show that FBW7 suppresses EZH2 activity and inhibits tumor migration and invasion via degradation of EZH2 in pancreatic cancer cells. Furthermore, immunohistochemistry analysis revealed that expression of EZH2 protein negatively correlates with FBW7 protein levels in a cohort of human pancreatic cancer specimens. Collectively, our findings demonstrate that FBW7 is a novel E3 ligase of EZH2 that regulates the EZH2 protein level in pancreatic cancer and represents a viable strategy for effective treatment of pancreatic cancer.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Movimiento Celular , Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteínas F-Box/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Quinasa 5 Dependiente de la Ciclina/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Humanos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Ubiquitina-Proteína Ligasas/genética
20.
J Exp Clin Cancer Res ; 36(1): 23, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28148279

RESUMEN

BACKGROUND: Recent evidence suggests that the aberrant activation of Hedgehog (Hh) signaling by Gli transcription factors is characteristic of a variety of aggressive human carcinomas, including colorectal cancer (CRC). Forkhead box M1 (FoxM1) controls the expression of a number of cell cycle regulatory proteins, and FoxM1 expression is elevated in a broad range of human malignancies, which suggests that it plays a crucial role in tumorigenesis. However, the mechanisms underlying FoxM1 expression are not fully understood. Here, we aim to further investigate the molecular mechanism by which Gli1 regulates FoxM1 in CRC. METHODS: Western blotting and immunohistochemistry (IHC) were used to evaluate FoxM1 and Gli1 protein expression, respectively, in CRC tissues and matched adjacent normal mucosa. BrdU (5-bromo-2'-deoxyuridine) and clone formation assays were used to clarify the influence of FoxM1 on CRC cell growth and proliferation. Chromatin immunoprecipitation (ChIP) and luciferase experiments were performed to explore the potential mechanisms by which Gli1 regulates FoxM1. Additionally, the protein and mRNA expression levels of Gli1 and FoxM1 in six CRC cell lines were measured using Western blotting and real-time PCR. Finally, the effect of Hh signaling on the expression of FoxM1 was studied in cell biology experiments, and the effects of Hh signaling activation and FoxM1 inhibition on the distribution of CRC cells among cell cycle phases was assessed by flow cytometry. RESULTS: Gli1 and FoxM1 were abnormally elevated in human CRC tissues compared with matched adjacent normal mucosa samples, and FoxM1 is a downstream target gene of the transcription factor Gli1 in CRC and promoted CRC cell growth and proliferation. Moreover, the aberrant activation of Hh signaling promoted CRC cell proliferation by directly binding to the promoter of FoxM1 and transactivating the activity of FoxM1 in CRC cells. CONCLUSION: The dysregulation of the Hh-Gli1-FoxM1 axis is essential for the proliferation and growth of human CRC cells and offers a potent target for therapeutic intervention in CRC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo , Células CACO-2 , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Proteínas Hedgehog/metabolismo , Humanos , Regiones Promotoras Genéticas , Transducción de Señal , Activación Transcripcional , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA