Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Commun Signal ; 20(1): 40, 2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35346238

RESUMEN

BACKGROUND: Tumor cells tend to utilize glycolysis rather than aerobic respiration even under aerobic conditions. OVOL2, an inhibitory C2H2 zinc finger transcription factor, is a potential tumor suppressor in cancers. However, the association between OVOL2 and tumor energy metabolism is unknown. METHODS: Western blotting was used to determine the expression of OVOL2 in different non-small cell lung cancer (NSCLC) cell lines and mouse models. The metabolic parameters in NSCLC cells following overexpression or knockdown OVOL2 were examined. To define the mechanism by which OVOL2 regulates aerobic glycolysis, interacting protein of OVOl2 and downstream molecular events were identified by luciferase assay and co-immunoprecipitation. We documented the regulatory mechanism in mouse xenograft models. Finally, clinical relevance of OVOL2, NF-κB signaling and GLUT1 was measured by immunostaining. RESULTS: OVOL2 is downregulated in NSCLC and overexpression of OVOL2 inhibits the survival of cancer cells. Moreover, OVOL2 directly binds to P65 and inhibits the recruitment of P300 but facilitates the binding of HDAC1 to P65, which in turn negatively regulates NF-κB signaling to suppress GLUT1 translocation and glucose import. In contrast, OVOL2 expression is negatively regulated by NF-κB signaling in NSCLC cells via the ubiquitin-proteasome pathway. Re-expression of OVOL2 significantly compromise NF-κB signaling-induced GLUT1 translocation, aerobic glycolysis in NSCLC cells and mouse models. Immunostaining revealed inverse correlations between the OVOL2 and phosphorylated P65 levels and between the OVOL2 and membrane GLUT1 levels, and a strong correlation between the phosphorylated P65 and membrane GLUT1 levels. CONCLUSIONS: These results suggest a regulatory circuit linking NF-κB and OVOL2, which highlights the role of NF-κB signaling and OVOL2 in the modulation of glucose metabolism in NSCLC. Video Abstract.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , FN-kappa B , Factores de Transcripción , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Supervivencia Celular , Glucosa/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , FN-kappa B/metabolismo , Factores de Transcripción/metabolismo
2.
Mol Cancer ; 17(1): 84, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29690888

RESUMEN

Conventional tumor markers for non-invasive diagnosis of gastric cancer (GC) exhibit insufficient sensitivity and specificity to facilitate detection of early gastric cancer (EGC). We aimed to identify EGC-specific exosomal lncRNA biomarkers that are highly sensitive and stable for the non-invasive diagnosis of EGC. Hence, in the present study, exosomes from the plasma of five healthy individuals and ten stage I GC patients and from culture media of four human primary stomach epithelial cells and four gastric cancer cells (GCCs) were isolated. Exosomal RNA profiling was performed using RNA sequencing to identify EGC-specific exosomal lncRNAs. A total of 79 and 285 exosomal RNAs were expressed at significantly higher levels in stage I GC patients and GCCs, respectively, than that in normal controls. Through combinational analysis of the RNA sequencing results, we found two EGC-specific exosomal lncRNAs, lncUEGC1 and lncUEGC2, which were further confirmed to be remarkably up-regulated in exosomes derived from EGC patients and GCCs. Furthermore, stability testing demonstrates that almost all the plasma lncUEGC1 was encapsulated within exosomes and thus protected from RNase degradation. The diagnostic accuracy of exosomal lncUEGC1 was evaluated, and lncUEGC1 exhibited AUC values of 0.8760 and 0.8406 in discriminating EGC patients from healthy individuals and those with premalignant chronic atrophic gastritis, respectively, which was higher than the diagnostic accuracy of carcinoembryonic antigen. Consequently, exosomal lncUEGC1 may be promising in the development of highly sensitive, stable, and non-invasive biomarkers for EGC diagnosis.


Asunto(s)
Biomarcadores de Tumor/sangre , Exosomas/genética , ARN Largo no Codificante/genética , Neoplasias Gástricas/patología , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Detección Precoz del Cáncer , Femenino , Humanos , Masculino , Estadificación de Neoplasias , ARN Largo no Codificante/sangre , Neoplasias Gástricas/sangre , Neoplasias Gástricas/genética
3.
Hepatology ; 65(4): 1206-1221, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27809333

RESUMEN

Great progress has been achieved in the study of Hippo signaling in regulating tumorigenesis; however, the downstream molecular events that mediate this process have not been completely defined. Moreover, regulation of Hippo signaling during tumorigenesis in hepatocellular carcinoma (HCC) remains largely unknown. In the present study, we systematically investigated the relationship between Yes-associated protein/TEA domain family member (YAP-TEAD) and hepatocyte nuclear factor 4-alpha (HNF4α) in the hepatocarcinogenesis of HCC cells. Our results indicated that HNF4α expression was negatively regulated by YAP1 in HCC cells by a ubiquitin proteasome pathway. By contrast, HNF4α was found to directly associate with TEAD4 to compete with YAP1 for binding to TEAD4, thus inhibiting the transcriptional activity of YAP-TEAD and expression of their target genes. Moreover, overexpression of HNF4α was found to significantly compromise YAP-TEAD-induced HCC cell proliferation and stem cell expansion. Finally, we documented the regulatory mechanism between YAP-TEAD and HNF4α in rat and mouse tumor models, which confirmed our in vitro results. CONCLUSION: There is a double-negative feedback mechanism that controls TEAD-YAP and HNF4α expression in vitro and in vivo, thereby regulating cellular proliferation and differentiation. Given that YAP acts as a dominant oncogene in HCC and plays a crucial role in stem cell homeostasis and tissue regeneration, manipulating the interaction between YAP, TEADs, and HNF4α may provide a new approach for HCC treatment and regenerative medicine. (Hepatology 2017;65:1206-1221).


Asunto(s)
Carcinoma Hepatocelular/genética , Factor Nuclear 4 del Hepatocito/genética , Neoplasias Hepáticas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Biopsia con Aguja , Carcinogénesis/genética , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Inmunohistoquímica , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/genética , Distribución Aleatoria , Ratas , Ratas Wistar , Sensibilidad y Especificidad , Transducción de Señal , Factores de Transcripción de Dominio TEA , Factores de Transcripción/genética , Proteínas Señalizadoras YAP
4.
Biochim Biophys Acta Gen Subj ; 1862(4): 1017-1030, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29369785

RESUMEN

BACKGROUND: Kinase inhibitor sorafenib is the most widely used drug for advanced HCC clinical treatment nowadays. However, sorafenib administration is only effective for a small portion of HCC patients, and the majority develop sorafenib-resistance during treatment. Thus, it is urgent to discover the endogenous mechanism and identify new pharmaceutical targets of sorafenib-resistance. METHODS: Pregnane X receptor (PXR) was detected by immunohistochemistry and quantitative PCR. GST-pull down and LC-MS/MS was used to detect the interaction of PXR and Sorafenib. To test the properties of HCC tumor growth and metastasis, in vivo tumor explant model, FACS, trans-well assay, cell-survival inhibitory assay and Western blot were performed. In terms of mechanistic study, additional assays such as ChIP and luciferase reporter gene assay were applied. RESULTS: In the present work, we found high PXR level in clinical specimens is related to the poor prognosis of Sorafenib treated patients. By the mechanistic studies, we show that sorafenib binds to PXR and activates PXR pathway, and by which HCC cells develop sorafenib-resistance via activating. Moreover, PXR overexpression helps HCC cells to persist to sorafenib treatment. CONCLUSION: This study reports the endogenous sorafenib-resistance mechanism in HCC cells, which offers an opportunity to design new therapeutic approaches for HCC treatment. GENERAL SIGNIFICANCE: PXR mediates sorafenib-resistance in HCC cells and targeting PXR can be a useful approach to facilitate HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Receptores de Esteroides/genética , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones SCID , Niacinamida/metabolismo , Niacinamida/uso terapéutico , Compuestos de Fenilurea/metabolismo , Receptor X de Pregnano , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Interferencia de ARN , Receptores de Esteroides/metabolismo , Sorafenib , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Gastroenterology ; 150(3): 659-671.e16, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26619963

RESUMEN

BACKGROUND & AIMS: Activation of WNT signaling promotes the invasive activities of several types of cancer cells, but it is not clear if it regulates the same processes in colorectal cancer (CRC) cells, or what mechanisms are involved. We studied the expression and function of OVOL2, a member of the Ovo family of conserved zinc-finger transcription factors regulated by the WNT signaling pathway, in intestinal tumors of mice and human beings. METHODS: We analyzed the expression of OVOL2 protein and messenger RNA in CRC cell lines and tissue arrays, as well as CRC samples from patients who underwent surgery at Xiamen University in China from 2009 to 2012; clinical information also was collected. CRC cell lines (SW620) were infected with lentivirus expressing OVOL2, analyzed in migration and invasion assays, and injected into nude mice to assess tumor growth and metastasis. Tandem affinity purification was used to purify the OVOL2-containing complex from CRC cells; the complex was analyzed by liquid chromatography, tandem mass spectrometry, and immunoprecipitation experiments. Gene promoter activities were measured in luciferase reporter assays. We analyzed mice with an intestine-specific disruption of Ovol2 (Ovol2(flox/+) transgenic mice), as well as Apc(min/+) mice; these mice were crossed and analyzed. RESULTS: Analysis of data from patients indicated that the levels of OVOL2 messenger RNA were significantly lower in colon carcinomas than adenomas, and decreased significantly as carcinomas progressed from grades 2 to 4. Immunohistochemical analysis of a tissue array of 275 CRC samples showed a negative association between tumor stage and OVOL2 level. Overexpression of OVOL2 in SW620 cells decreased their migration and invasion, reduced markers of the epithelial-to-mesenchymal transition, and suppressed their metastasis as xenograft tumors in nude mice; knockdown of OVOL2 caused LS174T cells to transition from epithelial to mesenchymal phenotypes. OVOL2 bound T-cell factor (TCF)4 and ß-catenin, facilitating recruitment of histone deacetylase 1 to the TCF4-ß-catenin complex; this inhibited expression of epithelial-to-mesenchymal transition-related genes regulated by WNT, such as SLUG, in CRC cell lines. OVOL2 was a downstream target of WNT signaling in LS174T and SW480 cells. The OVOL2 promoter was hypermethylated in late-stage CRC specimens from patients and in SW620 cells; hypermethylation resulted in OVOL2 down-regulation and an inability to inhibit WNT signaling. Disruption of Ovol2 in Apc(min/+) mice increased WNT activity in intestinal tissues and the formation of invasive intestinal tumors. CONCLUSIONS: OVOL2 is a colorectal tumor suppressor that blocks WNT signaling by facilitating the recruitment of histone deacetylase 1 to the TCF4-ß-catenin complex. Strategies to increase levels of OVOL2 might be developed to reduce colorectal tumor progression and metastasis.


Asunto(s)
Movimiento Celular , Neoplasias Colorrectales/metabolismo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Células CACO-2 , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/cirugía , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Genotipo , Células HCT116 , Células HEK293 , Histona Desacetilasa 1/metabolismo , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Fenotipo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Factores de Tiempo , Factor de Transcripción 4 , Factores de Transcripción/genética , Transfección , Carga Tumoral , beta Catenina/metabolismo
6.
J Cell Sci ; 126(Pt 13): 2877-89, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23613467

RESUMEN

Wnt signalling through ß-catenin and the lymphoid-enhancing factor 1/T-cell factor (LEF1/TCF) family of transcription factors maintains stem cell properties in both normal and malignant tissues; however, the underlying molecular pathway involved in this process has not been completely defined. Using a microRNA microarray screening assay, we identified let-7 miRNAs as downstream targets of the Wnt-ß-catenin pathway. Expression studies indicated that the Wnt-ß-catenin pathway suppresses mature let-7 miRNAs but not the primary transcripts, which suggests a post-transcriptional regulation of repression. Furthermore, we identified Lin28, a negative let-7 biogenesis regulator, as a novel direct downstream target of the Wnt-ß-catenin pathway. Loss of function of Lin28 impairs Wnt-ß-catenin-pathway-mediated let-7 inhibition and breast cancer stem cell expansion; enforced expression of let-7 blocks the Wnt-ß-catenin pathway-stimulated breast cancer stem cell phenotype. Finally, we demonstrated that the Wnt-ß-catenin pathway induces Lin28 upregulation and let-7 downregulation in both cancer samples and mouse tumour models. Moreover, the delivery of a modified lin28 siRNA or a let-7a agomir into the premalignant mammary tissues of MMTV-wnt-1 mice resulted in a complete rescue of the stem cell phenotype driven by the Wnt-ß-catenin pathway. These findings highlight a pivotal role for Lin28/let-7 in Wnt-ß-catenin-pathway-mediated cellular phenotypes. Thus, the Wnt-ß-catenin pathway, Lin28 and let-7 miRNAs, three of the most crucial stem cell regulators, connect in one signal cascade.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/genética , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Genes Reporteros , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Ratones , Ratones Noqueados , MicroARNs/genética , Células Madre Neoplásicas/patología , Proteínas de Unión al ARN/genética , Activación Transcripcional , Proteína Wnt1/genética , beta Catenina/genética
7.
J Cell Sci ; 126(Pt 24): 5692-703, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24101726

RESUMEN

Wnt-ß-catenin signaling participates in the epithelial-mesenchymal transition (EMT) in a variety of cancers; however, its involvement in hepatocellular carcinoma (HCC) and downstream molecular events is largely undefined. HNF4α is the most prominent and specific factor maintaining the differentiation of hepatic lineage cells and a potential EMT regulator in HCC cells. However, the molecular mechanisms by which HNF4α maintains the differentiated liver epithelium and inhibits EMT have not been completely defined. In this study, we systematically explored the relationship between Wnt-ß-catenin signaling and HNF4α in the EMT process of HCC cells. Our results indicated that HNF4α expression was negatively regulated during Wnt-ß-catenin signaling-induced EMT through Snail and Slug in HCC cells. In contrast, HNF4α was found to directly associate with TCF4 to compete with ß-catenin but facilitate transcription co-repressor activities, thus inhibiting expression of EMT-related Wnt-ß-catenin targets. Moreover, HNF4α may control the switch between the transcriptional and adhesion functions of ß-catenin. Overexpression of HNF4α was found to completely compromise the Wnt-ß-catenin-signaling-induced EMT phenotype. Finally, we determined the regulation pattern between Wnt-ß-catenin signaling and HNF4α in rat tumor models. Our studies have identified a double-negative feedback mechanism controlling Wnt-ß-catenin signaling and HNF4α expression in vitro and in vivo, which sheds new light on the regulation of EMT in HCC. The modulation of these molecular processes may be a method of inhibiting HCC invasion by blocking Wnt-ß-catenin signaling or restoring HNF4α expression to prevent EMT.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Neoplasias Hepáticas Experimentales/patología , Masculino , Unión Proteica , Ratas , Ratas Wistar , Factores de Transcripción de la Familia Snail , Factor de Transcripción 4 , Factores de Transcripción/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
8.
Gut ; 61(5): 714-24, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21873734

RESUMEN

AIMS: Wnt signalling is involved in cellular homeostasis and development. Dysregulation of the Wnt signalling pathway has been linked to colorectal cancer. The orphan nuclear receptor TR3 plays important roles in proliferation and apoptosis. In this study, we investigated how TR3 suppresses intestinal tumorigenesis by regulating Wnt signalling. METHODS: Intestinal polyps were quantified in Apc(min/+), Apc(min/+)/TR3(-/-) and Apc(min/+)/villin-TR3 mice. Wnt signalling activity was evaluated by assessing ß-galactosidase activity in a BAT-Gal reporter strain. The TR3 agonist cytosporone B was used to evaluate the role of TR3 in intestinal tumorigenesis. Crosstalk between TR3 and ß-catenin/TCF4 was analysed by molecular methods in colorectal cancer cells. The phosphorylation of TR3 by glycogen synthase kinase (GSK) 3ß and the correlation between GSK3ß activity and TR3 phosphorylation were evaluated in clinical samples and colorectal cancer cells. RESULTS: TR3 was found to significantly suppress Wnt signalling activity and the proliferation of intestinal epithelial cells. Apc(min/+)/TR3(-/-) mice developed more intestinal polyps than Apc(min/+)/TR3(+/+) mice, whereas either transgenic overexpression of TR3 in the intestine or treatment with cytosporone B in Apc(min/+) mice significantly decreased intestinal tumour number. Mechanistically, TR3 disrupted the association of ß-catenin and TCF4 on chromatin and facilitated the recruitment of transcriptional co-repressors to the promoters of Wnt signalling target genes. However, TR3 was phosphorylated by GSK3ß in most clinical colorectal cancers, which attenuated the inhibitory activity of TR3 towards Wnt signalling. CONCLUSIONS: TR3 is a negative regulator of Wnt signalling and thus significantly suppresses intestinal tumorigenesis in Apc(min/+) mice. This inhibitory effect of TR3 may be paradoxically overcome through phosphorylation by GSK3ß in clinical colorectal cancers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/metabolismo , Mucosa Intestinal/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Vía de Señalización Wnt , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proliferación Celular , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Mucosa Intestinal/patología , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Factor de Transcripción 4 , beta Catenina/metabolismo , beta-Galactosidasa/metabolismo
9.
Cell Death Dis ; 14(1): 64, 2023 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-36707511

RESUMEN

The pregnane X receptor (PXR) is an important regulator of hepatocellular carcinoma cellular resistance to antitumor drugs. Activation of PXR was modulated by the co-regulators. The target protein for the Xenopus plus end-directed kinesin-like protein (Xklp2) known as TPX2 that was previously considered as a tubulin regulator, also functions as the regulator of some transcription factors and pro-oncogenes in human malignances. However, the actions of TPX2 on PXR and HCC cells are still unclear. In the present study, our results demonstrate that the high expression of endogenous mRNA level of TPX2 not only correlated with the poor prognosis of advanced HCC patients who received sorafenib treatment but also with expression of PXR's downstream genes, cyp3a4 and/or mdr-1. Results from luciferase and real-time polymerase chain reaction (qPCR) showed that TPX2 leads to enhancement of the transcription factor activation of PXR. Protein-protein interactions between PXR and TPX2 were identified using co-immunoprecipitation. Mechanically, overexpression of TPX2 led to enhancement of PXR recruitment to its downstream gene cyp3a4's promoter region (the PXRE region) or enhancer region (the XREM region). Treatment of HCC cells with paclitaxel, a microtubule promoter, led to enhancement of the effects of TPX2, whereas vincristine, a microtubule depolymerizing agent caused a decrease in TPX2-associated effects. TPX2 was found to cause acceleration of the metabolism or clearance of sorafenib, a typical tyrosine kinase inhibitor (TKI) in HCC cells and in turn led to the resistance to sorafenib by HCC cells. By establishing novel actions of TXP2 on PXR in HCC cells, the results indicate that TPX2 could be considered a promising therapeutic target to enhance HCC cells sensitivity to antitumor drugs.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Receptor X de Pregnano/genética , Sorafenib/farmacología , Sorafenib/uso terapéutico , Factores de Transcripción/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Citocromo P-450 CYP3A/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Ciclo Celular/genética
10.
Front Oncol ; 12: 985082, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36106099

RESUMEN

Increasing evidence indicates that the oncoprotein murine double minute (MDM2) binding protein (MTBP) can be considered a pro-oncogene of human malignancies; however, its function and mechanisms in hepatocellular carcinoma (HCC) are still not clear. In the present work, our results demonstrate that MTBP could function as a co-activator of transcription factor E26 transformation-specific sequence (ETS-1), which plays an important role in HCC cell proliferation and/or metastasis and promotes proliferation of HCC cells. Using luciferase and real-time polymerase chain reaction (qPCR) assays, MTBP was found to enhance the transcription factor activation of ETS-1. The results from chromatin co-immunoprecipitation showed that MTBP enhanced the recruitment of ETS-1 to its downstream gene's (mmp1's) promoter region with ETS-1 binding sites. In cellular and nude mice models, overexpression of MTBP was shown to promote the proliferation of MHCC97-L cells with low endogenous MTBP levels, whereas the knockdown of MTBP led to inhibition of the proliferation of MHCC97-H cells that possessed high endogenous levels of MTBP. The effect of MTBP on ETS-1 was confirmed in the clinical specimens; the expression of MTBP was positively correlated with the downstream genes of ETS-1, mmp3, mmp9, and uPA. Therefore, by establishing the role of MTBP as a novel co-activator of ETS-1, this work expands our knowledge of MTBP or ETS-1 and helps to provide new ideas concerning HCC-related research.

11.
Sci China Chem ; 65(3): 630-640, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35126481

RESUMEN

Outbreaks of both influenza virus and the novel coronavirus SARS-CoV-2 are serious threats to human health and life. It is very important to establish a rapid, accurate test with large-scale detection potential to prevent the further spread of the epidemic. An optimized RPA-Cas12a-based platform combined with digital microfluidics (DMF), the RCD platform, was established to achieve the automated, rapid detection of influenza viruses and SARS-CoV-2. The probe in the RPA-Cas12a system was optimized to produce maximal fluorescence to increase the amplification signal. The reaction droplets in the platform were all at the microliter level and the detection could be accomplished within 30 min due to the effective mixing of droplets by digital microfluidic technology. The whole process from amplification to recognition is completed in the chip, which reduces the risk of aerosol contamination. One chip can contain multiple detection reaction areas, offering the potential for customized detection. The RCD platform demonstrated a high level of sensitivity, specificity (no false positives or negatives), speed (≤30 min), automation and multiplexing. We also used the RCD platform to detect nucleic acids from influenza patients and COVID-19 patients. The results were consistent with the findings of qPCR. The RCD platform is a one-step, rapid, highly sensitive and specific method with the advantages of digital microfluidic technology, which circumvents the shortcomings of manual operation. The development of the RCD platform provides potential for the isothermal automatic detection of nucleic acids during epidemics. Electronic Supplementary Material: Supplementary material is available in the online version of this article at 10.1007/s11426-021-1169-1.

12.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 38(2): 108-112, 2022 Mar.
Artículo en Zh | MEDLINE | ID: mdl-36031566

RESUMEN

Objective: To construct the lentivirus overexpression vector with two label genes fused with CopGFP and PuroR and to detect the emission of green fluorescence as well as resistance to puromycin in liver cancer cells infected with lentivirus packaged with the above vector. Methods: Firstly, two fragments containing copGFP and PuroR coding sequences were amplified from pCDH-CMV-MCS-copGFP and pLKO.1 respectively; secondly, the two amplified regions were fused with each other by recombinant PCR; thirdly, the fusion DNA fragment was cut and inserted into pCDH-CMV-MCS-copGFP vector, which was linearized with the same restriction endonuclease as used to digest fusion DNA fragment: BamH Ⅰ and Sal Ⅰ. The fusion region in the constructed vector was confirmed by DNA sequencing. The checked vector was co-transfected with package assistant plasmids, namely PLP1, PLP2 and VSVG into in 293T cells and the culture supernatant was subjected to centrifuge and infect liver cancer MHCC97H cells, which were then used to detect their resistance to puromycin (infected cells were treated with 1 mg/ml puromycin for 7 days after infection) and to observe green fluorescence emission in microscope. To determine its efficiency in expressing foreign target protein, the Sp1 coding region was inserted into the MCS sites of the vector, and Sp1 mRNA and protein expression levels were compared with the vehicle vector by RT-qPCR and Western blot. Results: The lentivirus overexpression vector with two label genes fused with CopGFP and PuroR was successfully constructed, and the liver cancer cells infected with lentivirus packaged with the vector expressing two labeling genes fused with CopGFP and PuroRshowed both emission of green fluorescence and resistance to puromycin simultaneously, while cells containing with the vector inserted with Sp1 coding region improved Sp1 mRNA level with 3.3 fold and protein level with 2.2 fold higher in comparison with cells containing the vehicle vector (P<0.01). Conclusion: The fused label genes consisting of copGFP and PuroR are correctly cloned into the lentivirus vector and confer cells with the ability to emission of green fluorescence and resistance to puromycin, besides, the vector may promote the expression of the target gene with long coding sequence.


Asunto(s)
Infecciones por Citomegalovirus , Neoplasias Hepáticas , Vectores Genéticos , Humanos , Lentivirus , Puromicina , ARN Mensajero , Transfección
13.
Emerg Infect Dis ; 17(6): 1053-5, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21749768

RESUMEN

We analyzed changes in immunologic values over time for 28 hospitalized patients with pandemic (H1N1) 2009. Levels of interleukin-6, interferon-y, and interleukin-10 increased 1 day after illness onset and then decreased to baseline levels. Levels of virus-specific antibody were undetectable 1 day after illness onset and peaked 36 days later.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/epidemiología , Gripe Humana/inmunología , Pandemias , Adolescente , Adulto , Anticuerpos Antivirales/sangre , China/epidemiología , Citocinas/sangre , Citocinas/inmunología , Humanos , Gripe Humana/sangre , Recuento de Linfocitos , Persona de Mediana Edad , Adulto Joven
14.
Zhonghua Gan Zang Bing Za Zhi ; 19(11): 815-7, 2011 Nov.
Artículo en Zh | MEDLINE | ID: mdl-22433301

RESUMEN

OBJECTIVE: To quantitatively detect intrahepatic HBV covalently closed circular DNA (cccDNA) and serum HBsAg; and to analyze the relationship between the two parameters and with serum HBV DNA level. METHODS: Intrahepatic cccDNA (copies/cell) was quantitated by plasmid-safe ATP-dependent Danes (PSAD) digestion in combination with rolling circle amplification and gap-spanning selective real-time PCR assay using formalin fixed paraffin-embedded liver biopsy samples. HBsAg was measured by chemiluminescence's reagent manufactured by Abbott Company using sera sampled at time-point of liver biopsy. RESULTS: Intrahepatic cccDNA level was positively correlated with serum HBsAg level (r = 0.459, P < 0.001), but not correlated with serum HBV DNA level. Serum HBsAg level was positively correlated with serum HBV DNA level (r = 0.328, P = 0.015), and reversely correlated with HBV replicative efficiency defined as the ratio of serum HBV DNA to cccDNA (r = -0.373, P = 0.005). CONCLUSION: In patients with chronic hepatitis B, intrahepatic cccDNA level is correlated with serum HBsAg level. The two parameters combined with serum HBV DNA may comprehensively reflect HBV replication activity and help evaluation of antiviral therapeutic efficacy.


Asunto(s)
ADN Circular/análisis , ADN Viral/análisis , Antígenos de Superficie de la Hepatitis B/sangre , Hepatitis B Crónica/sangre , Hepatitis B Crónica/virología , Femenino , Virus de la Hepatitis B/genética , Humanos , Hígado/virología , Masculino , Carga Viral
15.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 28(1): 129-34, 2011 Feb.
Artículo en Zh | MEDLINE | ID: mdl-21485199

RESUMEN

According to the specific construction of the pSUPER. retro. puro vector, RNA interfering with the hypoxia-inducible factor 1alpha (HIF-1alpha) plasmid was constructed in this research programme. The efficiency was (87.15 +/- 2.36)% and green fluorescent protein was observed after 36 hours when the constructed plasmid co-transformed into the prostatic carcinoma cell line PC-3; compared with the control groups, this constructed plasmid could reduce the expression of total RNA and protein in PC-3 cells significantly after 48 hours. The cells were checked out by the plasmid resistance against the puromycin biotin, the clones of which were selected and enlarged for two weeks, then the cell strain of pSUPER-siHIF-1alpha/PC-3 was collected. The HIF-1alpha protein expression of the pSUPER-siHIF-1alpha/ PC-3 cells of also decreased significantly. The results showed that the RNA interference could be used in the construction of expression vector of constructed siRNA inhibiting the expression of HIF-1alpha with PC-3 cells, which is the basis of researching the pathology, multiplication, and metastasis of HIF-1alpha in the prostatic carcinoma and other cancers.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias de la Próstata/genética , Transfección , Secuencia de Bases , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Datos de Secuencia Molecular , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
16.
Front Oncol ; 11: 715193, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34249768

RESUMEN

The MDM2 binding protein (MTBP) has been considered an important regulator of human malignancies. In this study, we demonstrate that the high level of MTBP's endogenous expression is correlated with poor prognosis of advanced hepatocellular carcinoma (HCC) patients who received sorafenib. MTBP interacted with the Pregnane X receptor (PXR) and enhanced the transcription factor activity of PXR. Moreover, MTBP enhanced the accumulation of PXR in HCC cells' nuclear and the recruitment of PXR to its downstream gene's (cyp3a4's) promoter region. Mechanically, the knockdown of MTBP in MHCC97-H cells with high levels of MTBP decelerated the clearance or metabolism of sorafenib in HCC cells and led to the resistance of HCC cells to sorafenib. Whereas overexpression of MTBP in in MHCC97-L cells with low levels of MTBP showed the opposite trend. By establishing the interaction between MTBP and PXR, our results indicate that MTBP could function as a co-activator of PXR and could be a promising therapeutic target to enhance the sensitivity of HCC cells to molecular targeting agents.

17.
HLA ; 98(5): 459-466, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34375029

RESUMEN

Mastering the SNP content in the HLA region can be based on it to judiciously select unrelated donor stem cells with preferable MHC matching to lower postoperative complications. Herein, quantitative PCR-based primers and probes were designed for 10 transplants outcome-associated SNP loci with two-allelic polymorphism, and then a new detection system ("HLA-10-SNP") was established. Compared with Sanger sequencing, its accuracy has been proven to reach 100%. Additionally, fluorescent PCR typing of 10 important SNPs via this system expressed excellent repeatability (sensitivity, 20 ng). Overall, the new system achieves single-sample classification precision and easily distinguishable results, equipped with the advantages of simple, rapid, accurate, and effective, promising to acquire widespread popularization and application in clinical settings.


Asunto(s)
Polimorfismo de Nucleótido Simple , Donante no Emparentado , Alelos , Prueba de Histocompatibilidad , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
Sci Adv ; 7(41): eabg6262, 2021 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-34623920

RESUMEN

Molecular pathways controlling emigration of mature thymocytes from thymus to the periphery remain incompletely understood. Here, we show that T cell­specific ablation of glycogen synthase kinase 3 (GSK3) led to severely impaired thymic egress. In the absence of GSK3, ß-catenin accumulated in the cytoplasm, where it associated with and activated Akt, leading to phosphorylation and degradation of Foxo1 and downregulation of Klf2 and S1P1 expression, thereby preventing emigration of thymocytes. A cytoplasmic membrane-localized ß-catenin excluded from the nucleus promoted Akt activation, suggesting a new function of ß-catenin independent of its role as a transcriptional activator. Furthermore, genetic ablation of ß-catenin, retroviral expression of a dominant negative Akt mutant, and transgenic expression of a constitutively active Foxo1 restored emigration of GSK3-deficient thymocytes. Our findings establish an essential role for GSK3 in thymocyte egress and reveal a previously unidentified signaling function of ß-catenin in the cytoplasm.

19.
J Neurooncol ; 100(1): 31-41, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20204459

RESUMEN

Gliomas are common malignant tumors of the human neural system, and Wnt signaling activation is closely related to glioma malignancy. Human Pygopus 2 (Pygo2) was recently discovered to be a component of the Wnt signaling pathway, which is required for ß-catenin/Tcf-dependent transcription. However, the role of Pygo2 in glioblastoma cell growth and survival remains uncertain. In the present study, Pygo2 expression was evaluated in 80 glioma tissue samples. Results demonstrated that tumor grade exhibited a positive correlation with overexpression of Pygo2. In addition, small hairpin RNA (shRNA) was used to specifically knockdown Pygo2 expression in human glioblastoma U251 cell lines. Results showed that inhibition of Pygo2 expression resulted in inhibited cell proliferation and invasiveness, as well as increased cell cycle arrest at the G(1) stage and decreased expression of the Wnt target gene cyclin D1. These results demonstrated that Pygo2 was highly expressed in glioma tissue and required for growth of glioblastoma cells.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación hacia Abajo/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Glioblastoma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , ARN Interferente Pequeño/farmacología , Adulto , Análisis de Varianza , Neoplasias Encefálicas/patología , Bromodesoxiuridina , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclina D/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/patología , Humanos , Indoles , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Factores de Tiempo , beta Catenina/metabolismo
20.
Onco Targets Ther ; 13: 7165-7176, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32801748

RESUMEN

BACKGROUND: Although molecular-targeted agents are still the first choice for advanced hepatocellular carcinoma (HCC) treatment, the therapeutic efficacy of these agents is not satisfactory. Recently, the mammalian target of rapamycin (mTOR) is considered to be a promising molecular target that can enhance the sensitivity of HCC cells to antitumor therapy. However, the reported mTOR inhibitors have some shortcomings, and novel mTOR inhibitors need to be developed to enhance the antitumor effect of molecularly targeted agents on advanced HCC. METHODS: In this study, five small-molecular compounds that could serve as potential mTOR-specific inhibitors were identified by virtual screening. The activity of tert-butyl (4-(9-(2-(1,3-dioxolan-2-yl)ethyl)-6-morpholino-9H-purin-2-yl)phenyl)carbamate (compound 4) was measured by enzyme test and Western blot, and its antitumor effect on HCC was examined in nude mice subcutaneous tumor model. RESULTS: The results showed that 4 is the most effective one in inhibiting the activation of mTOR kinase (mTOR IC50 = 17.52±3.67 nmol/L) among the five lead compounds. Further research in this study indicated that treatment with 4 enhanced the sensitivity of HCC cells to the molecular-targeted agents, such as sorafenib, regorafenib, lenvatinib, anlotinib, and apatinib. In addition, this research indicated that mTOR was correlated with the poor prognosis in patients with advanced HCC who received sorafenib. CONCLUSION: Our study identified a new type of small-molecular inhibitors of mTOR and confirmed their ability to enhance the antitumor effect of molecular-targeted agents on advanced HCC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA