Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 421
Filtrar
1.
Int J Mol Sci ; 24(1)2022 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-36613758

RESUMEN

Plant biomass is the most abundant renewable resource in nature. In a circular economy perspective, the implementation of its bioconversion into fermentable sugars is of great relevance. Lytic Polysaccharide MonoOxygenases (LPMOs) are accessory enzymes able to break recalcitrant polysaccharides, boosting biomass conversion and subsequently reducing costs. Among them, auxiliary activity of family 9 (AA9) acts on cellulose in synergism with traditional cellulolytic enzymes. Here, we report for the first time, the production of the AA9 LPMOs from the mesophilic Trichoderma reesei (TrAA9B) and the thermophilic Thermoascus aurantiacus (TaAA9B) microorganisms in tobacco by plastid transformation with the aim to test this technology as cheap and sustainable manufacture platform. In order to optimize recombinant protein accumulation, two different N-terminal regulatory sequences were used: 5' untranslated region (5'-UTR) from T7g10 gene (DC41 and DC51 plants), and 5' translation control region (5'-TCR), containing the 5'-UTR and the first 14 amino acids (Downstream Box, DB) of the plastid atpB gene (DC40 and DC50 plants). Protein yields ranged between 0.5 and 5% of total soluble proteins (TSP). The phenotype was unaltered in all transplastomic plants, except for the DC50 line accumulating AA9 LPMO at the highest level, that showed retarded growth and a mild pale green phenotype. Oxidase activity was spectrophotometrically assayed and resulted higher for the recombinant proteins without the N-terminal fusion (DC41 and DC51), with a 3.9- and 3.4-fold increase compared to the fused proteins.


Asunto(s)
Proteínas Fúngicas , Oxigenasas de Función Mixta , Celulosa/química , Proteínas Fúngicas/biosíntesis , Oxigenasas de Función Mixta/biosíntesis , Polisacáridos/metabolismo , Nicotiana/metabolismo , Plantas Modificadas Genéticamente/metabolismo , Plastidios
2.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830310

RESUMEN

Tryptophan metabolism plays a role in the occurrence and development of hepatocellular carcinoma cells. By degrading certain amino acids, tumor growth can be limited while maintaining the body's normal nutritional requirements. Tryptophan side-chain oxidase (TSO) enzyme can degrade tryptophan, and its inhibitory effect on hepatocellular carcinoma cells is worthy of further study. To investigate the degradation effect on tryptophan, TSO was isolated and purified from qq Pseudomonas. The reaction products were identified with high performance liquid chromatography (HPLC) and high-performance liquid chromatography tandem mass spectrometry (HPLC-MS). De novo sequencing provided the complete amino acid sequence of TSO. The results of CCK-8, colony formation, transwell, and qPCR confirmed that TSO had inhibitory effects on the proliferation and migration of HCCLM3 (human hepatocarcinoma cell line) and HepG2 cells. The results of flow cytometry confirmed its apoptotic activity. In animal experiments, we found that the tumor-suppressive effect was better in the oncotherapy group than the intraperitoneal injection group. The results of immunohistochemistry also suggested that TSO could inhibit proliferation and promote apoptosis. In conclusion, a specific enzyme that can degrade tryptophan and inhibit the growth of hepatoma cells was authenticated, and its basic information was obtained by extraction/purification and amino acid sequencing.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Bacterianas/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Oxigenasas de Función Mixta/farmacología , Triptófano/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Oxigenasas de Función Mixta/biosíntesis , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/aislamiento & purificación , Modelos Moleculares , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estructura Secundaria de Proteína , Pseudomonas/química , Pseudomonas/enzimología , Pseudomonas/genética , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
3.
Appl Immunohistochem Mol Morphol ; 29(10): 720-727, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34433181

RESUMEN

The human aspartyl ß-hydroxylase (ASPH) is overexpressed in tumor tissues. Bronchoalveolar lavage (BAL) is a diagnostic procedure for infections and malignancies. The aim of this study was to investigate whether tumor exosomes carrying ASPH gene marker were present in bronchoalveolar fluid of patients with non-small cell lung cancer (NSCLC). A tissue microarray analysis was applied to explore the expression of ASPH in different histologic NSCLC. The human NSCLC cell lines and normal bronchial cell lines were used to study exosomal ASPH exprerssion. A total of 27 NSCLC, 21 benign tumor, and 15 healthy controls underwent BAL. Immunohistochemistry was performed to study the ASPH expression in malignant and normal lung tissues. The expression characteristics of ASPH in different NSCLC and normal bronchial cells and pneumocytes were confirmed by cell blocks. A reverse transcription-quantitative polymerase chain reaction was carried out to study the levels of exosomal ASPH expression. Immunohistochemical staining of tissue microarray demonstrated that overexpression of ASPH was found in NSCLC tissues including adenocarcinoma, large cell carcinoma, and squamous cell carcinoma, but absent in adjacent normal tissues. All NSCLC specimens exhibited high levels of ASPH immunoreactivity, while nonmalignant and normal lung tissues exhibited a very low level of expression. Overexpression of ASPH was found in exosomes from NSCLC cell lines but absent from the normal bronchial cell line NL-20. ASPH level from BAL exosomes was significantly increased in NSCLC patients compared with that from nonmalignant or health group. Our method of isolation of BAL exosomes was easily performed in the clinical laboratory. BAL exosomal ASPH can be a potential biomarker for NSCLC diagnosis.


Asunto(s)
Lavado Broncoalveolar , Proteínas de Unión al Calcio/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas , Exosomas/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , Proteínas de la Membrana/biosíntesis , Oxigenasas de Función Mixta/biosíntesis , Proteínas Musculares/biosíntesis , Proteínas de Neoplasias/biosíntesis , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología
4.
Prostate ; 80(12): 977-985, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32542727

RESUMEN

BACKGROUND: Recently, resveratrol (Res) has been suggested to suppress the migration and invasion of prostate cancer (PCa). In the present study, we aimed to investigate the effects of Res on genomic DNA methylation, as well as the migration and invasion of PCa cells. METHODS: The suppression by Res of the growth of PCa cells was verified through a cytotoxicity assay. In addition, the effects of Res on 5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC), and ten-eleven translocation 1 (TET1) levels were assessed, and the cell migration and invasion were also determined. The expressions of TET1, tissue inhibitor of metalloproteinases (TIMP) 2, TIMP3, MMP2, and MMP9 were detected through Western blot analysis. Afterward, TET1 was silenced using lentiviral short hairpin RNA to examine the effect of TET1 on the Res-triggered inhibition of migration and invasion of PCa cells. RESULTS: Our results showed that Res upregulated the 5hmC and TET1 levels and downregulated the 5mC level. Moreover, Res also inhibited the migration and invasion of PCa cells, promoted the demethylation of TIMP2 and TIMP3 to upregulate their expressions, and suppressed the expressions of MMP2 and MMP9. The silencing of TET1 in the presence of Res showed that Res could exert its effect through TET1. CONCLUSIONS: Our findings indicated that Res inhibited the migration and invasion of PCa cells via the TET1/TIMP2/TIMP3 pathway, which might potentially serve as a target for the treatment of PCa.


Asunto(s)
Oxigenasas de Función Mixta/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Resveratrol/farmacología , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Células HEK293 , Humanos , Masculino , Oxigenasas de Función Mixta/biosíntesis , Oxigenasas de Función Mixta/genética , Invasividad Neoplásica , Células PC-3 , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Resveratrol/farmacocinética , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-3/biosíntesis , Inhibidor Tisular de Metaloproteinasa-3/genética , Regulación hacia Arriba
5.
Epigenetics Chromatin ; 12(1): 41, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31266538

RESUMEN

BACKGROUND: While aberrant DNA methylation is a characteristic feature of tumor cells, our knowledge of how these DNA methylation patterns are established and maintained is limited. DNA methyltransferases and ten-eleven translocation methylcytosine dioxygenases (TETs) function has been found altered in a variety of cancer types. RESULTS: Here, we report that in T cell acute lymphoblastic leukemia (T-ALL) the MYC oncogene controls the expression of TET1 and TET2 to maintain 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) patterns, which is associated with tumor cell-specific gene expression. We found that cellular senescence and tumor regression upon MYC inactivation in T-ALL was associated with genome-wide changes in 5mC and 5hmC patterns. Correlating with the changes in DNA (hydroxy)methylation, we found that T-ALL overexpress TET1, while suppressing TET2 in a MYC-dependent fashion. Consequently, MYC inactivation led to an inverse expression pattern, decreasing TET1, while increasing TET2 levels. Knockdown of TET1 or ectopic expression of TET2 in T-ALL was associated with genome-wide changes in 5mC and 5hmC enrichment and decreased cell proliferation, suggesting a tumor promoting function of TET1, and a tumor suppressing role for TET2. Among the genes and pathways controlled by TET1, we found ribosomal biogenesis and translational control of protein synthesis highly enriched. CONCLUSIONS: Our finding that MYC directly deregulates the expression of TET1 and TET2 in T-ALL provides novel evidence that MYC controls DNA (hydroxy)methylation in a genome-wide fashion. It reveals a coordinated interplay between the components of the DNA (de)methylating machinery that contribute to MYC-driven tumor maintenance, highlighting the potential of specific TET enzymes for therapeutic strategies.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/biosíntesis , Regulación Leucémica de la Expresión Génica , Oxigenasas de Función Mixta/biosíntesis , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , Línea Celular Tumoral , Citosina/metabolismo , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Humanos , Ratones , Ratones Transgénicos , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética
6.
Cancer Med ; 8(3): 990-1003, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30784212

RESUMEN

Increasing evidence revealed that ten-eleven translocation 1 (TET1) plays an important role in tumorigenesis and chemoresistance, but its functions in gemcitabine resistance in cholangiocarcinoma (CCA) remain unknown. This study aims to investigate the effect of TET1 on gemcitabine resistance in CCA and the possible effect on P-glycoprotein (P-gp) expression encoded by multidrug resistance (MDR) genes. We established two kinds of gemcitabine-resistant CCA cell lines and confirmed its specific features. The expression of TET1 and P-gp was evaluated in gemcitabine-resistant CCA cells and their parental cells at mRNA and protein level by quantitative RT-PCR and western blot analysis. After transfecting the gemcitabine-resistant CCA cell lines with TET1 gene or siRNA, the cell viability test was obtained to verify the effect of TET1 on the sensitivity of CCA cells to gemcitabine. And then, the possible effect of TET1 on the expression of P-gp was examined by western blot analysis. Xenograft tumor experiment was conducted to confirm the association between TET1 and P-gp expression under gemcitabine chemoresistance. The associations between clinical outcomes of CCA patients with chemotherapy and TET1 expression were analyzed in 82 patients. The results showed that TET1 expression was significantly decreased, and P-gp expression was increased in gemcitabine-resistant CCA cells. Additionally, overexpression of TET1 augmented the sensitivity of CCA cells to gemcitabine and induced the decreased expression of P-gp in gemcitabine-resistant CCA cells. Furthermore, multivariate Cox regression analysis indicated that TET1 expression and TNM stage were independent risk factors (P < 0.001) for the clinical outcomes of CCA patients with chemotherapy. Additionally, Kaplan-Meier survival and the log-rank test showed that decreased expression of TET1 was associated with poorer prognosis of CCA patients with chemotherapy. These findings suggest that TET1 expression reverses gemcitabine resistance in CCA accompanied by a reduction in P-gp expression. Thus, TET1 may be a promising target to overcome chemoresistance in CCA.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Oxigenasas de Función Mixta/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Animales , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Desoxicitidina/uso terapéutico , Resistencia a Antineoplásicos , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
7.
Cancer Lett ; 450: 53-62, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-30771438

RESUMEN

DNA hypermethylation is a driving force in carcinogenesis. However, the role of active DNA hypomethylation in cancer remains largely unknown. This process, facilitated by ten-eleven translocation methylcytosine dioxygenase 1 (TET1), which oxidizes 5-methylcytosine (5 mC) to 5-hydroxymethylcytosine (5hmC), has never been studied in cervical cancer. Here, we found that TET1 and 5hmC correlative increases from normal cervix to Low-grade squamous intraepithelial lesion (LSIL), maximizing in High-grade squamous intraepithelial lesion (HSIL), and decreasing in invasive cancer. Full-length HPV-immortalized HSIL cells demonstrated higher TET1/5hmC levels, and stemness properties, compared to invasive cancer cells. TET1 silencing promoted the epithelial-mesenchymal transition (EMT), to transform precancerous cells in vivo. TET1 increased 5hmC in the ZEB1 and VIM promoters, surprisingly, silencing both genes. TET1 interaction with the histone modifiers, LSD1 and EZH2, on the ZEB1 promoter, resulted in gene silencing, via loss of histone H3K4 trimethylation, and gain of histone H3K27 trimethylation. Taken together, TET1 promotes stemness properties, and inhibits EMT, in HSIL cells, through 5hmC-dependent and -independent mechanisms.


Asunto(s)
5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Células HeLa , Xenoinjertos , Humanos , Ratones , Oxigenasas de Función Mixta/biosíntesis , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas/biosíntesis , Lesiones Intraepiteliales Escamosas de Cuello Uterino/metabolismo , Lesiones Intraepiteliales Escamosas de Cuello Uterino/patología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Vimentina , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , Displasia del Cuello del Útero/metabolismo , Displasia del Cuello del Útero/patología
8.
Mol Med Rep ; 17(5): 6569-6575, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29512746

RESUMEN

Increasing evidence has shown that abnormal expression of miR-4284 participates in the progression of several types of cancer. However, the expression and the role of miR­4284 in gastric cancer remain largely unknown. Therefore, in the present study the miR­4284 expression levels in gastric cancer tissues and cell lines, was examined using reverse transcription­quantitative polymerase chain reaction (RT­qPCR) and found that miR­4284 was significantly upregulated in 40 pairs of gastric cancer tissues and five gastric cancer cell lines compared to the corresponding normal tissues and GES­1 cell line. In addition, increased miR­4284 expression was positively associated with TNM stage (P=0.035), distal metastasis (P=0.022) and poor prognosis in gastric cancer patients. Furthermore, the overexpression of miR­4284 expression was shown to promote cell proliferation, clone formation, invasion and migration, while the suppression of miR­4284 expression induced opposite effects. Additionally, luciferase reporter assay was conducted and showed that ten-eleven translocation 1 (TET1), a tumor suppressor gene that regulating cell survival and metastasis, was a direct target of miR­4284. Upregulated miR­4284 decreased the mRNA and protein levels of TET1 in SGC­7901 cells and downregulated miR­4284 increased the mRNA and protein levels of TET1 in AGS cells. In addition, miR­4284 expression was negatively correlated with the TET1 expression in gastric cancer tissues. Moreover, inhibition of TET1 suppressed the effect of miR­4284 inhibitors on cell proliferation in AGS cells. Therefore, data demonstrated that miR­4284 could promote tumor cell growth, migration and invasion by directly targeting TET1 in gastric cancer, which may provide a potential therapeutic target for gastric cancer treatment.


Asunto(s)
Movimiento Celular , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Oxigenasas de Función Mixta/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , ARN Neoplásico/metabolismo , Neoplasias Gástricas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Femenino , Humanos , Masculino , MicroARNs/genética , Oxigenasas de Función Mixta/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas/genética , ARN Neoplásico/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
9.
Int J Oncol ; 52(4): 1295-1304, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29484426

RESUMEN

Osteosarcoma has become one of the most common primary malignant tumors affecting children and adolescents. Although increasing evidence has indicated that microRNAs (miRNAs or miRs) play important roles in the development of osteosarcoma, the expression of miR­27a­3p and its effects on osteosarcoma are not yet fully understood. In the present study, our data demonstrated that the expression of miR­27a­3p in osteosarcoma cell lines was significantly higher than that in the normal human osteoblastic cell line, hFOB 1.19 cell (P<0.01). In order to explore the role of miR­27a­3p in the development and progression of osteosarcoma, the expression of miR­27a­3p was inhibited by transfection of the MG-63 cells with miR­27a­3p inhibitor. The results revealed that the cell proliferative ability significantly decreased (P<0.01), the number of apoptotic cells significantly increased (P<0.01) and the number of cells passing through the Transwell membrane was significantly reduced in the group transfected with the miR­27a­3p inhibitor (P<0.01). At the same time, the expression of E-cadherin and α-catenin was significantly upregulated (P<0.01), while the expression of vimentin was significantly downregulated in the group transfected with the miR­27a­3p inhibitor (P<0.01). Our results also revealed that the mRNA expression of ten-eleven translocation 1 (TET1) in the osteosarcoma cells was significantly downregulated compared with that in the hFOB 1.19 cells (P<0.01). Luciferase reporter system analysis indicated that miR­27a­3p recognized the TET1 3'-UTR. The protein expression of TET1 significantly increased in the group transfected with the miR­27a­3p inhibitor. The results from CCK-8 assay, flow cytometric assay and Transwell invasion analysis revealed that TET1 knockdown inhibited the biological effects induced by the downregulation of miR­27a­3p. Taken together, the findings of this study indicate that miR­27a­3p is upregulated, while TET1 is downregulated in human osteosarcoma cells. miR­27a­3p inhibition suppresses the proliferation and invasion of osteosarcoma cells, and promotes cell apoptosis via the negative regulation of TET1. miR­27a­3p/TET1 may thus be a potential target for the treatment of osteosarcoma.


Asunto(s)
Neoplasias Óseas/patología , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/metabolismo , Oxigenasas de Función Mixta/biosíntesis , Osteosarcoma/patología , Proteínas Proto-Oncogénicas/biosíntesis , Apoptosis/genética , Neoplasias Óseas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , MicroARNs/genética , Oxigenasas de Función Mixta/genética , Invasividad Neoplásica/genética , Osteosarcoma/genética , Proteínas Proto-Oncogénicas/genética
10.
Cell Prolif ; 51(2): e12426, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29277934

RESUMEN

OBJECTIVES: Ten-eleven translocation 1 (TET1) is a DNA methylcytosine (mC) dioxygenase discovered recently that can convert 5-mC into 5-hydroxymethylcytosine (5hmC). We previously reported that TET1 promotes odontoblastic differentiation of human dental pulp cells (hDPCs). The gene encoding the family with sequence similarity 20, member C (FAM20C) protein, is a potential TET1 target and showed demethylation during odontoblastic differentiation of hDPCs in our previous study. This study aimed to explore whether TET1-mediated hydroxymethylation could activate the FAM20C gene, thereby regulating hDPC differentiation. MATERIALS AND METHODS: The expression pattern of FAM20C and its potential changes during odontoblastic induction of hDPCs were assessed by Western blotting. Lentivirus-mediated transduction with short hairpin RNA (shRNA) was used to knock down FAM20C and TET1 expression in hDPCs. The mineralization potential of hDPCs was evaluated with an ALPase activity assay and by observing the mineralized matrix deposition and the expression of odontoblast-related markers DSPP and DMP1. Recombinant human FAM20C protein (rhFAM20C) was reintroduced into shTET1 cells in a rescue experiment. The dynamic hydroxymethylation status of the FAM20C gene promoter was examined using hydroxymethylated DNA immunoprecipitation (IP)-PCR. Chromatin IP-PCR and agarose gel electrophoresis were utilized to validate the recruitment of TET1 to its target loci in the FAM20C promoter. RESULTS: FAM20C protein level was upregulated after the odontoblastic induction of hDPCs. shRNA-mediated FAM20C suppression reduced the expression of DSPP and DMP1 after odontoblastic induction for 7 and 14 days. ALPase activity was reduced on day 7, and the formation of mineralized nodules was attenuated on day 14 after odontoblastic induction in FAM20C-inhibited hDPCs. Genomic 5hmC levels significantly decreased, and total 5mC levels increased in TET1-deficient hDPCs. In addition, a significant reduction in FAM20C also emerged. The rhFAM20C treatment of shTET1 cells attenuated the mineralization abnormalities caused by TET1 depletion. TET1 depletion prompted a decline in 5hmC levels in several regions on the FAM20C promoter. Enhanced TET1 recruitment was detected at the corresponding loci in the FAM20C promoter during odontoblastic induction. CONCLUSION: TET1 knockdown suppressed odontoblastic differentiation by restraining its direct binding to FAM20C promoter, and hence inhibiting FAM20C hydroxymethylation and subsequent transcription. These results suggest that TET1 potentially promotes the cytodifferentiation potential of hDPCs through its DNA demethylation machinery and upregulation of FAM20C protein expression.


Asunto(s)
Calcificación Fisiológica , Quinasa de la Caseína I/biosíntesis , Diferenciación Celular , Pulpa Dental/enzimología , Proteínas de la Matriz Extracelular/biosíntesis , Regulación Enzimológica de la Expresión Génica , Oxigenasas de Función Mixta/biosíntesis , Odontoblastos/enzimología , Proteínas Proto-Oncogénicas/biosíntesis , Adolescente , Adulto , Quinasa de la Caseína I/genética , Pulpa Dental/citología , Proteínas de la Matriz Extracelular/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Metilación , Oxigenasas de Función Mixta/genética , Odontoblastos/citología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética
11.
Cancer Sci ; 108(12): 2454-2461, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28985022

RESUMEN

Although prognostic markers for early estrogen receptor (ER)-positive breast cancer have been extensively developed, predictive markers for adjuvant endocrine therapy are still lacking. Focusing on the mechanisms underlying endocrine resistance, we investigated whether the endocrine sensitivity of ER-positive breast cancer cells was correlated with the expression of aspartate-ß-hydroxylase (ASPH), which is involved in the development of hepatocellular carcinoma. ASPH expression in ER-positive and tamoxifen-resistant breast cancer cells was upregulated by the MAPK and phosphoinositide-3 kinase (PI3K) pathways, which both play pivotal roles in endocrine resistance. In the clinical setting, ASPH expression was negatively correlated with recurrence-free survival of luminal B breast cancer patients that received adjuvant endocrine therapy, but not in patients that did not receive adjuvant endocrine therapy. Luminal B breast cancer is one of the intrinsic molecular subtypes identified by the Prediction Analysis of Microarray 50 (PAM50) multiple gene classifier, and because of its poor response to endocrine therapy, chemotherapy in addition to endocrine therapy is generally required after surgical resection. Our results suggest that the endocrine sensitivity of luminal B breast cancer can be assessed by examining ASPH expression, which promotes the consideration of a prospective study on the association between ASPH expression at the mRNA and protein levels in luminal B breast cancer and subsequent response to endocrine therapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Proteínas de Unión al Calcio/biosíntesis , Resistencia a Antineoplásicos/fisiología , Proteínas de la Membrana/biosíntesis , Oxigenasas de Función Mixta/biosíntesis , Proteínas Musculares/biosíntesis , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Receptores de Estrógenos/metabolismo
12.
Cancer Gene Ther ; 24(9): 373-380, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28776568

RESUMEN

Lung cancer is one of the leading causes of death from cancer worldwide, with a poor prognosis in advanced cases. In the past decade, epidermal growth factor receptor (EGFR) inhibitors have shown significant efficacy towards treatment for EGFR mutant lung cancer. Expanding our knowledge of oncogenic EGFR signaling pathways is therefore of highly importance for the cancer field. Recently it has been proposed that mutant EGFR transcriptionally silences the TET1 (ten-eleven translocation methylcytosine dioxygenase 1) gene in cellular and animal models of lung cancer. Since TET1 is a known DNA demethylase, EGFR-mediated TET1 silencing therefore downregulates demethylation of tumor suppressor genes, which then leads to tumor growth inhibition, potentiating the role of TET1 as a tumor suppressor gene in NSCLC. In our study, we examined the role of EGFR-TET1 silencing in NSCLC patient samples. By independently analyzing the TCGA (The Cancer Genome Atlas) NSCLC data set as well as a cohort of patient samples from our hospital and a data set from publicly deposited databases, we did not observe the aforementioned mutant EGFR silencing of TET1. Conversely, in our cohort, TET1 expression levels were significantly elevated in EGFR mutant samples (P=0.007). Patients with higher TET1 levels showed a trend of better response rates to EGFR inhibitors compared to low TET1 staining levels, although the result was not significant (P=0.08). Furthermore, we did not observe a correlation between TET1 expression levels and patient survival. We conclude that while oncogenic EGFR suppression of TET1 is established in cellular and animal models of lung cancer, its role in patient outcome and prognosis remains inconclusive and warrants further investigation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Regulación hacia Abajo , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , Oxigenasas de Función Mixta/biosíntesis , Mutación , Proteínas Proto-Oncogénicas/biosíntesis , Anciano , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Bases de Datos Genéticas , Receptores ErbB/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Oxigenasas de Función Mixta/genética , Proteínas Proto-Oncogénicas/genética
13.
Gynecol Oncol ; 147(2): 408-417, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28851501

RESUMEN

OBJECTIVE: Epithelial ovarian cancer (EOC) is the deadliest type of ovarian cancer, but the mechanisms contributing to its tumorigenesis are not well understood. Herein, we will elucidate the role of Ten-eleven translocation 1 (TET1) in EOC development. METHODS: The expression of TET1 in EOC cell lines and primary samples was examined by western blot and immunohistochemistry. The biological role of ectopic TET1 overexpression was revealed by a series of in vitro functional studies. Its downstream signaling pathway was predicted by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of microarray data. The methylation level and expression of Wnt/ß-catenin signaling inhibitors Dikkopf 1 (DKK1) and secreted Fzd receptor protein 2 (SFRP2) were examined by Chromatin immunoprecipitation (ChIP) assay, Epimark™ 5hmC and 5mC level analysis and quantitative RT-PCR. Small interference RNA (siRNA) technology was used to investigate the biological roles of DKK1 and SFRP2. RESULTS: TET1 expression was inversely correlated with clinical stage in patients with EOC by tissue microarray (TMA). TET1 expression was undetected in 6 types of EOC cell lines. Ectopic expression of TET1 inhibited colony formation, cell migration and invasion in SKOV3 and OVCAR3 cells. Furthermore, TET1 overexpression reversed the epithelial-mesenchymal transition (EMT) process of SKOV3 cells. Mechanistically, TET1 potently inhibited canonical Wnt/ß-catenin signaling by demethylating and upregulating two upstream antagonists of this pathway, SFRP2 and DKK1, which was associated with inhibition of EMT and cancer cell metastasis. CONCLUSION: This study uncovers that TET1 has potent tumor-suppressive effects in EOC by activating Wnt/ß-catenin signaling inhibitors DKK1 and SFRP2.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Oxigenasas de Función Mixta/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , beta Catenina/metabolismo , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Oxigenasas de Función Mixta/biosíntesis , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas/biosíntesis , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Análisis de Matrices Tisulares , Regulación hacia Arriba , Vía de Señalización Wnt
14.
Blood Cancer J ; 7(7): e587, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28731456

RESUMEN

The Ten Eleven Translocation (TET) enzymes have been found to be mutated in both diffuse large B-cell (DLBCL) and peripheral T-cell (PTCL) lymphomas resulting in DNA hypermethylation. Recent studies in embryonal stem cells showed that ascorbic acid (AA) is a cofactor for TET with a binding site at the catalytic domain, and enhances TET activity. We hypothesized that AA could potentially enhance TET activity in lymphoma cells to cause DNA demethylation, reactivate expression of tumor suppressor genes and enhance chemosensitivity. We demonstrate in vitro that AA treatment of DLBCL and PTCL cells using AA concentrations achievable intravenously increased TET activity leading to DNA demethylation. This epigenetic effect is independent of hydrogen peroxide. AA treatment increased the expression of SMAD1, a tumor suppressor gene known to be suppressed by methylation, and increased chemosensitivity of lymphoma cells. Twenty-nine percent (10/34) of unselected lymphoma patients had plasma AA levels that were deficient suggesting an additional clinical mechanism of TET hypofunction. These data indicate that AA has the potential to modify TET function in lymphoma and enhance chemosensitivity. In addition, the AA deficiency seen in some patients may further impair TET function and contribute to resistance. Clinical trials testing intravenous AA with chemotherapy are warranted.


Asunto(s)
Ácido Ascórbico/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Linfoma de Células B Grandes Difuso/embriología , Oxigenasas de Función Mixta/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Línea Celular Tumoral , Humanos , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología
15.
Environ Toxicol Pharmacol ; 54: 142-145, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28728133

RESUMEN

Zebrafish are an attractive model organism for toxicology; however, an important consideration in translating between species is xenobiotic metabolism/bioactivation. CYP2E1 metabolizes small hydrophobic molecules, e.g. ethanol, cigarette smoke, and diesel exhaust components. CYP2E1 is thought to only be conserved in mammals, but recent reports identified homologous zebrafish cytochrome P450s. Herein, ex vivo biochemical measurements show that unlike mammals, zebrafish possess a low-affinity 4-nitrophenol hydroxylase (Km ∼0.6 mM) in hepatic microsomes and mitochondria that is inducible only 1.5- to 2-fold by ethanol and is insensitive to 4-methylpyrazole inhibition. In closing, we suggest creating improved models to study CYP2E1 in zebrafish.


Asunto(s)
Etanol/farmacología , Hígado/metabolismo , Oxigenasas de Función Mixta/metabolismo , Nitrofenoles/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Encéfalo/metabolismo , Citocromo P-450 CYP2E1 , Inducción Enzimática , Masculino , Microsomas Hepáticos/metabolismo , Mitocondrias/metabolismo , Oxigenasas de Función Mixta/biosíntesis , Miocardio/metabolismo , Ratas , Pez Cebra , Proteínas de Pez Cebra/biosíntesis
16.
J Cell Biochem ; 118(10): 3193-3204, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28252217

RESUMEN

CpG methylation of DNA takes part in a specific epigenetic memory that plays crucial roles in the differentiation and abnormality of the cells. The methylation pattern aberration of genomes is affected in three ways, namely DNA methyltransferase (DNMT), ten-eleven translocation (TET), and methyl-binding domain (MBD) proteins. Of these, TET enzymes have recently been demonstrated to be master modifier enzymes in the DNA methylation process. Additionally, recent studies emphasize that not only epigenetic phenomena play a role in controlling hypoxia pathway, but the hypoxia condition also triggers hypomethylation of genomes that may help with the expression of hypoxia pathway genes. In this study, we suggested that TET1 and TET2 could play a role in the demethylation of genomes under chemical hypoxia conditions. Herein, the evaluating methylation status and mRNA expression of mentioned genes were utilized through real-time PCR and methylation-specific PCR (MSP), respectively. Our results showed that TET1 and TET2 genes were overexpressed (P < 0.05) under chemical hypoxia conditions in Retinal Pigment Epithelial (RPE) cells, whereas the promoter methylation status of them were hypomethylated in the same condition. Therefore, chemical hypoxia not only causes overexpression of TET1 and TET2 but also could gradually do promoter demethylation of same genes. This is the first study to show the relationship between epigenetics and the expression of mentioned genes related to hypoxia pathways. Furthermore, it seems that these associations in RPE cells are subjected to chemical hypoxia as a mechanism that could play a crucial role in methylation pattern changes of hypoxia-related diseases such as cancer and ischemia. J. Cell. Biochem. 118: 3193-3204, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/biosíntesis , Epigénesis Genética , Oxigenasas de Función Mixta/biosíntesis , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/biosíntesis , Epitelio Pigmentado de la Retina/metabolismo , Hipoxia de la Célula , Dioxigenasas , Femenino , Humanos , Masculino , Epitelio Pigmentado de la Retina/citología
17.
Cancer Res ; 77(9): 2351-2362, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28249902

RESUMEN

Obesity will soon surpass smoking as the most preventable cause of cancer. Hypercholesterolemia, a common comorbidity of obesity, has been shown to increase cancer risk, especially colorectal cancer. However, the mechanism by which hypercholesterolemia or any metabolic disorder increases cancer risk remains unknown. In this study, we show that hypercholesterolemia increases the incidence and pathologic severity of colorectal neoplasia in two independent mouse models. Hypocholesterolemia induced an oxidant stress-dependent increase in miR101c, which downregulated Tet1 in hematopoietic stem cells (HSC), resulting in reduced expression of genes critical to natural killer T cell (NKT) and γδ T-cell differentiation. These effects reduced the number and function of terminally differentiated NKT and γδ T cells in the thymus, the colon submucosa, and during early tumorigenesis. These results suggest a novel mechanism by which a metabolic disorder induces epigenetic changes to reduce lineage priming of HSC toward immune cells, thereby compromising immunosurveillance against cancer. Cancer Res; 77(9); 2351-62. ©2017 AACR.


Asunto(s)
Neoplasias Colorrectales/genética , Hipercolesterolemia/genética , MicroARNs/genética , Oxigenasas de Función Mixta/genética , Obesidad/genética , Proteínas Proto-Oncogénicas/genética , Animales , Carcinogénesis , Diferenciación Celular/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , Células Madre Hematopoyéticas/patología , Humanos , Hipercolesterolemia/complicaciones , Hipercolesterolemia/patología , Activación de Linfocitos/genética , Ratones , MicroARNs/biosíntesis , Oxigenasas de Función Mixta/biosíntesis , Células T Asesinas Naturales/patología , Obesidad/complicaciones , Obesidad/patología , Estrés Oxidativo/genética , Proteínas Proto-Oncogénicas/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Tumour Biol ; 39(3): 1010428317695017, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28349832

RESUMEN

Ten-eleven translocation proteins are α-ketoglutarate-dependent dioxygenases involved in the conversion of 5-methylcytosines (5-mC) to 5-hydroxymethylcytosine (5-hmC), 5-formylcytosine, and 5-carboxylcytosine that play a significant role in DNA demethylation. Deregulation of TET genes expression and changes in the level of 5-hmC are thought to be associated with the onset and progression of several types of cancer, but there are no such data related to endometrial cancer. The aim of the work was to investigate the messenger RNA expression levels of TET1, TET2, and TET3 in relation to clinicopathological characteristics of endometrial cancer as well as the correlation between expression of TET genes and the level of 5-hmC/5-mC. The prognostic significance of TETs expression for overall survival was established. We found that TET1 and TET2 messenger RNA expression was lower and TET3 was higher in cancers compared to normal tissues. Positive correlation between 5-hmC and the relative expression of TET1 and TET2 was found, but no correlation was observed in the case of TET3. Decreased expression of TET1 and TET2 was significantly associated with increased lymph node metastasis and International Federation of Gynecology and Obstetrics stage. Kaplan-Meier analysis indicated that low TET1 expression predicted poor overall survival (p = 0.038). Multivariate analysis identified the TET1 expression in endometrial cancer as an independent prognostic factor. Our results suggest that decreased expression of TET1 correlates with tumor progression and may serve as a potential prognostic biomarker in endometrial cancer.


Asunto(s)
Metilación de ADN/genética , Proteínas de Unión al ADN/biosíntesis , Dioxigenasas/biosíntesis , Neoplasias Endometriales/genética , Oxigenasas de Función Mixta/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Anciano , Citosina/análogos & derivados , Citosina/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Oxigenasas de Función Mixta/genética , Pronóstico , Proteínas Proto-Oncogénicas/genética
19.
Clin Epigenetics ; 9: 18, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28228863

RESUMEN

BACKGROUND: Alteration of DNA methylation (5-methylcytosine, 5mC) patterns represents one of the causes of tumorigenesis and cancer progression. Tet proteins can oxidise 5mC to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine and 5-carboxylcytosine (5caC). Although the roles of these oxidised forms of 5mC (oxi-mCs) in cancer pathogenesis are still largely unknown, there are indications that they may be involved in the mechanisms of malignant transformation. Thus, reduction of 5hmC content represents an epigenetic hallmark of human tumours, and according to our recent report, 5caC is enriched in a proportion of breast cancers and gliomas. Nevertheless, the distribution of oxi-mCs in paediatric brain tumours has not been assessed. FINDINGS: Here, we analyse the global levels and spatial distribution of 5hmC and 5caC in four brain tumour cell lines derived from paediatric sonic hedgehog (SHH) pathway-activated medulloblastomas (Daoy and UW228-3) and ependymomas (BXD-1425EPN and DKFZ-EP1NS). We show that, unlike HeLa cells, the paediatric tumour cell lines possess both 5hmC and 5caC at immunochemically detectable levels and demonstrate that both modifications display high degrees of spatial overlap in the nuclei of medulloblastomas and ependymomas. Moreover, although 5hmC levels are comparable in the four brain tumour cell lines, 5caC staining intensities differ dramatically between them with highest levels of this mark in a subpopulation of DKFZ-EP1NS cells. Remarkably, the 5caC enrichment does not correlate with 5hmC levels and is not associated with alterations in thymine DNA glycosylase (TDG) expression in SHH medulloblastoma and ependymoma cell lines but corresponds to elevated levels of TET1 transcript in UW228-3 and DKFZ-EP1NS cells. CONCLUSIONS: We demonstrate that both 5caC enrichment and elevated TET1 expression are observed in SHH medulloblastomas and ependymomas. Our results suggest that increased Tet-dependent 5mC oxidation may represent one of the epigenetic signatures of cancers with neural stem cell origin and, thus, may contribute to development of novel approaches for diagnosis and therapy of the brain tumours.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Cerebelosas/metabolismo , Citosina/análogos & derivados , Ependimoma/metabolismo , Meduloblastoma/metabolismo , Oxigenasas de Función Mixta/biosíntesis , Oxigenasas de Función Mixta/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , 5-Metilcitosina/metabolismo , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Neoplasias Cerebelosas/genética , Niño , Citosina/metabolismo , Ependimoma/genética , Expresión Génica , Humanos , Inmunohistoquímica , Meduloblastoma/genética , Regulación hacia Arriba
20.
Oncol Res ; 23(3): 89-98, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26931431

RESUMEN

In the present study, we found that lung cancer cell line (H460 cells) expressing Tet1 showed higher levels of adhesion, and Tet1 inhibited H460 cell proliferation. In addition, these cells showed a significantly reduced ability of collagen degradation and Smad2/3 phosphorylation compared to controls. Furthermore, vimentin was found to be highly expressed in larger metastatic cancer area. Tet1 overexpression was reduced in the epithelial marker E-cadherin. Moreover, Tet1 repressed cancer cell metastasis in nude mice. Collectively, these findings suggest that Tet1 expression plays a critical role in metastasis of lung cancer cells by suppression of invasion and epithelial-mesenchymal transition (EMT).


Asunto(s)
Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Neoplasias Pulmonares/genética , Oxigenasas de Función Mixta/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Animales , Adhesión Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Ratones , Oxigenasas de Función Mixta/genética , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas/genética , Proteína Smad2/biosíntesis , Proteína Smad2/genética , Vimentina/biosíntesis , Vimentina/genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA