Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 29(5): 1042-1054, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34815549

RESUMEN

Somatic cell reprogramming is an ideal model for studying epigenetic regulation as it undergoes dramatic chromatin remodeling. However, a role for phosphorylation signaling in chromatin protein modifications for reprogramming remains unclear. Here, we identified mitogen-activated protein kinase kinase 6 (Mkk6) as a chromatin relaxer and found that it could significantly enhance reprogramming. The function of Mkk6 in heterochromatin loosening and reprogramming requires its kinase activity but does not depend on its best-known target, P38. We identified Gatad2b as a novel target of Mkk6 phosphorylation that acts downstream to elevate histone acetylation levels and loosen heterochromatin. As a result, Mkk6 over-expression facilitates binding of Sox2 and Klf4 to their targets and promotes pluripotency gene expression during reprogramming. Our studies not only reveal an Mkk phosphorylation mediated modulation of chromatin status in reprogramming, but also provide new rationales to further investigate and improve the cell fate determination processes.


Asunto(s)
Cromatina , Heterocromatina , Reprogramación Celular , Epigénesis Genética , MAP Quinasa Quinasa 6/genética , MAP Quinasa Quinasa 6/metabolismo , Fosforilación
3.
Cell Death Discov ; 7(1): 117, 2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011971

RESUMEN

Steroid receptor RNA activator 1 (SRA1) has been described as a novel transcriptional co-activator that affects the migration of cancer cells. Through RT-PCR, we identified that skipping exon 3 of SRA1 produces two isoforms, including the truncated short isoform, SRA1-S, and the long isoform, SRA1-L. However, the effect of these two isomers on the migration of HCC cells, as well as the specific mechanism of exon 3 skipping remain unclear. In this study, we found up regulated expression of SRSF1 and SRA1-L in highly metastatic HCCLM3, as well as in HCCs with SRSF1 demonstrating the strongest correlation with SRA1-L. In contrast, we observed a constitutively low expression of SRA1-S and SRSF1 in lowly metastatic HepG2 cells. Overexpression of SRSF1 or SRA1-L promoted migration and invasion by increasing the expression of CD44, while SRA1-S reversed the effect of SRSF1 and SRA1-L in vitro. In addition, lung metastasis in mice revealed that, knockdown of SRSF1 or SRA1-L inhibited the migration of HCC cells, while SRA1-L overexpression abolished the effect of SRSF1 knockout and instead promoted HCC cells migration in vivo. More importantly, RNA immunoprecipitation and Cross-link immunoprecipitation analyses showed that SRSF1 interacts with exon 3 of SRA1 to up regulate the expression of SRA1-L in HCC cells. RNA pull-down results indicated that SRSF1 could also bind to exon 3 of SRA1 in vitro. Finally, minigene -MS2 mutation experiments showed that mutation of the SRA1 exon 3 binding site for SRSF1 prevented the binding of SRA1 pre-mRNA. In summary, our results provide experimental evidence that SRA1 exon 3 inclusion is up regulated by SRSF1 to promote tumor invasion and metastasis in hepatocellular carcinoma.

4.
Theranostics ; 10(13): 5719-5735, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32483414

RESUMEN

The Axl gene is known to encode for a receptor tyrosine kinase involved in the metastasis process of cancer. In this study, we investigated the underlying molecular mechanism of Axl alternative splicing. Methods: The expression levels of PTBP1 in hepatocellular carcinoma (HCC) tissues were obtained from TCGA samples and cell lines. The effect of Axl-L, Axl-S, and PTBP1 on cell growth, migration, invasion tumor formation, and metastasis of liver cancer cells were measured by cell proliferation, wound-healing, invasion, xenograft tumor formation, and metastasis. Interaction between PTBP1 and Axl was explored using cross-link immunoprecipitation, RNA pull-down assays and RNA immunoprecipitation assays. Results: Knockdown of the PTBP1 and exon 10 skipping isoform of Axl (Axl-S), led to impaired invasion and metastasis in hepatoma cells. Immunoprecipitation results indicated that Axl-S protein binds more robustly with Gas6 ligand than Axl-L (exon 10 including) and is more capable of promoting phosphorylation of ERK and AKT proteins. Furthermore, cross-link immunoprecipitation and RNA-pulldown assays revealed that PTBP1 binds to the polypyrimidine sequence(TCCTCTCTGTCCTTTCTTC) on Axl-Intron 9. MS2-GFP-IP experiments demonstrated that PTBP1 competes with U2AF2 for binding to the aforementioned polypyrimidine sequence, thereby inhibiting alternative splicing and ultimately promoting Axl-S production. Conclusion: Our results highlight the biological significance of Axl-S and PTBP1 in tumor metastasis, and show that PTBP1 affects the invasion and metastasis of hepatoma cells by modulating the alternative splicing of Axl exon 10.


Asunto(s)
Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Neoplasias Hepáticas/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Empalme Alternativo/genética , Animales , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Exones/genética , Regulación Neoplásica de la Expresión Génica/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , Hígado/patología , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Proteína de Unión al Tracto de Polipirimidina/genética , Proteínas Proto-Oncogénicas/metabolismo , Precursores del ARN/genética , Empalme del ARN/genética , ARN Mensajero/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factor de Empalme U2AF/genética , Factor de Empalme U2AF/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
5.
Biochim Biophys Acta Gene Regul Mech ; 1862(8): 858-869, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31075540

RESUMEN

Alternative splicing (AS) events occur in the majority of human genes. AS in a single gene can give rise to different functions among multiple isoforms. Human ortholog of mammalian enabled (Mena) is a conserved regulator of actin dynamics that plays an important role in metastasis. Mena has been shown to have multiple splice variants in human tumor cells due to AS. However, the mechanism mediated Mena AS has not been elucidated. Here we showed that polypyrimidine tract-binding protein 1 (PTBP1) could modulate Mena AS. First, PTBP1 levels were elevated in metastatic lung cancer cells as well as during epithelial-mesenchymal transition (EMT) process. Then, knockdown of PTBP1 using shRNA inhibited migration and invasion of lung carcinoma cells and decreased the Mena exon11a skipping, whereas overexpression of PTBP1 had the opposite effects. The results of RNA pull-down assays and mutation analyses demonstrated that PTBP1 functionally targeted and physically interacted with polypyrimidine sequences on both upstream intron11 (TTTTCCCCTT) and downstream intron11a (TTTTTTTTTCTTT). In addition, the results of migration and invasion assays as well as detection of filopodia revealed that the effect of PTBP1 was reversed by knockdown of Mena but not Mena11a+. Overexpressed MenaΔ11a also rescued the PTBP1-induced migration and invasion. Taken together, our study provides a novel mechanism that PTBP1 modulates Mena exon11a skipping, and indicates that PTBP1 depends on the level of Mena11a- to promote lung cancer cells migration and invasion. The regulation of Mena AS may be a potential prognostic marker and a promising target for treatment of lung carcinoma.


Asunto(s)
Empalme Alternativo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Neoplasias Pulmonares/genética , Proteínas de Microfilamentos/genética , Proteína de Unión al Tracto de Polipirimidina/genética , Células A549 , Movimiento Celular , Transición Epitelial-Mesenquimal , Exones , Regulación Neoplásica de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Invasividad Neoplásica , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Regulación hacia Arriba
6.
Eur J Pharmacol ; 852: 77-89, 2019 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-30831079

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a disease with a poor prognosis and high mortality, posing a major threat to human health. Increased levels of inflammatory cytokines, reactive oxygen species and coagulation cascade have been extensively reported in IPF. We previously fused Hirudin and human manganese superoxide dismutase (hSOD2) to generate a dual-feature fusion protein, denoted as rhSOD2-Hirudin fusion protein. In this study, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) and Hydroxyproline (HYP) assays were used to investigate the effects of rhSOD2-Hirudin protein on thrombin-induced fibroblast proliferation and collagen accumulation in vitro. Subsequently, the mice model of pulmonary fibrosis induced by bleomycin was used for evaluating the anti-inflammatory and anti-fibrotic effects of rhSOD2-Hirudin protein in vivo. Results showed that rhSOD2-Hirudin protein could inhibit the proliferation of fibroblasts and reduce the HYP production in vitro by inhibiting the activity of thrombin. In vivo experiments showed that lung inflammation and fibrosis were significantly decreased in rhSOD2-Hirudin protein-treated mice. Furthermore, rhSOD2-Hirudin protein treatment reduced profibrotic protein and gene expression while reducing the number of inflammatory cells in the lung. In conclusion, rhSOD2-Hirudin protein can effectively attenuate pulmonary fibrosis in vitro and in vivo, mainly by inhibiting the activity of thrombin meanwhile increasing SOD2 levels prevent cells from being damaged by reactive oxygen species, thereby mitigating IPF progression. This study provided important information on the feasibility and efficacy of rhSOD2-Hirudin protein as a novel therapeutic agent for IPF.


Asunto(s)
Bleomicina/farmacología , Hirudinas/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Superóxido Dismutasa/farmacología , Actinas/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidroxiprolina/biosíntesis , Ratones , Células 3T3 NIH , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Proteínas Recombinantes de Fusión/uso terapéutico , Factor de Crecimiento Transformador beta1/metabolismo
7.
Biotechnol Appl Biochem ; 66(1): 21-32, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30192404

RESUMEN

Lung cancer is a serious threat to human health. Studies have revealed that human manganese superoxide dismutase (hSOD2) and miRNAs play an essential role in the metastasis process of lung cancer. However, the miRNAs that associated with hSOD2 and involved in metastasis, remain elusive. After databases analysis and dual luciferase reporter validation, we demonstrated that miR-330-3p expression inversely correlated with hSOD2b expression level, and that miR-330-3p directly targeted the 3'untranslated region (3'UTR) of hSOD2b. Furthermore, overexpression of miR-330-3p promoted whereas knockdown of miR-330-3p inhibited invasion/migration and the epithelial-mesenchymal transition (EMT) process of lung cancer cells in vitro. Knockdown of miR-330-3p inhibited metastasis of lung cancer cells in vivo. Moreover, miR-330-3p-mediated enhancement of invasion/migration in 95-D cells could be rescued by over-expression of hSOD2. In conclusion, we demonstrated that miR-330-3p promoted metastasis of lung cancer cells by suppressing hSOD2b expression and unveiled a new clinical application of miR-330-3p in the therapy of lung cancer.


Asunto(s)
Movimiento Celular , Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Neoplásico/metabolismo , Superóxido Dismutasa/metabolismo , Células A549 , Transición Epitelial-Mesenquimal , Células HeLa , Células Hep G2 , Humanos , Células K562 , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Células MCF-7 , MicroARNs/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , ARN Neoplásico/genética , Superóxido Dismutasa/genética
8.
J Microbiol Biotechnol ; 26(11): 1881-1890, 2016 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-27435539

RESUMEN

Manganese superoxide dismutase (MnSOD) is a vital enzyme that protects cells from free radicals through eliminating superoxide radicals (O²â»). Hirudin, a kind of small active peptide molecule, is one of the strongest anticoagulants that can effectively cure thrombus diseases. In this study, we fused Hirudin to the C terminus of human MnSOD with the GGGGS linker to generate a novel dual-feature fusion protein, denoted as hMnSOD-Hirudin. The hMnSOD-Hirudin gene fragment was cloned into the pET15b (SmaI, CIAP) vector, forming a recombinant pET15b-hMnSOD-Hirudin plasmid, and then was transferred into Escherichia coli strain Rosetta-gami for expression. SDS-PAGE was used to detect the fusion protein, which was expected to be about 30 kDa upon IPTG induction. Furthermore, the hMnSOD-Hirudin protein was heavily detected as a soluble form in the supernatant. The purification rate observed after Ni NTA affinity chromatography was above 95%. The hMnSOD-Hirudin protein yield reached 67.25 mg per liter of bacterial culture. The identity of the purified protein was confirmed by western blotting. The hMnSOD-Hirudin protein activity assay evinced that the antioxidation activity of the hMnSOD-Hirudin protein obtained was 2,444.0 ± 96.0 U/mg, and the anticoagulant activity of the hMnSOD-Hirudin protein was 599.0 ± 35.0 ATU/mg. In addition, in vitro bioactivity assay showed that the hMnSODHirudin protein had no or little cytotoxicity in H9c2, HK-2, and H9 (human CD4⁺, T cell) cell lines. Transwell migration assay and invasion assay showed that the hMnSOD-Hirudin protein could suppress human lung cancer 95-D cell metastasis and invasion in vitro.


Asunto(s)
Hirudinas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Superóxido Dismutasa/genética , Superóxido Dismutasa/farmacología , Línea Celular Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Hirudinas/metabolismo , Hirudinas/farmacología , Humanos , Neoplasias Pulmonares/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Superóxido Dismutasa/metabolismo
9.
Mol Cell Biochem ; 385(1-2): 79-86, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24078003

RESUMEN

Human manganese superoxide dismutase (hMnSOD) is a new type of cancer suppressor. Nonamer of arginine (R9) is an efficient protein transduction domain (PTD). The aim of the study was to improve the transduction efficiency of hMnSOD and investigate its activity in vitro. In this study, we designed, constructed, expressed, and purified a novel fusion protein containing the hMnSOD domain and R9 PTD (hMnSOD­R9). The DNA damaged by Fenton's reagent was found to be significantly reduced when treated with hMnSOD­R9. hMnSOD­R9 fusion protein was successfully delivered into HeLa cells. The MTT assay showed that proliferation of various cancer cell lines were inhibited by hMnSOD­R9 in a dose-dependent manner. In addition, the cell cycle of HeLa cells was arrested at the sub-G0 phase by hMnSOD­R9. hMnSOD­R9 induced apoptosis of HeLa cells in a dose-dependent manner. With hMnSOD­R9 treatment, Bax, JNK, TBK1 gene expression was increased and STAT3 gene expression was gradually down-regulated in HeLa cells. We also found that apoptosis was induced by hMnSOD­R9 in HeLa cells via up-regulation of cleaved caspase-3 and down-regulation phospho-STAT3 pathway. These results indicated that hMnSOD­R9 may provide benefits to cervical cancer treatment.


Asunto(s)
Arginina/metabolismo , Membrana Celular/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Superóxido Dismutasa/metabolismo , Neoplasias del Cuello Uterino/patología , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Daño del ADN , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/metabolismo , Células HeLa , Humanos , Microscopía Confocal , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Transducción Genética , Neoplasias del Cuello Uterino/enzimología , Neoplasias del Cuello Uterino/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...