Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Oxid Med Cell Longev ; 2020: 5135893, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32190174

RESUMO

OBJECTIVE: The mechanism of enhanced radiosensitivity induced by mitochondrial uncoupling protein UCP2 was investigated in HeLa cells to provide a theoretical basis as a novel target for cervical cancer treatment. METHODS: HeLa cells were irradiated with 4 Gy X-radiation at 1.0 Gy/min. The expression of UCP2 mRNA and protein was assayed by real-time quantitative polymerase chain reaction and western blotting. UCP2 siRNA and negative control siRNA fragments were constructed and transfected into HeLa cells 24 h after irradiation. The effect of UCP2 silencing and irradiation on HeLa cells was determined by colony formation, CCK-8 cell viability, γH2AX immunofluorescence assay of DNA damage, Annexin V-FITC/PI apoptosis assay, and propidium iodide cell cycle assay. The effects on mitochondrial structure and function were investigated with fluorescent probes including dichlorodihydrofluorescein diacetate (DCFH-DA) assay of reactive oxygen species (ROS), rhodamine 123, and MitoTracker Green assay of mitochondrial structure and function. RESULTS: Irradiation upregulated UCP2 expression, and UCP2 knockdown decreased the survival of irradiated HeLa cells. UCP2 silencing sensitized HeLa cells to irradiation-induced DNA damage and led to increased apoptosis, cell cycle arrest in G2/M, and increased mitochondrial ROS. Increased radiosensitivity was associated with an activation of P53, decreased Bcl-2, Bcl-xl, cyclin B, CDC2, Ku70, and Rad51 expression, and increased Apaf-1, cytochrome c, caspase-3, and caspase-9 expression. CONCLUSIONS: UCP2 inhibition augmented the radiosensitivity of cervical cancer cells, and it may be a potential target of radiotherapy of advanced cervical cancer.


Assuntos
Tolerância a Radiação , Espécies Reativas de Oxigênio/metabolismo , Proteína Desacopladora 2/antagonistas & inibidores , Neoplasias do Colo do Útero/metabolismo , Apoptose/efeitos da radiação , Autofagia/efeitos da radiação , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Dano ao DNA , Reparo do DNA/efeitos da radiação , Regulação para Baixo/efeitos da radiação , Feminino , Inativação Gênica/efeitos da radiação , Células HeLa , Humanos , Potencial da Membrana Mitocondrial/efeitos da radiação , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Modelos Biológicos , Tolerância a Radiação/efeitos da radiação , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo , Neoplasias do Colo do Útero/patologia
3.
Gene ; 710: 178-185, 2019 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-31158449

RESUMO

In order to improve the therapeutic effect of non-small cell lung cancer (NSCLC), it is critical to combine radiation and gene therapy. Our study found that the activation of microRNA-9 (miR-9) conferred ionizing radiation (IR) sensitivity in cancer cells. Furthermore, increased microRNA-9 promoter methylation level was observed after IR. Our study combined the IR and microRNA-9 overexpression treatment which leads to a significant enhancement in the therapeutic efficiency in lung cancer both in vitro and in vivo. Therefore, it is plausible that microRNA-9 can be used as a novel therapeutic strategy of NSCLC. MTT assay was performed to detect the effect of microRNA-9 on the survival and growth of NSCLC cells. Flow cytometry results showed that microRNA-9 enhanced the apoptosis of NSCLC cells. Wound healing assay found that microRNA-9 can inhibit the migration of NSCLC cells and enhance the effect of radiation on the migration of NSCLC cells. In addition, bisulfate sequencing PCR was performed to analyze the methylation status of the microRNA-9 promoter. In order to determine the effect of microRNA-9 and its promoter methylation status on proliferation and radio-sensitivity in vivo, a subcutaneous tumor formation assay in nude mice was performed. Results have shown that microRNA-9 overexpression increased the radiosensitivity of A549 cells by inhibiting cell activity and migration, and by increasing apoptosis. In addition, the promoter methylation status of the microRNA-9 gene increased in response to ionizing radiation. Our study demonstrated that microRNA-9 enhanced radiosensitivity in NSCLC and this effect is highly regulated by its promoter methylation status. These results will help to clarify regulatory mechanisms of radiation resistance thus stimulate new methods for improving radiotherapy for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Metilação de DNA , Neoplasias Pulmonares/genética , MicroRNAs/genética , Tolerância a Radiação , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Epigênese Genética , Humanos , Neoplasias Pulmonares/radioterapia , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Regiões Promotoras Genéticas , Análise de Sequência de DNA
4.
Gene ; 678: 252-260, 2018 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-30099020

RESUMO

OBJECTIVE: To investigate the regulatory mechanism of MEN1 gene in radiation-induced lung fibrosis in mice and provide a new theoretical basis for the clinical treatment of radiation pulmonary fibrosis. METHODS: First, 80 C57BL/6 mice aged 8 weeks and weighing 18-22 g were selected, half of them were male and the other half were female. The mice were divided into control group and irradiation group (40 mice in each group) according to the method of the random number table. A radiation-induced lung fibrosis mouse model was established in which a single X-ray irradiation of 20 Gy was applied to the right lung in the irradiation group; H&E and Masson staining were used to verify whether the model was successful at 4, 8, 16 and 24 weeks after irradiation. The expression of MEN1, smooth muscle actin (α-SMA), Collagen-1 and transforming growth factor (TGF-ß) in lung tissue were detected by Western blot and qPCR. Secondly, in the mouse embryonic fibroblast cell line (MEF) and mouse lung epithelial cell line (MLE-12), we constructed cell models of MEN1 knockout and interference separately with the irradiation of 10 Gy X-rays. The expression of α-SMA, Collagen-1, and TGF-ß/Smads signaling pathway molecules was detected by qPCR. Finally, using the immunoprecipitation (IP) method, we can detect the interaction between Smad2 and the protein menin encoded by the MEN1 gene. RESULTS: The results of the radiation pulmonary fibrosis model in mice showed that compared with the control group, the alveolar septum widens, the alveolar integrity decreases, the lung tissue slightly thickens, and a small amount of collagen deposits appear after 4-8 weeks in the model group. At twenty-fourth weeks, a large number of cells in the interstitial space of the lung tissue and a localized focal fibrosis area were observed. Further study found that radiation induced fibrogenic inflammatory cytokines TGF-ß up-regulation, down-regulation of MEN1 gene expression, and then enhanced the expression of α-SMA and promotes the transformation of fibroblasts to myofibroblasts; At the same time, the expression of Collagen-1 was enhanced, which suggested that the extracellular matrix was overconcentrated and eventually promoted the formation of pulmonary fibrosis. In vitro, we found that knockout and interference of MEN1 gene can significantly enhance radiation-induced fibrosis, and up-regulate the expression of downstream molecules Smad2 and Smad3 of TGF-ß signaling pathway, and down-regulate the expression of Smad7. Furthermore, it played an important role in regulating the process of radionuclide fibrosis. CONCLUSION: MEN1 plays a key role in the formation of pulmonary fibrosis by regulating the secretion of TGF-ß and the activation of TGF-ß/Smads signaling pathway.


Assuntos
Actinas/metabolismo , Colágeno Tipo I/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fibrose Pulmonar/etiologia , Fator de Crescimento Transformador beta/metabolismo , Raios X/efeitos adversos , Actinas/genética , Animais , Linhagem Celular , Colágeno Tipo I/genética , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Transdução de Sinais , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/genética
5.
Oncol Rep ; 40(3): 1297-1306, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30015898

RESUMO

Transmembrane protein 165 (TMEM165), a Golgi protein, functions in ion homeostasis and vesicular trafficking in the Golgi apparatus. While mutations in TMEM165 are known to cause human 'congenital disorders of glycosylation', a recessive autosomal metabolic disease, the potential association of this protein with human cancer development has not been explored to date. In the present study, we revealed that TMEM165 is overexpressed in HCC and its depletion weakens the invasive activity of cancer cells through suppression of matrix metalloproteinase­2 (MMP­2) expression. Levels of TMEM165 mRNA and protein were clearly increased in HCC patient tissues and cell cultures. Quantitative real­time RT­PCR analysis of fresh HCC tissues (n=88) revealed association of TMEM165 overexpression with more frequent macroscopic vascular invasion, microscopic serosal invasion and higher α­fetoprotein levels. Notably, depletion of TMEM165 led to a marked decrease in the invasive activity of two different HCC cell types, Huh7 and SNU475, accompanied by downregulation of MMP­2. Our collective findings clearly indicated that TMEM165 contributed to the progression of HCC by promoting invasive activity, supporting its utility as a novel biomarker and therapeutic target for cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/patologia , Movimento Celular , Complexo de Golgi/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , Adulto , Idoso , Antiporters , Carcinoma Hepatocelular/metabolismo , Estudos de Casos e Controles , Proteínas de Transporte de Cátions , Feminino , Seguimentos , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , alfa-Fetoproteínas/metabolismo
6.
Biomed Pharmacother ; 105: 362-369, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29864624

RESUMO

Gastric cancer is characterized by resistance to ionizing radiation. The development of resistance to radiotherapy in gastric cancer patients is one of the obstacles to effective radiotherapy. MicroRNAs are small well-conserved non-coding RNA species that regulate post-transcriptional activation. Our study aimed to investigate the role of miR-196b in radiation-induced gastric cancer. In the present study, we found that miR-196b expression was significantly reduced following radiation. The ectopic miR-196b expression sensitized SNU-638 gastric cancer cells and increased γ-H2AX foci upon radiation treatment. Bioinformatics analysis suggested that the DNA repair protein RAD23B was a putative target gene of miR-196b. Overexpression of miR-196b suppressed RAD23B expression in SNU-638 cells. Reporter assays further showed that miR-196b inhibited RAD23B 3'-UTR luciferase activity. Knockdown of RAD23B by small interfering RNA transfection closely mimicked the outcomes of miR-196b transfection, leading to impaired DNA damage repair in gastric cancer cells. Our results show that miR-196b improved radiosensitivity of SNU-638 cells by targeting RAD23B. Our data indicate that miR-196b is a potential target to enhance the effect of radiation treatment on gastric cancer cells. These findings will provide evidence for a new therapeutic target in radiotherapy.


Assuntos
Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , MicroRNAs/metabolismo , Tolerância a Radiação/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Sequência de Bases , Linhagem Celular Tumoral , Dano ao DNA/genética , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Técnicas de Silenciamento de Genes , Humanos , MicroRNAs/genética , Radiação Ionizante
7.
Oncol Rep ; 40(1): 536-544, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29749478

RESUMO

Protein arginine methyltransferase 5 (PRMT5) is a protein that catalyzes transfer of methyl groups to the arginine residues of proteins and is involved in diverse cellular and biological responses. While the participation of PRMT5 in cancer progression has been increasingly documented, its association with the invasive phenotype currently remains poorly understood. In the present study, we revealed that PRMT5 is overexpressed in human hepatocellular carcinoma (HCC) and in colon cancer and its depletion leads to the suppression of cell invasive activity via the reduction of the expression of MMP-2. Real-time quantitative RT-PCR analysis of 120 HCC patient tissues revealed the overexpression of PRMT5 in HCC and the association of PRMT5 with aggressive clinicopathological parameters, such as poorer differentiation (P=0.004), more frequent hepatic vein invasion (P=0.019), larger tumor size (P=0.011) and higher α-fetoprotein levels (P=0.020). Similarly to the data obtained with HCC, overexpression of PRMT5 was also displayed in colon cancer tissues, compared to matched non-tumor regions. Consistent with the significant association of the overexpression of PRMT5 with hepatic vein invasion in patient specimens, PRMT5 depletion via siRNA transfection led to a marked reduction in the invasion rate in both HCC and colon cancer cells. Reduced invasion associated with PRMT5 depletion was accompanied by a decrease in the expression of MMP-2. Collectively, our results indicated that PRMT5 overexpression in HCC and colon cancer cells contributed to their acquisition of aggressive characteristics, such as invasiveness, thus presenting a promising therapeutic target for the treatment of these diseases.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteína-Arginina N-Metiltransferases/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Metaloproteinase 2 da Matriz/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , RNA Interferente Pequeno/genética
8.
Cancer Lett ; 346(2): 249-56, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24434653

RESUMO

DNA lesion-induced centrosomal abnormalities during the replication phase are relatively unknown. Here, we report that RNAi-mediated depletion of RRM1 induces cell-cycle arrest at the replication phase, along with severe DNA damage and centrosomal amplification. Interestingly, CHK1 depletion synergistically increased RRM1-depletion-induced centrosomal amplification. In response to hydroxyurea, CHK1 was delocalized from the centrosome by RRM1 depletion. Moreover, CDK1, which functions in centrosome separation and is inhibited by CHK1, was found to be essential for RRMI1-depletion-induced centrosomal amplification. Thus, we herein demonstrate that RRM1 preserves chromosomal stability via the CHK1- and CDK1-dependent stabilization of the centrosomal integrity at the replication stage.


Assuntos
Proteína Quinase CDC2/metabolismo , Centrossomo/fisiologia , Replicação do DNA/fisiologia , Proteínas Quinases/metabolismo , Proteínas Supressoras de Tumor/genética , Proteína Quinase CDC2/genética , Linhagem Celular Tumoral , Centrossomo/metabolismo , Quinase 1 do Ponto de Checagem , Ciclina A/metabolismo , Ciclina B/metabolismo , Dano ao DNA , Replicação do DNA/genética , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Proteínas Quinases/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Ribonucleosídeo Difosfato Redutase , Transdução de Sinais , Transfecção , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/metabolismo
9.
Biochem Biophys Res Commun ; 443(3): 796-801, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24333416

RESUMO

Hyaluronan synthase 2 (HAS2), a synthetic enzyme for hyaluronan, regulates various aspects of cancer progression, including migration, invasion and angiogenesis. However, the possible association of HAS2 with the response of cancer cells to anticancer radiotherapy, has not yet been elucidated. Here, we show that HAS2 knockdown potentiates irradiation-induced DNA damage and apoptosis in cancer cells. Upon exposure to radiation, all of the tested human cancer cell lines exhibited marked (up to 10-fold) up-regulation of HAS2 within 24h. Inhibition of HAS2 induction significantly reduced the survival of irradiated radioresistant and -sensitive cells. Interestingly, HAS2 depletion rendered the cells to sustain irradiation-induced DNA damage, thereby leading to an increase of apoptotic death. These findings indicate that HAS2 knockdown sensitizes cancer cells to radiation via persistent DNA damage, further suggesting that the irradiation-induced up-regulation of HAS2 contributes to the radioresistance of cancer cells. Thus, HAS2 could potentially be targeted for therapeutic interventions aimed at radiosensitizing cancer cells.


Assuntos
Dano ao DNA , Glucuronosiltransferase/biossíntese , Tolerância a Radiação , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Indução Enzimática/efeitos da radiação , Raios gama , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Técnicas de Silenciamento de Genes , Glucuronosiltransferase/genética , Histonas/metabolismo , Humanos , Hialuronan Sintases , Regulação para Cima/genética , Regulação para Cima/efeitos da radiação
10.
Biochem Biophys Res Commun ; 441(4): 831-7, 2013 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-24211209

RESUMO

ß-Transducin repeat-containing protein (ß-TrCP), an E3 ligase, promotes the degradation of substrate proteins in response to various stimuli. Even though several ß-TrCP substrates have been identified to date, limited information of its upstream regulators is available. Here, we showed that SIRT1 suppresses ß-TrCP protein synthesis via post-translational degradation. SIRT1 depletion led to a significant increase in the ß-TrCP accumulation without affecting the mRNA level. Consistently, ß-TrCP protein accumulation induced by resveratrol was further enhanced upon SIRT1 depletion. Rescue of SIRT1 reversed the effect of resveratrol, leading to reduced ß-TrCP protein levels. Proteasomal inhibition led to recovery of ß-TrCP in cells with SIRT1 overexpression. Notably, the recovered ß-TrCP colocalized mostly with SIRT1. Thus, SIRT1 acts as a negative regulator of ß-TrCP synthesis via promoting protein degradation.


Assuntos
Sirtuína 1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Glucose/deficiência , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Biossíntese de Proteínas , Proteólise , RNA Mensageiro/metabolismo , Resveratrol , Sirtuína 1/genética , Estilbenos/farmacologia , Transcrição Gênica
11.
Biochem Biophys Res Commun ; 424(4): 681-6, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22789853

RESUMO

Upon apoptotic stimulation, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a cytosolic enzyme normally active in glycolysis, translocates into the nucleus and activates an apoptotic cascade therein. In the present work, we show that SIRT1 prevents nuclear translocation of GAPDH via interaction with GAPDH. SIRT1 depletion triggered nuclear translocation of cytosolic GAPDH even in the absence of apoptotic stress. Such translocation was not, however, observed when SIRT1 enzymatic activity was inhibited, indicating that SIRT1 protein per se, rather than the deacetylase activity of the protein, is required to inhibit GAPDH translocation. Upon irradiation, SIRT1 prevented irradiation-induced nuclear translocation of GAPDH, accompanied by interaction of SIRT1 and GAPDH. Thus, SIRT1 functions to retain GAPDH in the cytosol, protecting the enzyme from nuclear translocation via interaction with these two proteins. This serves as a mechanism whereby SIRT1 regulates cell survival upon induction of apoptotic stress by means that include irradiation.


Assuntos
Apoptose/fisiologia , Núcleo Celular/enzimologia , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Tolerância a Radiação , Sirtuína 3/metabolismo , Transporte Ativo do Núcleo Celular , Apoptose/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Citosol/enzimologia , Células HeLa , Humanos , RNA Interferente Pequeno/genética , Sirtuína 3/genética
12.
Int J Oncol ; 40(1): 277-86, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21887464

RESUMO

Proteins involved in the G1 phase of the cell cycle are aberrantly expressed, sometimes in mutated forms, in human cancers including human hepatocellular carcinoma. Upon attack by a DNA-damaging anticancer drug, a cell arrests at the G1 phase; this is a safety feature prohibiting entry of DNA-damaged cells into S-phase. p21WAF1/CIP1 prevents damaged cells from progressing to the next cell cycle. Here, we show that, in response to mitomycin C and doxorubicin, human hepatocellular carcinoma cells generate conflicting signals, mediated by cyclin E and p21WAF1/CIP1, which respectively accelerates and represses cell cycle transition. Exposure to these anticancer drugs led to rapid accumulation of cyclin E in both p53-proficient HepG2 and p53-deficient Hep3B cells. Such anticancer drug-induced cyclin E accumulation influenced the G1-S-phase transition, but not DNA fragmentation-mediated death. In p53-proficient HepG2 cells, accumulation of cyclin E was followed by an increase in the level of p53-dependent p21WAF1/CIP1, thereby inhibiting further the G1-S-phase transition. Sublethal drug concentrations also induced rapid accumulation of cyclin E, but p21WAF1/CIP1 accumulation was delayed, further facilitating the G1-S-phase transition. Eventually, most cells arrested in G2/M. Thus, mitomycin C- or doxorubicin-induced conflicting signals, mediated by cyclin E and p21WAF1/CIP1, are in play in human hepatocellular carcinoma cells. Damaged G1 cells either immediately enter S-phase, or do not do so at all, depending on the extent of DNA damage.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Ciclina E/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Doxorrubicina/administração & dosagem , Fase G1/efeitos dos fármacos , Células Hep G2 , Humanos , Mitomicina/administração & dosagem , Fase S/efeitos dos fármacos , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo
13.
BMB Rep ; 42(10): 691-6, 2009 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-19874716

RESUMO

The E2F gene family appears to regulate the proliferation and differentiation of events that are required for adipogenesis. Pref-1 is a transmembrane protein that inhibits adipocyte differentiation in 3T3-L1 cells. In this study, we found that the expression of pref-1 is regulated by the transcription factor E2F1. The expression of pref-1 and E2F1 was strongly induced in preadipocytes and at the late differentiation stage. Using luciferase reporter assay, ChIP assay and EMSA, we found that the -211/-194 region of the pref-1 promoter is essential for the binding of E2F1 as well as E2F1-dependent transcriptional activation. Knockdown of E2F1 reduced both pref-1 promoter activity and the level of pref-1 mRNA. Taken together, our data suggest that transcriptional activation of pref-1 is stimulated by E2F1 protein in adipocytes.


Assuntos
Fator de Transcrição E2F1/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Ativação Transcricional/genética , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Sequência de Bases , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Fator de Transcrição E2F1/genética , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
J Biochem Mol Biol ; 40(2): 212-7, 2007 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-17394771

RESUMO

The chicken oviduct is a dynamic organ that produces secretory proteins such as ovalbumin during the laying period. In this study, we identified oviduct-specific proteins in hens during the egg-laying period by proteomic analysis. Proteins extracted from the magnum of hens of different ages (5, 35, and 65 weeks) were analyzed by two-dimensional gel electrophoresis to compare the intensity of proteins among samples. Approximately 300 spots were detected on each gel. Based on the comparison of image gels, we found that the intensity of eight spots in 35-week magnums was increased at least by 2-fold compared with the others. Five of the eight spots were identified as calumenin, acidic ribosomal phosphoproteins (ARP), prohibitin, heart fatty acid-binding protein, and anterior gradient-2 (AGR-2). In particular, ARP and AGR-2 were highly expressed in 35- week magnums compared with 5- and 65-week magnums. In addition, the level of these proteins was consistent with their RNA levels. Expression of AGR-2 mRNA was detected in the mature magnum, whereas no signal was observed in premature tissue. Among various tissues, expression of AGR-2 mRNA was highest in the magnum, high in the isthmus, and five fold lower in muscle. It was undetectable in the liver and in other tissues (heart and kidney). However, the mRNA levels of other proteins were ubiquitous among tissues. In transcriptional activity of AGR-2, a 3.0 kb fragment of promoter region containing potential estrogen receptor binding sites had enhanced its activity strongly. In conclusion, these results suggest that AGR-2 has functional regulatory roles in the chicken oviduct during the egglaying period.


Assuntos
Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Galinhas/metabolismo , Oviductos/metabolismo , Oviposição/fisiologia , Animais , Eletroforese em Gel Bidimensional , Estrogênios/farmacologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Oviductos/efeitos dos fármacos , Oviposição/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...