Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Food Sci Biotechnol ; 32(5): 723-727, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37009039

RESUMEN

[This corrects the article DOI: 10.1007/s10068-022-01118-8.].

2.
Int J Biol Macromol ; 227: 839-850, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36563804

RESUMEN

Obesity is a global epidemic, it can induce glucose and lipid metabolism disorder and non-alcoholic fatty liver disease (NAFLD). This study explored a new way to control weight and improve fatty liver, namely, living in hypoxia environment and supplement with lactoferrin (Lf). Sixty male C57BL/6J mice were divided into six groups, namely, control, hypoxia, high-fat diet, hypoxia + high-fat diet, hypoxia + high-fat diet + low dose Lf intervention, and hypoxia + high-fat diet + high-dose Lf intervention. Mice in the hypoxia treatment groups were treated with approximately 11.5 % oxygen for 6 h every day for 8 weeks. Results showed that interventions combining Lf and hypoxia treatments showed better effect against obesity and NAFLD than hypoxia treatment alone. The interventions controlled weight gain in mice, improved glucolipid metabolism in mice. The combination intervention reduced cholesterol absorption by reducing the level of hydrophobic bile acids, and elevating the level of hydrophilic bile acids. Gut microbiota analysis revealed that the combination intervention considerably elevated short chain fatty acids (SCFAs)-producing bacteria level, and reduced the Desulfovibrionaceae_unclassified level. Thus, Lf combined with hypoxia intervention effectively prevents obesity and NAFLD by restoring gut microbiota composition and bile acid profile.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Masculino , Ratones , Ácidos y Sales Biliares/metabolismo , Dieta Alta en Grasa , Hipoxia/complicaciones , Lactoferrina/metabolismo , Hígado , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/tratamiento farmacológico
3.
Food Sci Biotechnol ; 31(10): 1315-1323, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35992325

RESUMEN

Radiation-induced liver damage (RILD) is a spiny problem in radiotherapy or other circumstances that exposure to radiation. The need for radioprotective agent is increasing to protect liver tissue. This study aimed to explore the hepatoprotective effect of p-coumaric acid (CA) against RILD. C57BL/6 male mice were exposed to 4 Gy irradiation and administrated with CA for 4 days starting on the same day of irradiation. Mice were sacrificed to obtain blood and liver tissues on day 3.5 or 14 post irradiation, respectively. The blood and liver tissues were collected. As compared with the only irradiated group, CA supplementation improved liver morphology, decreased serum alanine aminotransferase and aspartate aminotransferase, inhibited BCL2-associated X (BAX) protein expression, and improved the mice hematopoietic function. CA at the dose of 100 mg/kg body weight showed better effect compared to the other doses. Thus, CA might possess potential to protect against RILD.

4.
Chin J Cancer ; 36(1): 11, 2017 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-28088228

RESUMEN

BACKGROUND: Ankyrin repeat and SOCS box protein 3 (ASB3) is a member of ASB family and contains ankyrin repeat sequence and SOCS box domain. Previous studies indicated that it mediates the ubiquitination and degradation of tumor necrosis factor receptor 2 and is likely involved in inflammatory responses. However, its effects on oncogenesis are unclear. This study aimed to investigate the effects of ASB3 on the growth and metastasis of colorectal cancer (CRC). METHODS: We used next-generation sequencing or Sanger sequencing to detect ASB3 mutations in CRC specimens or cell lines, and used real-time quantitative polymerase chain reaction, Western blotting, and immunohistochemical or immunofluorescence assay to determine gene expression. We evaluated cell proliferation by MTT and colony formation assays, tested cell cycle distribution by flow cytometry, and assessed cell migration and invasion by transwell and wound healing assays. We also performed nude mouse experiments to evaluate tumorigenicity and hepatic metastasis potential of tumor cells. RESULTS: We found that ASB3 gene was frequently mutated (5.3%) and down-regulated (70.4%) in CRC cases. Knockdown of endogenous ASB3 expression promoted CRC cell proliferation, migration, and invasion in vitro and facilitated tumorigenicity and hepatic metastasis in vivo. Conversely, the ectopic overexpression of wild-type ASB3, but not that of ASB3 mutants that occurred in clinical CRC tissues, inhibited tumor growth and metastasis. Further analysis showed that ASB3 inhibited CRC metastasis likely by retarding epithelial-mesenchymal transition, which was characterized by the up-regulation of ß-catenin and E-cadherin and the down-regulation of transcription factor 8, N-cadherin, and vimentin. CONCLUSION: ASB3 dysfunction resulted from gene mutations or down-regulated expression frequently exists in CRC and likely plays a key role in the pathogenesis and progression of CRC.


Asunto(s)
Neoplasias Colorrectales , Proteínas Supresoras de la Señalización de Citocinas/genética , Animales , Pueblo Asiatico/genética , Ciclo Celular , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Endogámicos BALB C , Mutación , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Cicatrización de Heridas
5.
Oncoimmunology ; 7(1): e1373234, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29296528

RESUMEN

Interleukin (IL)-17 has been reported to play a controversial role in tumor immunity. Our previous studies showed that infiltration of IL-17-producing cells in esophageal squamous cell carcinoma (ESCC) induced tumor protective immunity by recruiting CD8+T lymphocytes, natural killer (NK) cells, and B lymphocytes into the tumor microenvironment. However, the mechanism of IL-17 regulation of tumor-associated neutrophils remains elusive in ESCC. In this study, we therefore evaluated the accumulation of myeloperoxidase (MPO)+ neutrophils and its association with IL-17-producing cells within ESCC tumor nests. We also investigated the effects of IL-17 on the recruitment and antitumor activity of neutrophils. MPO+ neutrophil infiltration was found to predict a favorable prognosis in ESCC patients and was positively correlated with IL-17+ cell density. IL-17 stimulated ESCC tumor cells to release more of the CXC chemokines CXCL2 and CXCL3, which are involved in neutrophil migration. Furthermore, IL-17 potentiates the direct killing capability of neutrophils by enhancing the production of cytotoxic molecules, including reactive oxygen species (ROS), MPO, TNF-related apoptosis-inducing ligand (TRAIL), and IFN-γ. Experiments in mice suggested that IL-17 alone might not affect tumor progression in the tumor-bearing host, but IL-17 can inhibit tumor growth by promoting beneficial neutrophil infiltration and activation at tumor sites. As emerging evidence indicates that targeting tumor-associated neutrophils is a strategy for antitumor therapy, our findings reveal a positive contribution of IL-17 to the modulation of neutrophil-mediated antitumor immunity in ESCC. This study provides further understanding of the mechanisms that selectively regulate functional activities of neutrophils, which may be critical for developing new tumor immunotherapy.

6.
Biochem Biophys Res Commun ; 442(3-4): 171-6, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24269822

RESUMEN

A replication-deficient adenovirus (Ad) encoding secreted human endostatin (Ad-Endo) has been demonstrated to have promising antiangiogenic and antitumoral effects. The E1B55k-deleted Ad H101 can selectively lyse cancer cells. In this study, we explored the antitumor effects and cross-interactions of Ad-Endo and H101 on nasopharyngeal carcinoma (NPC). The results showed that H101 dramatically promoted endostatin expression by Ad-Endo via rescuing Ad-Endo replication in NPC cells, and the expressed endostatin proteins significantly inhibited the proliferation of human umbilical vein endothelial cells. E1A and E1B19k products are required for the rescuing of H101 to Ad-Endo replication in CNE-1 and CNE-2 cells, but not in C666-1 cells. On the other hand, Ad-Endo enhanced the cytotoxicity of H101 by enhancing Ad replication in NPC cells. The combination of H101 and Ad-Endo significantly inhibited CNE-2 xenografts growth through the increased endostatin expression and Ad replication. These findings indicate that the combination of Ad-Endo gene therapy and oncolytic Ad therapeutics could be promising in comprehensive treatment of NPC.


Asunto(s)
Adenoviridae/fisiología , Endostatinas/genética , Terapia Genética/métodos , Neoplasias Nasofaríngeas/terapia , Neovascularización Patológica/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Replicación Viral , Adenoviridae/genética , Animales , Carcinoma , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/irrigación sanguínea , Virus Oncolíticos/genética , Proteínas Recombinantes/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Transl Med ; 11: 257, 2013 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-24124726

RESUMEN

BACKGROUND: Gene therapy using a recombinant adenovirus (Ad) encoding secretory human endostatin (Ad-Endo) has been demonstrated to be a promising antiangiogenesis and antitumor strategy of in animal models and clinical trials. The E1B55KD-deficient Ad dl1520 was also found to replicate selectively in and destroy cancer cells. In this study, we aimed to investigate the antitumor effects of antiangiogenic agent Ad-Endo combined with the oncolytic Ad dl1520 on gastric cancer (GC) in vitro and in vivo and determine the mechanisms of these effects. METHODS: The Ad DNA copy number was determined by real-time PCR, and gene expression was assessed by ELISA, Western blotting or immunohistochemistry. The anti-proliferation effect (cytotoxicity) of Ad was assessed using the colorimetry-based MTT cell viability assay. The antitumor effects were evaluated in BALB/c nude mice carrying SGC-7901 GC xenografts. The microvessel density and Ad replication in tumor tissue were evaluated by checking the expression of CD34 and hexon proteins, respectively. RESULTS: dl1520 replicated selectively in GC cells harboring an abnormal p53 pathway, including p53 mutation and the loss of p14(ARF) expression, but did not in normal epithelial cells. In cultured GC cells, dl1520 rescued Ad-Endo replication, and dramatically promoted endostatin expression by Ad-Endo in a dose- and time-dependent manner. In turn, the addition of Ad-Endo enhanced the inhibitory effect of dl1520 on the proliferation of GC cells. The transgenic expression of Ad5 E1A and E1B19K simulated the rescue effect of dl1520 supporting Ad-Endo replication in GC cells. In the nude mouse xenograft model, the combined treatment with dl1520 and Ad-Endo significantly inhibited tumor angiogenesis and the growth of GC xenografts through the increased endostatin expression and oncolytic effects. CONCLUSIONS: Ad-Endo combined with dl1520 has more antitumor efficacy against GC than Ad-Endo or dl1520 alone. These findings indicate that the combination of Ad-mediated antiangiogenic gene therapy and oncolytic Ad therapeutics could be one of promising comprehensive treatment strategies for GC.


Asunto(s)
Adenoviridae/metabolismo , Antineoplásicos/uso terapéutico , Endostatinas/uso terapéutico , Recombinación Genética/genética , Neoplasias Gástricas/tratamiento farmacológico , Proteínas Virales/metabolismo , Adenoviridae/efectos de los fármacos , Proteínas E1B de Adenovirus/metabolismo , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Endostatinas/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/patología , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/metabolismo , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Transl Med ; 10: 256, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23272637

RESUMEN

BACKGROUND: Interferon-γ (IFN-γ) is regarded as a potent antitumor agent, but its clinical application is limited by its short half-life and significant side effects. In this paper, we tried to develop IFN-γ gene therapy by a replication defective adenovirus encoding the human IFN-γ (Ad-IFNγ), and evaluate the antitumoral effects of Ad-IFNγ on nasopharyngeal carcinoma (NPC) cell lines in vitro and in xenografts model. METHODS: The mRNA levels of human IFN-γ in Ad-IFNγ-infected NPC cells were detected by reverse transcription-polymerase chain reaction (RT-PCR), and IFN-γ protein concentrations were measured by enzyme-linked immunosorbent assay (ELISA) in the culture supernatants of NPC cells and tumor tissues and bloods of nude mice treated with Ad-IFNγ. The effects of Ad-IFNγ on NPC cell proliferation was determined using MTT assay, cell cycle distribution was determined by flow cytometry analysis for DNA content, and cells apoptosis were analyzed by Annexin V-FITC/7-AAD binding assay and hoechst 33342/PI double staining. The anti-tumor effects and toxicity of Ad-IFNγ were evaluated in BALB/c nude mice carrying NPC xenografts. RESULTS: The results demonstrated that Ad-IFNγ efficiently expressed human IFN-γ protein in NPC cell lines in vitro and in vivo. Ad-IFNγ infection resulted in antiproliferative effects on NPC cells by inducing G1 phase arrest and cell apoptosis. Intratumoral administration of Ad-IFNγ significantly inhibited the growth of CNE-2 and C666-1 cell xenografts in nude mice, while no significant toxicity was observed. CONCLUSIONS: These findings indicate IFN-γ gene therapy mediated by replication defective adenoviral vector is likely a promising approach in the treatment of nasopharyngeal carcinoma.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Terapia Genética , Interferón gamma/genética , Interferón gamma/uso terapéutico , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/terapia , Animales , Apoptosis , Carcinoma , Proliferación Celular , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Ratones , Ratones Desnudos , Carcinoma Nasofaríngeo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Leuk Res ; 33(11): 1512-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19394692

RESUMEN

ZD6474 is an orally available, small-molecule tyrosine kinase inhibitor. This study explores the effect of ZD6474 on imatinib-resistant K562 cell lines, which show markedly increased SRC family kinases (SFKs) activity. ZD6474 induces growth arrest and apoptosis of imatinib-resistant and parental K562 cells, as well as inhibition of Src activity and its downstream effectors, the anti-apoptotic Bcl-2 family. ZD6474 treatment also inhibits the activity of STAT3 and reactivation of its activity results in suppression of the anti-tumor effects of SFKs inhibitors. A single oral administration of ZD6474 produced dose-dependent inhibition of imatinib-resistant K562 cells xenograft tumors. These results suggest that clinical assessment of ZD6474 against imatinib-resistant CML is warranted.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos , Piperazinas/farmacología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Quinazolinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Secuencia de Bases , Benzamidas , Ciclo Celular/efectos de los fármacos , Cartilla de ADN , Femenino , Humanos , Mesilato de Imatinib , Inmunohistoquímica , Células K562 , Trasplante Heterólogo
10.
Life Sci ; 81(9): 695-701, 2007 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-17714738

RESUMEN

Interferon gamma (IFNgamma) is regarded as a potent antitumor agent, but therapy with IFNgamma is hampered by its short half-life and significant side effects. We developed a replication defective adenovirus carrying the human IFNgamma gene and evaluated the effects of adenovirus-mediated IFNgamma (Ad-IFNgamma) gene transfer on human prostate cancer cell lines in vitro and on xenografts in vivo. Our results showed infection of prostate cancer cells with Ad-IFNgamma led to production of an active cytokine and resulted in an antiproliferative effect on the prostate cancer cells. Intratumoral injection of Ad-IFNgamma significantly inhibited the growth of DU-145 cell xenografts in vivo, while no significant toxicity effect was observed. RT-PCR analysis indicated transgene expression mainly enriched in tumors in vivo, and slightly distributed in livers. These findings suggest adenovirus-mediated IFNgamma gene transfer is a promising approach in the treatment of advanced prostate cancer.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Interferón gamma/genética , Neoplasias de la Próstata/terapia , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Interferón gamma/biosíntesis , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Immunol Immunother ; 56(11): 1831-43, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17503043

RESUMEN

BACKGROUND AND OBJECTIVE: [corrected] Dendritic cells play an important role in initiation and regulation of immune responses. Previous studies demonstrated that intratumoral administration of 6Ckine-modified DCs enhanced local and systemic antitumor effects. Herein we report the investigation of the specific CTL responses elicited by adenoviral 6Ckine/IFNgamma fusion gene-modified DCs in vitro. METHODS: Human monocyte-derived DCs were modified with an adenoviral vector encoding 6Ckine/IFNgamma fusion protein (Ad-6Ckine/IFNgamma), and then investigated the effect of 6Ckine/IFNgamma fusion protein on the maturation, cytokine and chemokine secretion of DCs, and their activities of recruiting and activating T cells in vitro were investigated. RESULTS: 6Ckine/IFNgamma fusion protein induced DC maturation characterized with the upregulation of CD83 and CCR7. And it up-regulated the expression of RANTES and IL-12p70, down-regulated that of IL-10 in DCs. Additionally, 6Ckine/IFNgamma markedly increased DC's recruiting ability for naive T cells, benefiting from the enhanced expression of chemokines 6Ckine and RANTES in DCs. Fusion gene-modified DCs significantly promoted the proliferation of autologous T cells, induced Th1 differentiation by upregulating the expression of IL-2 and T-bet in T cells, and increased specific cytotoxicity of CTLs against specific tumor cells, HepG2 or LoVo cells, respectively. CONCLUSION: Combining the effects of 6Ckine and IFNgamma, Ad-6Ckine/IFNgamma modified DCs induced enhanced CTL responses in vitro, which indicated that Ad-6Ckine/IFNgamma modified DCs might be used as an adjuvant to trigger an effective antitumor immune response.


Asunto(s)
Quimiocinas CC , Células Dendríticas/inmunología , Terapia Genética , Interferón gamma/metabolismo , Linfocitos T Citotóxicos/inmunología , Adenoviridae/genética , Inhibidores de la Angiogénesis , Línea Celular Tumoral , Quimiocina CCL21 , Células Dendríticas/trasplante , Vectores Genéticos , Humanos , Interferón gamma/genética , Activación de Linfocitos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
12.
Hum Gene Ther ; 18(3): 207-21, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17346097

RESUMEN

E10A, a recombinant adenovirus type 5 vector carrying the human endostatin gene, may be a promising gene therapy drug in the treatment of solid tumors by antiangiogenesis, but a preclinical safety evaluation of E10A has not yet been performed. With high and low doses equivalent to 30 and 7.5 times the human curative dose, respectively, intramuscular injections of E10A were given once daily, 6 days/week, for 3 months, followed by a 1-month recovery period. As of 4 months, all experimental animals appeared generally healthy: normal behavior and eating habits, no nausea, vomiting, or salivation, no abnormal changes in urination or defecation, and increased body weight with the time of experiment. Urinalysis, hemogram, blood biochemistry, electrocardiogram, macroscopic and microscopic studies of organs and tissues were done before treatment, at month 3 of treatment, and 1 month posttreatment. At all time points, no significant abnormal toxic effects were noted. Preliminary investigation of E10A immunotoxicity in dogs indicated that anti-adenoviral antibodies were generated, in a dose- and time-independent manner, after E10A injection. Our data demonstrated that, long term, high-dose intramuscular administration of recombinant human endostatin-carrying adenovirus (E10A) was not notably toxic and might be safe for clinical therapeutic use, although additional long-term toxicity studies by other administration routes are still necessary.


Asunto(s)
Adenoviridae/genética , Endostatinas/genética , Terapia Genética , Vectores Genéticos/toxicidad , Animales , Análisis Químico de la Sangre , Peso Corporal/efectos de los fármacos , Defecación/efectos de los fármacos , Perros , Conducta Alimentaria/efectos de los fármacos , Femenino , Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacocinética , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunohistoquímica , Inyecciones Intramusculares , Masculino , Urinálisis , Micción/efectos de los fármacos , Vómitos/inducido químicamente
13.
Ai Zheng ; 25(9): 1069-75, 2006 Sep.
Artículo en Chino | MEDLINE | ID: mdl-16965644

RESUMEN

BACKGROUND & OBJECTIVE: T-bet (T box expressed in T cells), a Th1-specific T box transcription factor, controls many kinds of immune cells, such as Th1, NK, CD8+, dendritic cells, and B cells. This study was to explore potential effects of T-bet gene on biological functions of mouse macrophage Raw264.7 cells in vitro. METHODS: The eukaryotic expression vector carrying mouse T-bet (pcDNA3.0-mT-bet) was constructed and identified by consequence analysis, double restrictive endonucleases digestion and polymerase chain reaction (PCR). The gene expression in Raw264.7 cells was detected by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot. PBS, pcDNA3.0, and pcDNA3.0-mT-bet were transiently transfected into Raw264.7 cells respectively; cell cycle, MHC I/II expression levels, phagocytic activity of FITC-dextran, nitric oxide (NO) secretion level, and the cytotoxicity of Raw264.7 cells to mouse leukemia cell line L1210 were evaluated 48 hours after transfection. RESULTS: Eukaryotic expression vector which could express T-bet protein in Raw264.7 cells was successfully constructed. There was no difference in cell cycle between pcDNA3.0 group and pcDNA3.0-mT-bet group. There was significant difference in MHC I expression level between pcDNA3.0 group (20.8+/-0.7) and pcDNA3.0-mT-bet group (24.8+/-0.6, P<0.05), but not in MHC II expression level; there was also difference in mean fluorescence intensity of phagocytized dextran between pcDNA3.0 group (28.2+/-0.4) and pcDNA3.0-mT-bet group (32.8+/-0.8, P<0.05); there was also significant difference in NO secretion level between pcDNA3 group (0 pmol) and pcDNA3.0-mT-bet group [(1.7+/-0.6) pmol, P<0.05] without lipopolysaccharide (LPS) stimulation; meanwhile, significant difference was also observed between pcDNA3.0 group [(10.5 +/-1.3) pmol] and pcDNA3.0-mT-bet group [(15.6+/-1.6) pmol, P<0.05] under the stimulation of LPS (10 microg/ml) for 20 h; there was also difference in cytotoxicity of Raw264.7 cells to L1210 cells in vitro between pcDNA3 group [(35.6+/-2.1)%] and pcDNA3.0-mT-bet group [(51.9+/-3.5)%, P<0.05]. CONCLUSION: T-bet up-regulates MHC I expression and NO secretion level in Raw264.7 cells, increases their cytotoxicity to L1210 cells, but has no influences on the cell cycle and MHC II expression.


Asunto(s)
Genes MHC Clase I , Antígenos de Histocompatibilidad Clase I/metabolismo , Macrófagos/citología , Óxido Nítrico/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Ciclo Celular , Línea Celular Tumoral , Vectores Genéticos , Leucemia L1210/patología , Macrófagos/metabolismo , Ratones , Proteínas Nucleares/metabolismo , Fagocitosis , Plásmidos , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología , Transactivadores/metabolismo , Transfección , Regulación hacia Arriba
14.
Life Sci ; 77(12): 1331-40, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15936041

RESUMEN

Endostatin, a 20-kDa carboxyl-terminal fragment of collagen XVIII, is a potent inhibitor of endothelial cell proliferation and tumor angiogenesis. We have constructed replication-deficient recombinant adenovirus (Ad-rhE), which encoded secreted human endostatin, and our previous studies showed that Ad-rhE had a potent suppression of tumor growth in vivo. In the present study, we investigated the dynamic distribution and expression of human endostatin gene in vivo using fluorogenic real-time quantitative PCR and enzyme-linked immunosorbent assay(ELISA), respectively, with an injection of 2.0 x10(9)pfu of Ad-rhE. After injection, the Ad-rhE DNAs decreased sharply, but lasted a relative long-term at low concentration (10,000--20,000 copies/mg tissues). Whereas the expressed endostatin rose up rapidly, and reached to the top on day 5 after injection of Ad-rhE, and then decreased sharply, but endostatin in tumors sustained to over 9 days at a certain level. Both Ad-rhE DNAs and endostatin mainly enriched in tumors in vivo, and then in livers. These results suggest that endostatin gene delivered by adenoviral vector can generate a high expression in vivo, and both the metabolism pathways of Ad-rhE DNAs and endostatin in vivo are through the systems of livers.


Asunto(s)
Adenoviridae/genética , Endostatinas/genética , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Inyecciones , Hígado/metabolismo , Melanoma Experimental/metabolismo , Trasplante de Neoplasias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Ai Zheng ; 24(3): 376-84, 2005 Mar.
Artículo en Chino | MEDLINE | ID: mdl-15757547

RESUMEN

Angiogenesis is required for invasive tumor growth and metastasis. Inhibition of angiogenesis is considered to be a promising approach of antitumor therapy. Recently, many endogenous angiogenesis inhibitors have been discovered, some of them are currently in various stages of clinical trials. This review focused on the structure, function, and mechanism of endogenous angiogenesis inhibitors, and their potential in treating tumor.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/fisiología , Angiostatinas/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Endostatinas/farmacología , Células Endoteliales/citología , Humanos , Interferones/farmacología , Metaloproteinasas de la Matriz/farmacología , Neoplasias/irrigación sanguínea , Inhibidores de Serina Proteinasa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...