Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
PLoS One ; 10(5): e0124420, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25933295

RESUMEN

BACKGROUND: Effective therapeutic strategies to address intestinal complications after radiation exposure are currently lacking. Mesenchymal stem cells (MSCs), which display the ability to repair the injured intestine, have been considered as delivery vehicles for repair genes. In this study, we evaluated the therapeutic effect of hepatocyte growth factor (HGF)-gene-modified MSCs on radiation-induced intestinal injury (RIII). METHODS: Female 6- to 8-week-old mice were radiated locally at the abdomen with a single 13-Gy dose of radiation and then treated with saline control, Ad-HGF or Ad-Null-modified MSCs therapy. The transient engraftment of human MSCs was detected via real-time PCR and immunostaining. The therapeutic effects of non- and HGF-modified MSCs were evaluated via FACS to determine the lymphocyte immunophenotypes; via ELISA to measure cytokine expression; via immunostaining to determine tight junction protein expression; via PCNA staining to examine intestinal epithelial cell proliferation; and via TUNEL staining to detect intestinal epithelial cell apoptosis. RESULTS: The histopathological recovery of the radiation-injured intestine was significantly enhanced following non- or HGF-modified MSCs treatment. Importantly, the radiation-induced immunophenotypic disorders of the mesenteric lymph nodes and Peyer's patches were attenuated in both MSCs-treated groups. Treatment with HGF-modified MSCs reduced the expression and secretion of inflammatory cytokines, including tumor necrosis factor alpha (TNF-α) and interferon-gamma (IFN-γ), increased the expression of the anti-inflammatory cytokine IL-10 and the tight junction protein ZO-1, and promoted the proliferation and reduced the apoptosis of intestinal epithelial cells. CONCLUSIONS: Treatment of RIII with HGF-gene-modified MSCs reduces local inflammation and promotes the recovery of small intestinal histopathology in a mouse model. These findings might provide an effective therapeutic strategy for RIII.


Asunto(s)
Terapia Genética , Factor de Crecimiento de Hepatocito/genética , Inmunidad , Intestinos/lesiones , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Traumatismos por Radiación/terapia , Animales , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Factor de Crecimiento de Hepatocito/uso terapéutico , Humanos , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Intestinos/inmunología , Ganglios Linfáticos/patología , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados/patología , Traumatismos por Radiación/sangre , Traumatismos por Radiación/inmunología , Transducción Genética
2.
Clin Chim Acta ; 437: 183-6, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25086284

RESUMEN

BACKGROUND/AIMS: To determine whether the combination of tumor markers (CA72-4, CA125, CA19-9 and CEA) could increase the sensitivity and accuracy for in the diagnosis of gastric cancer (GC). METHODS: This study is a retrospective analysis. A total of 426 patients, including 106 patients with GC, 149 patients with benign gastric diseases and 171 healthy people, who visited Zhejiang Xiaoshan Hospital from January 2011 to December 2013, were measured by serum markers, including CA72-4, CA125, CA19-9 and CEA. Statistical analyses including area under curve (AUC) of receiver operating characteristic (ROC) curve, and logistic regression analysis, were performed to evaluate the diagnostic value of these markers on GC. RESULTS: Serum levels of CA72-4, CEA, CA125 and CA19-9 were higher in the GC group than those in the benign gastric disease group and the healthy control group (P<0.005). The sensitivities of CA72-4, CEA, CA125 and CA19-9 at the recommended cut-off level for all patients were 33.0%, 25.5%, 31.1% and 38.7%, respectively. However, when all four markers were used in combination the sensitivity increased to 66.0%. But by using an optimal cut-off value, the sensitivities of all four markers for the diagnosis of GC were improved. Especially the sensitivity of CEA increased to 73.6% and the sensitivity of the combination of the tumor markers increased to 75.5%. The age and gender had no effects on the diagnostic value of these markers. CONCLUSIONS: With the help of optimal cut-off values based on ROC curve and logistic regression analysis, the combination of these markers could improve the sensitivity for the diagnosis of GC based on common serum tumor markers.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/sangre , Biomarcadores de Tumor/sangre , Antígeno Ca-125/sangre , Antígeno CA-19-9/sangre , Antígeno Carcinoembrionario/sangre , Proteínas de la Membrana/sangre , Neoplasias Gástricas/sangre , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Detección Precoz del Cáncer/normas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Neoplasias Gástricas/diagnóstico , Adulto Joven
3.
Hum Gene Ther ; 24(3): 343-53, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23458413

RESUMEN

Abstract Effective therapeutic strategies for radiation-induced lung injury (RILI) are lacking. Mesenchymal stem cells (MSCs), as gene therapy delivery vehicles, possess the ability to repair injured lung. In this study, we conducted MSC-based hepatocyte growth factor (HGF) gene therapy for RILI. Mice received single-dose radiation with 20 Gy of γ rays locally to the lung, and then were administered normal sodium, Ad-HGF-modified MSCs, or Ad-Null-modified MSCs. Ad-HGF-modified MSCs (MSCs-HGF) improved histopathological and biochemical markers of lung injury. MSCs-HGF could reduce secretion and expression of proinflammatory cytokines, including tumor necrosis factor-α, interferon-γ, interleukin (IL)-6, and intercellular adhesion molecule-1, and increase the expression of antiinflammatory cytokine IL-10. It could also decrease expression levels of profibrosis factors transforming growth factor-ß, Col1a1 (collagen type 1, α1), and Col3a1, and inhibit fibrosis progress. MSCs-HGF could promote proliferation of lung epithelial cells and protect them from apoptosis, and improve the expression of endogenous HGF and its receptor c-Met significantly. We also found that sphingosine-1-phosphate receptor-1 expression was increased in injured lung. These results suggest MSC-based HGF gene therapy not only reduces inflammation but also inhibits lung fibrosis.


Asunto(s)
Factor de Crecimiento de Hepatocito/genética , Lesión Pulmonar/terapia , Células Madre Mesenquimatosas/metabolismo , Traumatismos Experimentales por Radiación/terapia , Adenoviridae/genética , Células Epiteliales Alveolares/metabolismo , Animales , Apoptosis , Proliferación Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Regulación de la Expresión Génica , Terapia Genética , Vectores Genéticos/genética , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Trasplante de Células Madre Mesenquimatosas , Ratones , Permeabilidad , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo
4.
Biochem Biophys Res Commun ; 410(4): 803-8, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21703232

RESUMEN

Members of the Spred gene family are negative regulators of the Ras/Raf-1/ERK pathway, which has been associated with several features of the tumor malignancy. However, the effect of Spred genes on hepatocellular carcinoma (HCC) remains uninvestigated. In the present work, we analyzed the in vitro and in vivo effects of Spred2 expression on the hepatic carcinoma cell line, SMMC-7721. In addition to attenuated ERK activation, which inhibited the proliferation and migration of unstimulated and HGF-stimulated SMMC-7721 cells. Adenovirus-mediated Spred2 overexpression induced the activation of caspase-3 and apoptosis, as well as reduced the expression level of Mcl-1. Most importantly, the knockdown of Spred2 markedly enhanced tumor growth in vivo. In conclusion, these results suggest that Spred2 could qualify as a potential therapeutic target in HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas Represoras/genética , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño/genética
5.
Hepatobiliary Pancreat Dis Int ; 10(2): 201-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21459729

RESUMEN

BACKGROUND: KAI1/CD82 has been reported to attenuate the process of metastases in a variety of tumors; however, its mechanism of action in invasion has not been fully elucidated. The present study aimed to investigate the importance of KAI1 in invasion and its correlation with activation of sphingosine kinase (SPK) in human pancreatic cancer PANC1 and Miapaca-2 cell lines. METHODS: The expression of KAI1 in PANC1 and Miapaca-2 cells, which was mediated by recombinant adenovirus (Ad-KAI1), was assessed by a flow cytometer and Western blotting. After successful infection was established, in vitro growth curve and invasive ability in Boyden Chamber assay were studied. The presence of KAI1 correlating with c-Met and SPK was detected by co-immunoprecipitation and [gamma-32P] ATP incorporation. RESULTS: KAI1 genes had no significant effects on the curve representing cell growth. After infection with the KAI1 gene, decreased invasive ability in the Boyden Chamber assay was observed in PANC1 and Miapaca-2 cells that were induced by hepatocyte growth factor. Over-expression of KAI1 in the cells led to the deactivation of SPK and a decreased level of intracellular sphingosine-1-phosphate. No correlation was observed between c-Met and KAI1 during co-immunoprecipitation. CONCLUSION: The results of this study for the first time demonstrated a regulatory role for KAI1 in SPK activation, which leads to decreased invasive ability in disease progression of human pancreatic cancer.


Asunto(s)
Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Proteína Kangai-1/fisiología , Neoplasias Pancreáticas/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Adenoviridae/genética , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Activación Enzimática , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Proteína Kangai-1/genética , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-met/fisiología , Transfección
6.
Biochem Biophys Res Commun ; 404(3): 802-8, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21168384

RESUMEN

KAI1, a metastasis-suppressor gene belonging to the tetraspanin family, is known to inhibit cancer metastasis without affecting the primary tumorigenicity by inhibiting the epidermal growth factor (EGF) signaling pathway. Recent studies have shown that hypoxic conditions of solid tumors induce high-level autophagy and KAI1 expression. However, the relationship between autophagy and KAI1 remains unclear. By using transmission electron microscopy, confocal microscopy, and Western blotting, we found that KAI1 can induce autophagy in a dose- and time-dependent manner in the human pancreatic cell line MiaPaCa-2. KAI1-induced autophagy was confirmed by the expression of autophagy-related proteins LC3 and Beclin 1. KAI1 induces autophagy through phosphorylation of extracellular signal-related kinases rather than that of AKT. KAI1-induced autophagy protects MiaPaCa-2 cells from apoptosis and proliferation inhibition partially through the downregulation of poly [adenosine diphosphate (ADP)-ribose] polymerase (PARP) cleavage and caspase-3 activation.


Asunto(s)
Autofagia , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Kangai-1/biosíntesis , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Humanos , Proteína Kangai-1/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Fosforilación , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Factor de Transcripción STAT3/metabolismo
7.
Leuk Lymphoma ; 51(10): 1902-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20795791

RESUMEN

Prokineticin-1 (PK1) has been identified as a mitogen-specific protein for the endothelium of steroidogenic glands. Here we report a novel function of PK1 in the regulation of multiple myeloma (MM) cells. PK1 activates multiple signals including mitogen-activated protein kinase (MAPK), PI3K-AKT, and Jak-STAT3, sphingosine kinase-1 (SPK1) in MM cells. Treatment of MM cells with PK1 causes a time- and dose-dependent phosphorylation of MAPK, AKT and STAT3 and upregulation of SPK1 expression and cellular activity. We also show that PK1 upregulates Mcl-1 expression in a time- and dose-dependent manner in human MM cell lines and in the cells of patients with MM. Pertussis toxin, a pan-PK1 receptor inhibitor, can block PK1-induced upregulation of Mcl-1, indicating it relates to a G-protein-coupled receptor. We also show that PK1 protects MM cells against apoptosis induced by starvation for fetal calf serum (FBS), or for FBS and IL-6. Taken together, PK1 activates multiple signaling pathways and, upregulates Mcl-1 expression, leading to proliferation and survival of MM cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/farmacología , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero/farmacología , Expresión Génica , Humanos , Inmunohistoquímica , Interleucina-6/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Fosforilación/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Células Tumorales Cultivadas , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo
8.
Biochem Biophys Res Commun ; 393(4): 637-42, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20153728

RESUMEN

Spreds, a recently established class of negative regulators of the Ras-ERK (extracellular signal-regulated kinase) pathway, are involved in hematogenesises, allergic disorders and tumourigenesis. However, their role in hematologic neoplasms is largely unknown. Possible effects of Spreds on other signal pathways closely related to Ras-ERK have been poorly investigated. In this study, we investigated the in vitro effects of Spred2 on chronic myeloid leukemia (CML) cells. In addition to inhibiting the well-established Ras-ERK cascade, adenovirus-mediated Spred2 over-expression inhibits constitutive and stem cell factor (SCF)-stimulated sphingosine kinase-1 (SPHK1) and Mcl-1 expression, as well as inhibiting proliferation and inducing apoptosis in CML cells. In K562 cells and primary CML cells, imatinib induces endogenous Spred2 expression. Spred2 silencing by stable RNA interference partly protects K562 cells against imatinib-induced apoptosis. Together, these data implicate Spred2 in imatinib-induced cytotoxicity in CML cells, possibly by inhibiting the Ras-ERK cascade and the pro-survival signaling molecules SPHK1 and Mcl-1. These findings reveal potential targets for selective therapy of CML.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Proteínas Represoras/metabolismo , Benzamidas , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Silenciador del Gen , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Represoras/genética
9.
Acta Pharmacol Sin ; 30(5): 617-27, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19363518

RESUMEN

AIM: Oncolytic adenovirus, also called conditionally replicating adenovirus (CRAD), can selectively propagate in tumor cells and cause cell lysis. The released viral progeny can infect neighboring cancer cells, initiating a cascade that can lead to the ultimate destruction of the tumor. Suicide gene therapy using herpes simplex virus thymidine kinase (HSV-TK) and ganciclovir (GCV) offers a potential treatment strategy for cancer and is undergoing preclinical trials for a variety of tumors. We hypothesized that HSV-TK gene therapy combined with oncolytic adenoviral therapy would have an enhanced effect compared with the individual effects of the therapies and is a potential novel therapeutic strategy to treat liver cancer. METHODS: To address our hypothesis, a novel CRAD was created, which consisted of a telomerase-dependent oncolytic adenovirus engineered to express E1A and HSV-TK genes (Ad-ETK). The combined effect of Ad-ETK and GCV was assessed both in vitro and in vivo in nude mice bearing HepG2 cell-derived tumors. Expression of the therapeutic genes by the transduced tumor cells was analyzed by RT-PCR and Western blotting. RESULTS: We confirmed that Ad-ETK had antitumorigenic effects on human hepatocellular carcinoma (HCC) both in vitro and in vivo, and the TK/GCV system enhanced oncolytic adenoviral therapy. We confirmed that both E1A and HSV-TK genes were expressed in vivo. CONCLUSION: The Ad-ETK construct should provide a relatively safe and selective approach to killing cancer cells and should be investigated as an adjuvant therapy for hepatocellular carcinoma.


Asunto(s)
Adenoviridae/genética , Carcinoma Hepatocelular/terapia , Ganciclovir/administración & dosificación , Terapia Genética/métodos , Neoplasias Hepáticas Experimentales/terapia , Viroterapia Oncolítica/métodos , Timidina Quinasa/genética , Animales , Línea Celular Tumoral , Terapia Combinada , Genes Transgénicos Suicidas , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Simplexvirus/genética
10.
Sheng Wu Gong Cheng Xue Bao ; 23(5): 915-8, 2007 Sep.
Artículo en Chino | MEDLINE | ID: mdl-18051875

RESUMEN

Adenovirus vectors are one of the most promising gene transfer systems. They are of great value for gene therapy because these vectors achieve temporal high-level transgene expression and high gene transfer efficiency. To meet increasing needs of adenovirus vectors for gene therapy programs, parallel development of efficient, scalable and reproducible production processes is required. Perfusion cultivation of 293 cells is one of the most commonly used methods to produce adenovirus vectors and it is suitable for industrialized production specially. Experimental studies had been carried out to produce recombinant adenovirus containing the green fluorescent protein gene (Ad-GFP) by perfusion cultivation of HEK-293 N3S cells in a 5L stirring bioreactors. Perfusion rate was 1-2 volume/day. To infect the 293 N3S cells with Ad-GFP at the density of (2-4) x 10(6) cells/ ml. The time of collecting cells was 48 hours post infection. After three rounds of freeze/thaw and centrifugation, the crude viral lysates were stored at--80 degrees C until use. Then to get the Ad-GFP products by 2 x CsCl-gradient purification. The purity of the products was determined by the A260/A280 ratio and a high performance liquid chromatography (HPLC) assay. The infective titer was determined by a TCID50 assay. The culture term was 10-12 days. The infectious titer, the number of virus particle and the ratio of infectious titer to virus particle for the product were 1.0 x 10(11) IU/mL, 1.68 x 10(12) VP/mL and 6.0% IU/VP respectively. The A260/A280 ratio was 1.33, and the purity determined by HPLC was 99.2%. The cell specific productivity was around 1000 IU/cell. By perfusion cultivation of 293 N3S cells in a 5L stirring bioreactors, we established the production process for Ad-GFP, which paves a way to produce other recombinant adenovirus for gene therapy.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Riñón/citología , Recombinación Genética , Adenoviridae/crecimiento & desarrollo , Adenoviridae/aislamiento & purificación , Reactores Biológicos/microbiología , Línea Celular , Técnicas de Transferencia de Gen , Humanos , Riñón/virología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Cultivo de Virus/instrumentación , Cultivo de Virus/métodos
11.
Hum Gene Ther ; 18(11): 1119-28, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17939750

RESUMEN

Sphingosine kinase 1 (SPK1) has been identified as a central mediator of ischemia preconditioning and plays a protective role in ischemia/reperfusion (I/R)-induced cardiomyocyte death. In the present study, we investigated the protective effect of adenovirus-mediated SPK1 gene (Ad-SPK1) transfer on I/R-induced cardiac injury, and evaluated its therapeutic action on postinfarction heart failure. Cardiac SPK1 activity was increased about 5-fold by injection of Ad-SPK1, compared with injection of adenovirus carrying the green fluorescent protein gene (Ad-GFP). A more potent performance and a lower incidence of arrhythmia were observed in Ad-SPK1-injected hearts during the reperfusion period, compared with Ad-GFP-injected hearts. An enzymatic activity assay showed that creatine kinase release was also less in Ad-SPK1-injected hearts. To investigate the therapeutic action of the SPK1 gene on postischemic heart failure, the left anterior descending branch of the coronary artery in Wistar rats was ligated after direct intramyocardial injection of Ad-SPK1 or Ad-GFP as a control. Ad-SPK1 injection significantly preserved cardiac systolic and diastolic function, as evidenced by left ventricular (LV) systolic pressure, LV end-diastolic pressure, and peak velocity of contraction (dP/dt). The LV morphometric parameters of Ad-SPK1-treated animals were also preserved. In addition, SPK1 gene delivery significantly enhanced angiogenesis and reduced fibrosis. These results demonstrate that adenovirus-mediated SPK1 gene transfer could efficiently prevent I/R-induced myocardial injury and attenuate postischemic heart failure. Thus, SPK1 gene delivery would be a novel strategy for the treatment of coronary heart disease.


Asunto(s)
Adenoviridae , Vectores Genéticos , Daño por Reperfusión Miocárdica/prevención & control , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Adenoviridae/genética , Animales , Arritmias Cardíacas/prevención & control , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Fibrosis/prevención & control , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Contracción Miocárdica/genética , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/terapia , Miocardio/enzimología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/virología , Neovascularización Fisiológica/genética , Técnicas de Cultivo de Órganos , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/uso terapéutico , Ratas , Ratas Wistar , Recuperación de la Función/genética
12.
Br J Haematol ; 138(5): 632-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17686057

RESUMEN

Interleukin 6 (IL-6) influences the growth and survival of multiple myeloma (MM) cells via the activation of multiple signalling cascades. Although sphingosine kinase (SPHK) signalling is known to play important roles in the regulation of cell proliferation and apoptosis, the role of SPHK activation in IL-6 signalling and in the pathology of MM remains unclear. This study found that IL-6 activated SPHK in MM cells, which mediates the suppressive effects of IL-6 on MM cell apoptosis. Both MM cell lines and primary MM cells constitutively expressed SPHK, and treatment of MM cells with IL-6 resulted in activation of SPHK in a concentration-dependent manner. Specific inhibitors of the phosphatidylinositol-3 kinase and extracellular signal-regulated kinase/mitogen-activated protein kinase pathways blocked the IL-6-induced activation of SPHK. It was further demonstrated that IL-6-induced activation of SPHK inhibited dexamethasone-induced apoptosis of MM cells. IL-6 stimulation or retroviral-mediated overexpression of SPHK1 in MM cells resulted in increased intracellular SPHK activity and upregulation of myeloid cell leukaemia-1 (Mcl-1), leading to increased cell proliferation and survival. Conversely, inhibition of SPHK1 by small interfering RNA reduced IL-6-induced upregulation of Mcl-1 and blocked the suppressive effect of IL-6 on MM cell apoptosis. Taken together, these results delineate a key role for SPHK activation in IL-6-induced proliferation and survival of MM cells, and suggest that SPHK may be a potential new therapeutic target in MM.


Asunto(s)
Apoptosis/efectos de los fármacos , Activación Enzimática , Interleucina-6/farmacología , Mieloma Múltiple/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proliferación Celular , Dexametasona/farmacología , Glucocorticoides/farmacología , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Mieloma Múltiple/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
13.
Artículo en Chino | MEDLINE | ID: mdl-17374184

RESUMEN

OBJECTIVE: To study the pulmonary toxicity to rats induced by the nanosized SiO(2) or the standard SiO(2). METHODS: Seventy-two male SD rats were divided into three groups: the nanosized SiO(2) group, the standard SiO(2) group and the control group. 24 rats each group. The nanosized SiO(2) group and the standard SiO(2) group were instilled intratracheally with 0.5 ml suspension of 0.6 mg/ml nanosized SiO(2) or standard SiO(2) respectively while the control group was instilled with 0.5 ml physiological saline. On the 3rd, 7th, 14th, and 28th day after exposure, six rats were sacrificed at each time point and the total white cells counts and total protein in BALF and the histopathological changes were observed. The pulmonary toxicities of the two SiO(2) dusts were compared. RESULTS: Nanosized SiO(2) caused significant increase at 3rd, 7th, 14th day after the exposure [(16.0 +/- 6.0) x 10(6), (11.1 +/- 4.0) x 10(6), (12.2 +/- 4.6) x 10(6)] compared with saline (P < 0.05 or P < 0.01) in the total numbers of white cells and on the 3rd after the exposure compared with standard SiO(2) [(5.7 +/- 3.7) x 10(6), P < 0.01]. Meanwhile, Nanosized SiO(2) significantly increased the total protein on the 14th, 28th day after the exposure (0.41 +/- 0.14, 0.41 +/- 0.19 g/L) compared with saline or standard SiO(2) and nanosized SiO(2) on the 3rd, 7th day after the exposure (P < 0.05 or P < 0.01). Nanosized SiO(2)-treated rats showed marked white cell infiltration in alveolar space or around brondum the blood vessel. Standard SiO(2) caused similar but less severe responses compared with nanosized SiO(2). Van Gieson's-stained sections showed no significant fibrosis in these dust-exposed rats at 28th day after the exposure. CONCLUSION: Nanosized SiO(2) can cause severer and longer pulmonary toxicity in rats than standard SiO(2). The pulmonary particle load threshold of nanosized SiO(2) may be lower than that of standard SiO(2).


Asunto(s)
Nanopartículas/toxicidad , Fibrosis Pulmonar/inducido químicamente , Dióxido de Silicio/toxicidad , Animales , Masculino , Tamaño de la Partícula , Ratas , Ratas Sprague-Dawley
14.
Exp Hematol ; 34(9): 1171-82, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16939810

RESUMEN

OBJECTIVE: Adenoviral vectors (Ad) were widely used in gene therapy and study of gene function, but the commonly used serotype 5 adenovirus-based vectors (Ad5) could poorly transduce hematopoietic cells because of low expression of viral receptors on these cells. To overcome this limitation, we developed a retargeting adenovector with a chimeric fiber of Ad5 and Ad11p (Ad5F11p) and evaluated its gene transfer ability in hematopoietic cells. MATERIALS AND METHODS: An Ad11p fiber pseudotyped Ad5 vector was generated by modifying the fiber gene of pAdEasy-1 backbone plasmid. Ad5F11p-GFP encoding enhanced green fluorescence protein (GFP) gene was transferred into human leukemic cell lines, primary leukemic cells, and CD34(+) hematopoietic cells. The gene transduction efficiency was determined by fluorescence-activated cell sorting assay. RESULTS: More than 90% of U937 or K562 cells could be infected by Ad5F11p-GFP at a moderate multiplicity of infection (MOI). Ad5F11p-GFP is also significantly more effective than control Ad5-GFP in infection of primary myeloid leukemic cells. At 200 MOI, GFP-positive percentages of Ad5F11p-GFP transduced myeloid leukemic cells range from 10.58% to 92.63% with a median of 28.65%. Ad5F11p-GFP could transduce about 50% human hematopoietic stem/progenitor (CD34(+)) cells, while Ad5-GFP could transduce <15% at 200 MOI. CD46 was reported to be the receptor of Ad11p. Our data suggest that CD46 participates in the process of Ad5F11p-GFP infection but is not the unique molecule determining its gene transfer efficiency of host cells. CONCLUSION: We established a retargeting adenovector system, which could infect hematopoietic cells effectively and would benefit research work on Ad tropism.


Asunto(s)
Adenoviridae , Antígenos CD34 , Terapia Genética , Células Madre Hematopoyéticas , Transducción Genética , Transgenes , Línea Celular Tumoral , Terapia Genética/métodos , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Especificidad de la Especie , Transducción Genética/métodos
15.
Cell Signal ; 18(11): 2049-55, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16765027

RESUMEN

Shp-2, a ubiquitously expressed protein tyrosine phosphatase containing two Src homology 2 domains, plays an important role in integrating signaling from the cell surface receptors to intracellular signaling mechanisms. Previous studies have demonstrated that the Shp-2 is involved in hepatocyte growth factor (HGF)-induced cell scattering. Here we report that Shp-2 is required for the HGF-induced activation of sphingosine kinase-1 (SPK1), a highly conserved lipid kinase that plays an important role in cell migration. Loss-of-function mutation of Shp-2 did not affect the expression of SPK1, but resulted in its inactivation and the blockage of HGF-induced migration in embryonic fibroblasts. Reintroduction of functional wild type (WT) Shp-2 into the mutant cells partially restored SPK1 activation, and overexpression of SPK1 in these mutant cells enhanced HGF-induced cell migration. Inhibition of expression or activity of SPK1 in WT cells markedly decreased intracellular S1P levels and HGF-induced cell migration. Furthermore, we found that Shp-2 co-immunoprecipitated with SPK1 and c-Met in embryonic fibroblasts. These studies suggest that Shp-2 is an SPK1-interacting protein and that it plays an indispensable role in HGF-induced SPK1 activation.


Asunto(s)
Movimiento Celular/fisiología , Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/farmacología , Línea Celular , Quinasa de Punto de Control 2 , Embrión de Mamíferos/citología , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Fosfotransferasas (Aceptor de Grupo Alcohol)/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/farmacología
17.
Artículo en Chino | MEDLINE | ID: mdl-21166179

RESUMEN

AIM: To assess whether hepatocyte growth factor recruits bone marrow-derived endothelial progenitor cells into blood circulation to participate in postnatal angiogenesis and endothelium repair. METHODS: The adenovirus vector encoding HGF gene (Ad-HGF) were intravenous administrated into BALB/c mice, and then serum HGF was determined by enzyme-linked immunosorbent assay, the number of CD34+ cells in peripheral blood was assayed by flow cytometry, and the nucleated cells in peripheral blood were isolated, cultured and the endothelial cell colonies were characterized by staining with antibodies against tie-2, vWF. The carbon tetrachloride-induced liver damage model of female mice was established. The peripheral blood nucleated cells of Ad-HGF treated male mice were intravenous administrated into these mice, and 4 weeks later, in situ hybridization for the sry gene was used to identify the implanted cells in the damaged tissues. RESULTS: Intravenous administration of Ad-HGF resulted in significant elevation of serum hepatocyte growth factor level and induced profoundly increase of endothelial progenitor cells in the peripheral blood, which were characterized by their ability to form endothelial cell colonies in culture and expression of CD34, tie-2, and vW factor. HGF-mobilized endothelial progenitors could incorporate into sites of neovascularization in a liver regeneration model. CONCLUSION: Hepatocyte growth factor could markedly recruit bone marrow-derived endothelial progenitor cells into blood circulation.


Asunto(s)
Células de la Médula Ósea/citología , Factor de Crecimiento de Hepatocito/farmacología , Células Madre/citología , Animales , Células Endoteliales/citología , Femenino , Movilización de Célula Madre Hematopoyética , Masculino , Ratones , Ratones Endogámicos BALB C
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 13(6): 1044-8, 2005 Dec.
Artículo en Chino | MEDLINE | ID: mdl-16403277

RESUMEN

Hepatocyte growth factor (HGF) is one of major growth factors in the bone marrow microenvironments with which the proliferation, differentiation and migration of bone marrow-derived mesenchymal stem cells were closely contacted. However, its roles in the regulation of proliferation, differentiation and migration of bone marrow-derived mesenchymal stem cells remain unclear. This study was aimed to investigate the effect of HGF on biological characteristics of bone marrow-derived mesenchymal stem cells. Expression of c-Met, the receptor for HGF was detected by immunohistochemistry assay, cell proliferation was determined by MTT, activity of ALP was quantitatively assayed, cell migration and anoikis-induced MSC apoptosis were analyzed. The results showed that HGF not influenced the proliferation and osteogenic differentiation of bone marrow-derived mesenchymal stem cells. Treatment of bone marrow-derived mesenchymal stem cells with recombinant human hepatocyte growth factor resulted in inhibition of anoikis-induced apoptosis. HGF significantly stimulated the migration of bone marrow-derived mesenchymal stem cells. Both PI-3 kinase and MAPK kinase were proved to be involved in HGF-induced migration. It is concluded that HGF/c-Met signal regulates the apoptosis and migration of bone marrow-derived mesenchymal stem cells.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/biosíntesis , Anoicis/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo
19.
Exp Cell Res ; 298(2): 593-601, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15265705

RESUMEN

Hepatocyte growth factor (HGF)-induced migration of endothelial cells is critical for angiogenesis. Sphingosine kinase (SPK) is a key enzyme catalyzing the formation of sphingosine-1-phosphate (S1P), a lipid messenger that is implicated in the regulation of a wide variety of important cellular events through both intracellular and extracellular mechanisms. The aim of this study was to investigate whether activation of SPK is involved in the migration of endothelial cells induced by HGF. The biological functions of HGF are mediated through the activation of its high-affinity tyrosine kinase receptor, c-met protooncogene. In the present study, Treatment of ECV304 endothelial cells with HGF resulted in tyrosine phosphorylation of c-Met and activation of SPK in a concentration-dependent manner. Either Ly294002 or PD98059, specific inhibitor of the PI3K and ERK/MAPK pathways, respectively, blocked the HGF-induced activation of SPK. HGF stimulation significantly increased intracellular S1P level, but no detectable secretion of S1P into the cell culture medium was observed. Treatment of ECV304 cells with pertussis toxin (PTX) has no effect on the HGF-induced migration, indicating extracellular S1P is dispensable for this process. Overexpression of wild-type SPK gene in ECV 304 cells increased the intracellular S1P and enhanced the HGF-induced migration, whereas inhibition of cellular SPK activity by N,N-dimethylsphingosine (DMS), a potent inhibitor of SPK, or by expression of a dominant-negative SPK (DN-SK) blocked the HGF-induced migration of ECV 304 cells. It is suggested that PI3K and ERK/MAPK mediated the activation of SPK and would be involved in the HGF-induced migration of endothelial cells. These results elucidate a novel mechanism by which intracellularly generated S1P mediates signaling from HGF/c-Met to the endothelial cell migration.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/enzimología , Factor de Crecimiento de Hepatocito/metabolismo , Lisofosfolípidos/metabolismo , Neovascularización Fisiológica/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Toxina del Pertussis/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas c-met/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esfingosina/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
20.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 12(3): 335-9, 2004 Jun.
Artículo en Chino | MEDLINE | ID: mdl-15228662

RESUMEN

The proliferation and apoptosis of multiple myeloma (MM) cells were regulated by bone marrow microenvironments in which Notch signal plays important role in mediating cell-cell communication. However, the regulatory effect of Notch signal on the proliferation and apoptosis of multiple myeloma cells remains unclear. In this study, regulatory effect of Notch signal on the apoptosis of MM cells induced by DMS (N, N-dimethylsphingosine) was investigated. RT-PCR was used to identify the expression of Notch receptor and related molecules such as Dll-1, Jagged-1, Deltex-1 in MM cell lines. The intracellular domain of Notch (ICN), active form of Notch, was transferred into MM cells by retrovirus. The apoptosis of MM cells was determined by trypan blue exclusion tests and TdT-mediated dUTP nick end labeling (TUNEL) assay. The results showed that multiple myeloma cells expressed the Notch-1 and its related molecules. Notch activated multiple myeloma cell lines were obtained. Activation of Notch protected the multiple myeloma cells from the apoptosis induced by DMS,which was determined by cell viability and TUNEL assay. In conclusion, Notch signal suppressed the apoptosis of multiple myeloma cells and would possibly be a novel therapeutic target.


Asunto(s)
Apoptosis , Mieloma Múltiple/patología , Receptores de Superficie Celular/fisiología , Factores de Transcripción/fisiología , División Celular , Humanos , Mieloma Múltiple/tratamiento farmacológico , Receptor Notch1 , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...