Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Oncol Rep ; 22(5): 1069-75, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19787223

RESUMEN

Down-regulated in renal cell carcinoma gene (DRR1) is one of the candidate tumor suppressor genes (TSGs) on human 3p21.1. This study was performed to validate the expression status of DRR1 gene in cancer cells and the expression pattern of the protein in clinical specimens of human lung cancer and to examine its potential as a molecular target for treatment of lung cancer in vivo. DRR1 expression was analyzed in 7 human lung cancer cell lines. DRR1 protein expression was also examined in clinical non-small cell lung cancer (NSCLC) specimens. Furthermore, effects of DRR1 re-expression on A549 cells in vitro and A549 xenograft tumors in nude mice were evaluated. Loss of DRR1 mRNA expression was detected in 6 of the 7 human cancer cell lines, the exception was the renal cancer cell line OS-RC-2. DRR1 protein expression was absent in 15 of 20 (75%) human NSCLC specimens by immunostaining. Transfection of DRR1 gene into DRR1-negative-expressing A549 cells resulted in significant cell growth suppression and apoptosis. Plasmids containing DRR1 cDNA complexed with DOTAP:Chol liposomes were administered intravenously via tail vein to nude mice bearing A549 xenograft tumors resulting in tumor growth inhibition and elevation of apoptosis compared with the controls. DRR1 is a potent growth suppressor of NSCLC, acting through apoptosis pathway in vivo and it may be a potential therapeutic gene for human lung cancer.


Asunto(s)
Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/genética , Cromosomas Humanos Par 3/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Terapia Genética , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Animales , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Proliferación Celular , Femenino , Genes Supresores de Tumor , Vectores Genéticos , Humanos , Técnicas para Inmunoenzimas , Liposomas , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Plast Reconstr Surg ; 143(5): 1371-1382, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30807498

RESUMEN

BACKGROUND: This study investigated whether a hyaluronic acid-povidone-iodine compound can enhance diabetic wound healing. METHODS: A dorsal skin defect (6 × 5 cm) in a streptozotocin-induced diabetes rodent model was used. Seventy male Wistar rats were divided into seven groups: I, normal control; II, diabetic control, no treatment; III, diabetic rats, lower molecular weight (100 kDa) hyaluronic acid; IV, rats, higher molecular weight (1000 kDa) hyaluronic acid; V, rats, 0.1% povidone-iodine; VI, rats, lower molecular weight hyaluronic acid plus povidone-iodine; and VII, rats, higher molecular weight hyaluronic acid plus povidone-iodine. Histologic examination was performed with hematoxylin and eosin staining. CD45, Ki-67, prolyl 4-hydroxylase, and vascular endothelial growth factor were evaluated with immunohistochemical staining. RESULTS: Compared with the control, higher molecular weight hyaluronic acid plus povidone-iodine-treated rats had significantly reduced wound area (p < 0.001). Higher molecular weight hyaluronic acid plus povidone-iodine increased wound healing time when compared with higher molecular weight hyaluronic acid, povidone-iodine, or lower molecular weight hyaluronic acid plus povidone-iodine. Histology revealed significantly increased neovessels and suppressed inflammatory response in the higher molecular weight hyaluronic acid plus povidone-iodine group when compared with the control group. Immunohistochemical staining revealed significantly increased Ki67, prolyl 4-hydroxylase, and vascular endothelial growth factor expression, and suppressed CD45 expression in the higher molecular weight hyaluronic acid plus povidone-iodine group when compared with the other groups. CONCLUSION: Higher molecular weight hyaluronic acid plus povidone-iodine complex dressing significantly facilitated diabetic wound healing via increasing neovascularization and tissue regeneration and suppressing a proinflammatory response.


Asunto(s)
Antiinfecciosos Locales/farmacología , Diabetes Mellitus Experimental/complicaciones , Ácido Hialurónico/farmacología , Povidona Yodada/farmacología , Cicatrización de Heridas/efectos de los fármacos , Administración Cutánea , Animales , Antiinfecciosos Locales/química , Antiinfecciosos Locales/uso terapéutico , Vendajes , Diabetes Mellitus Experimental/inducido químicamente , Pie Diabético/tratamiento farmacológico , Pie Diabético/etiología , Combinación de Medicamentos , Humanos , Ácido Hialurónico/química , Ácido Hialurónico/uso terapéutico , Masculino , Peso Molecular , Povidona Yodada/química , Povidona Yodada/uso terapéutico , Ratas , Ratas Wistar , Piel/efectos de los fármacos , Piel/patología , Estreptozocina/toxicidad , Resultado del Tratamiento
4.
Oncol Res ; 20(12): 579-87, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-24139416

RESUMEN

Metastasis is the most lethal attribute of human malignancy. High-level expression of survivin is involved in both carcinogenesis and angiogenesis in cancer. Previous studies indicate that a mutation of the threonine residue at position 34 (Thr34Ala) of survivin generates a dominant-negative mutant that induces apoptosis, inhibits angiogenesis, and suppresses highly metastatic breast carcinoma in mouse models. We investigated the efficacy of gene therapy with a survivin dominant-negative mutant and possible factors related to lymph node metastasis. The metastasis rate was compared between each group in order to find a survivin-targeted therapy against lymphangiogenesis in its earliest stages. We established lymph node metastasis models and treated animals with H22 tumors with Lip-mSurvivinT34A (Lip-mS), Lip-plasmid (Lip-P), or normal saline (NS). Eight days after the last dose, five randomly chosen mice from each group were sacrificed. We detected the apoptotic index, microvessel density (MVD), lymphatic microvessel density (LMVD), and the expression of VEGF-D with immunohistochemistry. After the remaining animals were sacrificed, we compared the tumor-infiltrated lymph nodes in each group. Administration of mSurvivinT34A plasmid complexed with cationic liposome (DOTAP/chol) resulted in the efficacious inhibition of tumor growth and lymph node metastasis within the mouse H22 tumor model. These responses were associated with tumor cell apoptosis, and angiogenesis and lymphangiogenesis inhibition. Our results suggested that Lip-mSurvivinT34A induced apoptosis and inhibited tumor angiogenesis and lymphangiogenesis, thus suppressing tumor growth and lymphatic metastasis. The mSurvivinT34A survivin mutant is a promising strategy of gene therapy to inhibit lymphatic metastasis.


Asunto(s)
Terapia Genética/métodos , Proteínas Inhibidoras de la Apoptosis/genética , Plásmidos/administración & dosificación , Plásmidos/genética , Proteínas Represoras/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Liposomas/administración & dosificación , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/terapia , Linfangiogénesis/genética , Metástasis Linfática , Ratones , Ratones Endogámicos BALB C , Mutación , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Proteínas Represoras/biosíntesis , Survivin
5.
J Cancer Res Clin Oncol ; 137(1): 19-28, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20217127

RESUMEN

PURPOSE: Patients with localized prostate cancer can usually achieve initial response to conventional treatment. However, most of them will inevitably progress to advanced disease stage. There is a clear need to develop innovative and effective therapeutics for prostate cancer. Mouse survivin T34A (mS-T34A) is a phosphorylation-defective Thr34 → Ala dominant negative mutant, which represents a potential promising target for cancer gene therapy. This study was designed to determine whether mS-T34A plasmid encapsuled by DOTAP-chol liposome (Lip-mS) has the anti-tumor activity against prostate cancer, if so, to further investigate the possible mechanisms. METHODS: In vitro, TRAMP-C1 cells were transfected with Lip-mS and examined for apoptosis by PI staining and flow cytometric analysis. In vivo, subcutaneous prostate cancer models were established in C57BL/6 mice, which were randomly assigned into three groups to receive i.v. administrations of Lip-mS, pVITRO2-null plasmid complexed with DOTAP-chol liposome (Lip-null) or normal saline every 2 days for eight doses. Tumor volume was measured. Tumor tissues were inspected for apoptosis by TUNEL assay. Microvessel density (MVD) was determined by CD31 immunohistochemistry. Alginate-encapsulated tumor cell test was conducted to evaluate the treatment effect on angiogenesis. RESULTS: Administration of Lip-mS resulted in significant inhibition in the growth of mouse TRAMP-C1 tumors. The anti-tumor response was associated with increased tumor cell apoptosis and decreased microvessel density. CONCLUSIONS: The present study may be of importance in the exploration of the potential application of Lip-mS in the treatment of a broad spectrum of tumors.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/genética , Mutación , Neoplasias de la Próstata/terapia , Proteínas Represoras/genética , Animales , Línea Celular Tumoral , Ácidos Grasos Monoinsaturados/administración & dosificación , Terapia Genética , Proteínas Inhibidoras de la Apoptosis/uso terapéutico , Liposomas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Compuestos de Amonio Cuaternario/administración & dosificación , Proteínas Represoras/uso terapéutico , Survivin
6.
J Exp Clin Cancer Res ; 29: 46, 2010 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-20462440

RESUMEN

BACKGROUND: A high concentration of cisplatin (CDDP) induces apoptosis in many tumor cell lines. CDDP has been administered by infusion to avoid severe toxicity. Recently, it has been reported that changes in survivin expression or function may lead to tumor sensitization to chemical and physical agents. The aim of this study was to determine whether a dominant-negative mouse survivin mutant could enhance the anti-tumor activity of CDDP. METHODS: A plasmid encoding the phosphorylation-defective dominant-negative mouse survivin threonine 34-->alanine mutant (survivin T34A) complexed to a DOTAP-chol liposome (Lip-mS) was administered with or without CDDP in Lewis Lung Carcinoma (LLC) cells and in mice bearing LLC tumors, and the effects on apoptosis, tumor growth and angiogenesis were assessed. Data were analyzed using one-way analysis of variance(ANOVA), and a value of P < 0.05 was considered to be statistically significant. RESULTS: LLC cells treated with a combination of Lip-mS and CDDP displayed increased apoptosis compared with those treated with Lip-mS or CDDP alone. In mice bearing LLC tumors and treated with intravenous injections of Lip-mS and/or CDDP, combination treatment significantly reduced the mean tumor volume compared with either treatment alone. Moreover, the antitumor effect of Lip-mS combined with CDDP was greater than their anticipated additive effects. CONCLUSION: These data suggest that the dominant-negative survivin mutant, survivin T34A, sensitized LLC cells to chemotherapy of CDDP. The synergistic antitumor activity of the combination treatment may in part result from an increase in the apoptosis of tumor cells, inhibition of tumor angiogenesis and induction of a tumor-protective immune response.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Cisplatino/farmacología , Liposomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Animales , Apoptosis , Terapia Combinada , Sinergismo Farmacológico , Femenino , Terapia Genética/métodos , Proteínas Inhibidoras de la Apoptosis , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica , Survivin
7.
Oncol Rep ; 23(1): 97-103, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19956869

RESUMEN

Radiosensitivity of tumors is due to a complex interaction of various factors, it has been reported that survivin also acts as a constitutive and inducible radioresistance factor in a panel of tumor cells and approaches designed to inhibit survivin expression or function may lead to tumor sensitisation to chemical and physical agents. Previously, we found that the plasmid encoding the phosphorylation-defective mouse survivin threonine 34-->alanine mutant complexed to DOTAP-chol liposome (Lip-mS) can suppress murine primary breast carcinoma. However, little is known regarding the biological effect of Lip-mS combined with radiation. The present study was designed to determine whether Lip-mS could enhance the anti-tumor activity of radiation. The Lewis Lung Carcinoma (LLC) cells treated with a combination of Lip-mS and radiation displayed apparently increased apoptosis compared with those treated with Lip-mS or radiation alone. Mice bearing LLC tumors were treated with intravenous injections of Lip-mS and radiation, the combined treatment significantly reduced mean tumor volume compared with either treatment alone. Moreover, the anti-tumor effect of Lip-mS combined with radiation was greater than their additive effect when compared with the expected effect of the combined treatment. These data suggest that inhibition of survivin using a dominant-negative mutant, survivin T34A, could sensitize LLC cells to radiation efficiently and the synergistic anti-tumor activity may in part result from increasing the apoptosis of tumor cells, inhibiting tumor angiogenesis and inducing a tumor-protective immune response in the combined treatment.


Asunto(s)
Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/radioterapia , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/fisiología , Tolerancia a Radiación/genética , Animales , Apoptosis , Colesterol/farmacología , Terapia Combinada/métodos , Ácidos Grasos Monoinsaturados/farmacología , Genes Dominantes , Proteínas Inhibidoras de la Apoptosis , Liposomas/farmacología , Ratones , Ratones Endogámicos C57BL , Plásmidos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Compuestos de Amonio Cuaternario/farmacología , Radioterapia/métodos , Proteínas Represoras , Survivin , Sales de Tetrazolio/farmacología , Tiazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA