Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cancer Sci ; 109(5): 1660-1671, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29601143

RESUMO

We previously reported that, in a mouse model of mammary cancer, α-mangostin alone exhibits anti-metastatic properties. To enhance this anti-metastatic effect, we examined the efficacy of synthetic α-mangostin dilaurate (MGD), prepared by adding lauric acid to α-mangostin, in the same experimental system wherein mice bearing mammary tumors are exposed to dietary MGD at 0, 2000 and 4000 ppm. Lauric acid has a high propensity for lymphatic absorption, which is the most common pathway of initial dissemination of many solid malignancies. Both mammary tumor volumes and wide-spectrum organ metastasis were markedly reduced at 2000 and 4000 ppm: furthermore, survival in the 4000-ppm group was significantly greater than in control mice. Apoptosis in mammary carcinomas was also significantly increased in the 4000-ppm group, whereas blood microvessel density and lymphatic vessel invasion were markedly reduced. In real-time PCR analyses of tumor samples, increased p21 and decreased Pcna expression were observed with 4000 ppm but values were not statistically significant when compared to expression in control tumors. However, exposure to 4000 ppm significantly decreased expression of phospho-Akt (Ser473/Thr308) as compared to the control, indicating a role in the anti-tumorigenic effects of MGD. These findings suggest that MGD may be useful for adjuvant therapy and chemoprevention and that conjugated medium-chain fatty acids may enhance the efficacy of certain chemotherapeutic agents.


Assuntos
Neoplasias Mamárias Experimentais/tratamento farmacológico , Xantonas/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Lauratos , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica , Antígeno Nuclear de Célula em Proliferação/análise , Proteínas Proto-Oncogênicas c-akt/metabolismo
2.
BMC Med ; 9: 69, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21639868

RESUMO

BACKGROUND: The mangosteen fruit has a long history of medicinal use in Chinese and Ayurvedic medicine. Recently, the compound α-mangostin, which is isolated from the pericarp of the fruit, was shown to induce cell death in various types of cancer cells in in vitro studies. This led us to investigate the antitumor growth and antimetastatic activities of α-mangostin in an immunocompetent xenograft model of mouse metastatic mammary cancer having a p53 mutation that induces a metastatic spectrum similar to that seen in human breast cancers. METHODS: Mammary tumors, induced by inoculation of BALB/c mice syngeneic with metastatic BJMC3879luc2 cells, were subsequently treated with α-mangostin at 0, 10 and 20 mg/kg/day using mini-osmotic pumps and histopathologically examined. To investigate the mechanisms of antitumor ability by α-mangostin, in vitro studies were also conducted. RESULTS: Not only were in vivo survival rates significantly higher in the 20 mg/kg/day α-mangostin group versus controls, but both tumor volume and the multiplicity of lymph node metastases were significantly suppressed. Apoptotic levels were significantly increased in the mammary tumors of mice receiving 20 mg/kg/day and were associated with increased expression of active caspase-3 and -9. Other significant effects noted at this dose level were decreased microvessel density and lower numbers of dilated lymphatic vessels containing intraluminal tumor cells in mammary carcinoma tissues. In vitro, α-mangostin induced mitochondria-mediated apoptosis and G1-phase arrest and S-phase suppression in the cell cycle. Since activation by Akt phosphorylation plays a central role in a variety of oncogenic processes, including cell proliferation, anti-apoptotic cell death, angiogenesis and metastasis, we also investigated alterations in Akt phosphorylation induced by α-mangostin treatment both in vitro and in vivo. Quantitative analysis and immunohistochemistry showed that α-mangostin significantly decreased the levels of phospho-Akt-threonine 308 (Thr308), but not serine 473 (Ser473), in both mammary carcinoma cell cultures and mammary carcinoma tissues in vivo. CONCLUSIONS: Since lymph node involvement is the most important prognostic factor in breast cancer patients, the antimetastatic activity of α-mangostin as detected in mammary cancers carrying a p53 mutation in the present study may have specific clinical applications. In addition, α-mangostin may have chemopreventive benefits and/or prove useful as an adjuvant therapy, or as a complementary alternative medicine in the treatment of breast cancer.


Assuntos
Antineoplásicos/farmacologia , Garcinia mangostana/química , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Metástase Neoplásica/prevenção & controle , Neoplasias/tratamento farmacológico , Xantonas/farmacologia , Animais , Antineoplásicos/isolamento & purificação , Feminino , Humanos , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Transplante Heterólogo , Resultado do Tratamento , Proteína Supressora de Tumor p53/genética , Xantonas/isolamento & purificação
3.
BMC Med ; 8: 69, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21047425

RESUMO

BACKGROUND: Cancer metastasis contributes significantly to cancer mortality and is facilitated by lymphangiogenesis and angiogenesis. A new splicing variant, endogenous soluble vascular endothelial growth factor receptor-2 (esVEGFR-2) that we recently identified is an endogenous selective inhibitor of lymphangiogenesis. To evaluate the antimetastatic potential of esVEGFR-2, gene therapy with vector expressing esVEGFR-2 (pesVEGFR-2) or endostatin (pEndo) as a positive control was conducted on murine metastatic mammary cancer. METHODS: Syngeneic inoculated metastatic mammary cancers received direct intratumoral injection of pesVEGFR-2, pEndo or pVec as control, once a week for six weeks. In vivo gene electrotransfer was performed on the tumors after each injection. RESULTS: Deaths from metastasis were much lower in the pesVEGFR-2 and pEndo groups than in those of the pVec. Tumor volume was significantly lower in the pesVEGFR-2 and the pEndo groups throughout the study. Multiplicity of lymph node and lung metastatic nodules was significantly suppressed in the pesVEGFR-2 and pEndo groups. Moreover, the total number of overall metastasis including the other organs was also decreased in these groups. However, pesVEGFR-2 was not able to decrease the number of lungs, ovaries, kidneys and adrenals with metastasis as counted by unilateral or bilateral metastasis. The number of CD34+/Lyve-1⁻ blood microvessels was significantly decreased in the pEndo group, while the number of CD34⁻/Lyve-1+ lymphatic vessels was significantly decreased in the pesVEGFR-2 and pEndo groups. In addition, a significant reduction in the number of dilated lymphatic vessels containing intraluminal cancer cells was observed in the pesVEGFR-2 and pEndo groups. Levels of apoptosis were significantly increased in the pEndo group, whereas the rates of cell proliferation were significantly decreased in the pesVEGFR-2 and pEndo groups. CONCLUSIONS: Our data demonstrate that esVEGFR-2 can inhibit mainly lymph node metastasis. The antimetastatic activity of esVEGFR-2 may be of high clinical significance in the treatment of metastatic breast cancer because lymph node involvement is a most important prognostic factor in cancer patients.


Assuntos
Linfangiogênese/efeitos dos fármacos , Metástase Linfática/genética , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Fatores de Crescimento do Endotélio Vascular/genética , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Terapia Genética , Vetores Genéticos , Imunocompetência , Neoplasias Pulmonares/secundário , Linfangiogênese/genética , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Metástase Neoplásica , Isoformas de Proteínas , Receptores de Fatores de Crescimento do Endotélio Vascular/farmacologia , Carga Tumoral/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/farmacologia
4.
BMC Cancer ; 10: 566, 2010 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-20958960

RESUMO

BACKGROUND: The effects of raloxifene, a novel selective estrogen receptor modulator, were studied in a mouse metastatic mammary cancer model expressing cytoplasmic ERα. METHODS: Mammary tumors, induced by inoculation of syngeneic BALB/c mice with BJMC3879luc2 cells, were subsequently treated with raloxifene at 0, 18 and 27 mg/kg/day using mini-osmotic pumps. RESULTS: In vitro study demonstrated that the ERα in BJMC3879luc2 cells was smaller (between 50 and 64 kDa) than the normal-sized ERα (66 kDa) and showed cytoplasmic localization. A statistically significant but weak estradiol response was observed in this cell line. When BJMC3879luc2 tumors were implanted into mice, the ERα mRNA levels were significantly higher in females than in males. In vitro studies showed that raloxifene induced mitochondria-mediated apoptosis and cell-cycle arrest in the G1-phase and a decrease in the cell population in the S-phase. In animal experiments, tumor volumes were significantly suppressed in the raloxifene-treated groups. The multiplicity of lymph node metastasis was significantly decreased in the 27 mg/kg group. Levels of apoptosis were significantly increased in the raloxifene-treated groups, whereas the levels of DNA synthesis were significantly decreased in these groups. No differences in microvessel density in tumors were observed between the control and raloxifene-treated groups. The numbers of dilated lymphatic vessels containing intraluminal tumor cells were significantly reduced in mammary tumors in the raloxifene-treated groups. The levels of ERα mRNA in mammary tumors tended to be decreased in the raloxifene-treated groups. CONCLUSION: These results suggest that the antimetastatic activity of raloxifene in mammary cancer expressing cytoplasmic ERα may be a crucial finding with clinical applications and that raloxifene may be useful as an adjuvant therapy and for the chemoprevention of breast cancer development.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/metabolismo , Neoplasias/tratamento farmacológico , Cloridrato de Raloxifeno/farmacologia , Animais , Apoptose , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Humanos , Metástase Linfática , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Fatores Sexuais
5.
Anticancer Res ; 40(5): 2687-2694, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32366413

RESUMO

BACKGROUND/AIM: Transforming growth factor-ß (TGF-ß) plays dual suppressive and oncogenic roles in mammary carcinogenesis. MATERIALS AND METHODS: To analyze whether TGF-ß exerts suppressive or oncogenic actions on mammary carcinogenesis, transgenic mice overexpressing a dominant-negative mutant type II TGF-ß receptor (TßRII-DNR) driven by the mouse mammary tumor virus (MMTV) promoter were treated with a low dose of urethane, a carcinogen present in fermented food products and alcoholic beverages. RESULTS: Lobular proliferative lesions, showing high ß-casein expression, developed in the mammary glands of TßRII-DNR+/+ mice aged >61 weeks. Compared with wild-type mice, TßRII-DNR+/+ mice administered with urethane showed significant increases in dysplastic hyperplasias and adenocarcinomas of the mammary glands. CONCLUSION: The functional decline of TGF-ß signaling in mammary glands led to a high susceptibility to urethane-induced mammary carcinogenesis. TGF-ß signaling may act as a tumor suppressor during mammary tumor development.


Assuntos
Carcinogênese/patologia , Genes Dominantes , Neoplasias Mamárias Animais/genética , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Neoplasias Pulmonares/patologia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/induzido quimicamente , Neoplasias Mamárias Animais/patologia , Camundongos Transgênicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Transgenes , Uretana
6.
Cancer Chemother Pharmacol ; 60(5): 681-91, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17256131

RESUMO

PURPOSE: The effects of vaticanol C (Vat-C), a novel resveratrol tetramer, were studied in a mouse metastatic mammary cancer model carrying mutations in p53 that produce a metastatic spectrum similar to that seen in human breast cancers. METHODS: Mammary tumors, induced by inoculation of syngeneic BALB/c mice with BJMC3879 cells, were subsequently treated with Vat-C at 0, 100 and 200 ppm in their diet. RESULTS: The in vitro study demonstrated that Vat-C induced apoptosis, as inferred by morphological changes, nucleosomal DNA fragmentation and elevated activities of caspases. Although tumor volumes were not apparently suppressed in mice treated with Vat-C, the multiplicity of lymph node metastasis was significantly decreased in the 200-ppm group. Furthermore, the multiplicity of lung metastasis was also significantly lower in the 200-ppm group. In any category of organ metastasis, the number of organs with metastasis tended to be lower in the 200-ppm group, but these findings were not statistically significant. The levels of apoptosis were significantly higher in the 200-ppm group, but DNA synthesis only a tended to be lower in this group. Microvessel density in tumors also tended to be lower in the Vat-C-treated groups. Moreover, the numbers of lymphatic vessels having intraluminal tumor cells was significantly lower in mammary tumors of mice given 100 and 200-ppm Vat-C, indicating a reduction in migrating tumor cells into the lymphatic vessels of tumor tissue. CONCLUSIONS: These results suggest that the observed antimetastatic activity of Vat-C may be of clinical significance as an adjuvant therapy in metastatic human breast cancer having p53 mutations, and may also be useful as a chemopreventative of breast cancer development.


Assuntos
Genes p53 , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Metástase Linfática/prevenção & controle , Neoplasias Mamárias Experimentais/patologia , Mutação , Estilbenos/uso terapêutico , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose , Peso Corporal , Feminino , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Células Tumorais Cultivadas
7.
Anticancer Res ; 26(6B): 4245-54, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17201140

RESUMO

BACKGROUND: The antitumor growth and antimetastatic actions of celecoxib [a selective cyclooxygenase-2 (COX-2) inhibitor] were investigated in a metastatic murine mammary cancer model. MATERIALS AND METHODS: Mice bearing mammary tumors, developed after inoculation of syngeneic BALBIc mice with a mammary carcinoma cell line carrying a p53 mutation, were treated with celecoxib at 0, 7.5 and 15 mg/kg five times a week for seven weeks. RESULTS: Tumor volumes were significantly reduced in association with an increase in apoptosis and a decrease in DNA synthesis in tumor tissues. In vitro studies demonstrated a significant increase in the number of cells undergoing apoptosis, with significantly elevated activities of caspase-3 and caspase-9, but not caspase-8, and a dose-dependent decrease in mitochondrial membrane potential, indicating the mitochondrial pathway of apoptosis. In addition, treatment with celecoxib showed cell cycle arrest in the G -phase and decreased cell population in the S- and G2/M-phases. Furthermore, tumor microvessel formation and mRNA levels for VEGF-A and COX-2 were markedly decreased. CONCLUSION: Celecoxib may be useful as an adjuvant therapy for breast cancer containing p53 mutations due to its ability to both induce p53-independent mitochondria-mediated apoptosis and exert anti-angiogenic potential.


Assuntos
Divisão Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Celecoxib , Replicação do DNA/efeitos dos fármacos , Neoplasias Pulmonares/prevenção & controle , Neoplasias Mamárias Experimentais/irrigação sanguínea , Camundongos , Camundongos Endogâmicos BALB C
8.
Cancer Res ; 62(5): 1410-9, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11888913

RESUMO

FTY720 is a unique immunosuppressive agent that exerts its activity by inducing apoptosis in lymphocytes. We conducted the present study to investigate the effects of FTY720 on cancer growth and metastasis, as well as its mechanism of action. In vitro treatment with FTY720 induced dramatic cancer cell apoptosis in a mouse breast cancer cell line, JygMC(A). Electron microscopy revealed distinct changes on the cell surface with decreased filopodias and microvilli in cancer cells treated with FTY720 at 2 microM and clear evidence of apoptosis at 10 microM. Interestingly, the effect of FTY720 was significantly less in the normal fibroblasts than in the cancer cells, indicating greater susceptibility of cancer cells to the agent. We then tested the in vivo effect of FTY720 in a mouse breast cancer model created by inoculating JygMC(A) cells (s.c.) in the flank region of BALB/c-nu/nu mice at three different dosages (2, 5, and 10 mg/kg/day; n = 30/group). Tumor growth was markedly suppressed at a dosage of 5 mg/kg or more without notable side effects. In addition, tumor metastasis, which was dramatically evident in control mice, was significantly prevented even at a low dose (2 mg/kg/day), resulting in a significant prolongation of animal survival. These data led us to additionally investigate the mechanism of action, especially the prevention of metastasis at a low dose. FTY720 treatment at 2 microM caused a remarkable cytoskeletal change with deformed and decreased filopodias in cancer cells. In addition, it significantly decreased the ability of cancer cells to adhere and migrate to extracellular matrix components, and markedly reduced the expression of integrins on the cancer cell surface. These results indicate that FTY720 is a potent anticancer agent that induces cancer cell apoptosis and is markedly effective for prevention of metastasis. The changes of cellular structure with reduction of integrin expression may be one of its underlying mechanisms of action.


Assuntos
Imunossupressores/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Metástase Neoplásica/prevenção & controle , Propilenoglicóis/farmacologia , Actinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Feminino , Cloridrato de Fingolimode , Integrinas/análise , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Propilenoglicóis/efeitos adversos , Esfingosina/análogos & derivados , Células Tumorais Cultivadas
9.
Cancer Gene Ther ; 9(1): 16-27, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11916241

RESUMO

The effectiveness of electroporation as a means of gene transfection, both in vitro and in vivo, was tested using the herpes simplex virus 1 thymidine kinase (HSVtk) gene in combination with ganciclovir (GCV) administration as therapy against murine mammary cancer. Approximately 80% of BJMC3879 metastatic mammary carcinoma cells, derived from MMTV-infected BALB/c mice, died as a result of HSVtk/GCV treatment 72 hours after the transfection; decreased DNA synthesis was also seen. Mammary tumors induced by inoculation of syngeneic mice with BJMC3879 cells were subsequently treated by direct injection of vector containing HSVtk (pHSVtk) alone, empty vector or saline alone twice a week. After each injection, the tumors were subjected to in vivo electroporation. Mice treated with pHSVtk or saline were intraperitoneally injected with GCV at 40 mg/kg five times a week. Significantly reduced tumor volumes were observed for the pHSVtk+GCV group in experimental week 2 and thereafter throughout the 2-month study. DNA synthesis was significantly decreased as well in the pHSVtk+GCV group compared with all other groups. Furthermore, metastasis to lymph nodes and lungs was significantly suppressed by HSVtk/GCV treatment. Expression of HSVtk in the tumors was confirmed by RT-PCR. Macrophage accumulations were frequently observed in the peripheries of necrotic regions in HSVtk/GCV-treated tumors, where levels of apoptosis were significantly higher than those observed in other groups. We therefore conclude that in vivo electroporation can result in efficient gene transfer and that the HSVtk/GCV prodrug system strongly suppresses tumor growth and metastases in this model.


Assuntos
Antivirais/uso terapêutico , Ganciclovir/uso terapêutico , Vetores Genéticos/uso terapêutico , Herpesvirus Humano 1/genética , Neoplasias Pulmonares/terapia , Neoplasias Mamárias Experimentais/terapia , Timidina Quinase/genética , Animais , Apoptose , Efeito Espectador , Primers do DNA/química , Eletroporação/métodos , Feminino , Terapia Genética , Herpesvirus Humano 1/enzimologia , Humanos , Neoplasias Pulmonares/secundário , Linfonodos/patologia , Metástase Linfática , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timidina Quinase/metabolismo , Transplante Heterólogo
10.
Anticancer Res ; 23(4): 3183-93, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12926052

RESUMO

BACKGROUND: FTY720 is a unique immunosuppressant that induces apoptosis in activated lymphocytes, but not in other hematopoietic cells. We conducted the present study to investigate its anticancer effect and molecular pathway in inducing apoptosis using murine breast cancer models. MATERIALS AND METHODS: The difference in drug susceptibility to FTY720 between cancer cells and non-cancer cells was examined by MTT assay and cell growth assay. FTY720-induced apoptosis was determined by electron microscopy and DNA electrophoresis, and its molecular pathway was evaluated by Western blot analysis. We then tested in vivo the effect of this agent using two murine breast cancer models. RESULTS: FTY720 treatment induced selective cancer cell apoptosis in vitro at a concentration of less than 10 microM. In vivo tumor growth was significantly prevented with induction of apoptosis in both models without any severe systemic adverse reactions. The evaluation of intracellular protease activity demonstrated that FTY720-induced apoptosis was mediated by a Fas-independent, Bcl-associated signal transduction pathway. Inhibition of extracellular signal-regulated kinase (ERK) activity may be involved in its underlying mechanism of action. CONCLUSION: FTY720 may be a promising candidate for a new anticancer therapy, which potentially induces selective apoptosis in cancer cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Imunossupressores/farmacologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Propilenoglicóis/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Antineoplásicos/efeitos adversos , Divisão Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Cloridrato de Fingolimode , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Imunossupressores/efeitos adversos , Masculino , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Propilenoglicóis/efeitos adversos , Esfingosina/análogos & derivados , Proteína bcl-X
11.
Anticancer Res ; 33(10): 4237-47, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24122987

RESUMO

Cancer metastasis contributes significantly to cancer mortality and is facilitated by lymphangiogenesis and angiogenesis. Vascular endothelial growth factors-C and D (VEGF-C and VEGF-D) are heavily involved in lymphangiogenesis. Using small interfering RNA (siRNA) against mouse Vegf-c, and Vegf-d, we sought to inhibit metastasis in a model of metastatic murine mammary cancer. BJMC3879Luc2 cell-induced mammary carcinomas received direct intratumoral injections in vivo of either plasmid VEGF-C/D siRNA (psiVEGF-C, psiVEGF-D) or a vector control followed by in vivo gene electrotransfer weekly for seven weeks. Treatment with psiVEGF-C and with psiVEGF-D resulted in lower tumor volumes as compared to the controls. Treatment with psiVEGF-C suppressed wide-spectrum organ metastasis involving lung and lymph nodes. Treatment with psiVEGF-C further reduced the number of dilated lymphatic vessels with invading cancer cells and inhibited tumor blood microvessel density. In contrast, although treatment with psiVEGF-D was not effective and gave equivocal results, it did induce some insignificant reduction in tumor volume increment, average numbers of lymph node metastases and average number of intratumoral dilated lymphatic vessels. In conclusion, specific silencing of the Vegf-c gene suppresses wide-spectrum organ metastasis, including the lung and lymph nodes. However, therapy with siRNA for Vegf-d was not adequately effective in this murine system.


Assuntos
Adenocarcinoma/terapia , Neoplasias Pulmonares/terapia , Neoplasias Mamárias Experimentais/terapia , RNA Interferente Pequeno/genética , Fator C de Crescimento do Endotélio Vascular/genética , Fator D de Crescimento do Endotélio Vascular/genética , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/secundário , Animais , Modelos Animais de Doenças , Feminino , Terapia Genética , Imunocompetência , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/secundário , Metástase Linfática , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Microvasos/patologia , Transplante de Neoplasias , Carga Tumoral , Fator C de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo
12.
Anticancer Res ; 31(4): 1225-34, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21508369

RESUMO

BACKGROUND: Sunitinib is an inhibitor that blocks tyrosine phosphorylation (p-Tyr) of receptors including vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor. Sunitinib suppresses angiogenesis and cell proliferation and is an effective treatment for renal cell carcinoma and gastrointestinal stromal tumors. In the present study, we examined the antitumor and antimetastatic activities of sunitinib in mouse metastatic mammary cancer. MATERIALS AND METHODS: Mammary tumors induced by inoculation of BJMC3879 cells into mice were treated with sunitinib using mini-osmotic pumps. At 1 week and 7 weeks after initiation of drug administration, cancer tissue was removed and carried out histopathological and immunohistochemical examination. RESULTS: Tumor growth, as well as metastasis to the lungs and other organs, was significantly inhibited in sunitinib-treated mice. Cell death areas in mammary carcinomas were much larger in the sunitinib-treated groups than in the control group. In addition, sunitinib induced necrotic cell death rather than apoptosis. Although microvessel density was significantly lower in the sunitinib-treated mammary tumors, numbers of metastases to lymph nodes and the number of lymphatic vessels in the mammary tumors were not significantly different among groups. Cell proliferation, as assessed by BrdU-labeling indices, was significantly lower in mammary carcinomas of sunitinib-treated mice. The amounts of p-VEGFR-2 and p-Tyr, as determined by immunohistochemistry, were greatly reduced in sunitinib-treated mice. CONCLUSION: In a mouse model of mammary cancer, sunitinib inhibited tumor growth and metastasis (with the exception of lymph node metastasis), angiogenesis, and cell proliferation possibly due to reduced levels of p-VEGFR-2 and p-Tyr.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Apoptose/efeitos dos fármacos , Indóis/uso terapêutico , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/prevenção & controle , Neovascularização Patológica/prevenção & controle , Pirróis/uso terapêutico , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Feminino , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação/efeitos dos fármacos , Sunitinibe , Células Tumorais Cultivadas , Tirosina/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Int J Breast Cancer ; 2011: 867152, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22295235

RESUMO

Lymphangiogenesis and the expression of vascular endothelial cell growth factor C (VEGF-C) in tumors have been considered to be causally promoting lymphatic metastasis. There are only a few studies on lymphatic metastasis in immunocompetent allograft mouse models. To study the relationship between VEGF-C-mediated lymphangiogenesis and axillary lymph node metastasis, we used two mouse mammary carcinoma cell lines; the BJMC338 has a low metastatic propensity, whereas the BJMC3879 has a high metastatic propensity although it originated from the former cell line. Each cell line was injected separately into two groups of female BALB/c mice creating in vivo mammary cancer models. The expression level of VEGF-C in BJMC3879 was higher than BJMC338. As the parent cell line, BJMC3879-derived tumors showed higher expression of VEGF-C compared to BJMC338-derived tumors. This higher expression of VEGF-C in BJMC3879-derived tumors was associated with marked increase in infiltrating macrophages and enhanced expression of lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) reflecting increased tumoral lymphatic density and subsequent induction of axillary lymph node metastasis. Our mouse mammary carcinoma models are allotransplanted tumors showing the same axillary lymph node metastatic spectrum as human breast cancers. Therefore, our mouse models are ideal for exploring the various molecular mechanisms of cancer metastasis.

14.
Anticancer Res ; 29(11): 4389-95, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20032383

RESUMO

BACKGROUND: Many areas of research, including gene and pharmacological therapeutics, would benefit from longitudinal in vivo monitoring methodologies. To investigate the feasibility of one such methodology, we developed a murine mammary cancer model amenable to sequential bioluminescent imaging of tumor growth and metastasis in living animals. MATERIALS AND METHODS: Metastatic mouse mammary carcinoma BJMC3879 cells were transfected to stably express firefly luciferase and inoculated into immunocompetent female BALB/c mice. RESULTS: Sequential analysis using bioluminescent imaging showed increasing photon counts correlated to expanding mammary tumor volumes; in addition, strong signals from axillary, mandibular, femoral, thoracic and abdominal regions in mice were histopathologically determined to be due to metastases, the majority of which occurred in lymph nodes and lungs. CONCLUSION: The bioluminescent mouse mammary cancer model we established provides a method for quantifiable longitudinal in vivo imaging that can be used in gene and pharmacological therapy applications.


Assuntos
Luciferases de Vaga-Lume/análise , Neoplasias Mamárias Experimentais/enzimologia , Animais , Linhagem Celular Tumoral , Feminino , Luciferases de Vaga-Lume/biossíntese , Luciferases de Vaga-Lume/genética , Medições Luminescentes/métodos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transfecção
15.
Anticancer Res ; 29(7): 2485-95, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19596918

RESUMO

BACKGROUND: The antitumor growth and antimetastatic activity of panaxanthone (approximately 80% alpha-mangostin and 20% gamma-mangostin) were studied in a mouse metastatic mammary cancer model that produces a metastatic spectrum similar to that seen in human breast cancer. MATERIALS AND METHODS: Mammary tumors, induced by inoculation of syngeneic BALB/c mice with BJMC3879 cells, were subsequently treated with panaxanthone at 0, 2,500, or 5,000 ppm in their diet. In vitro studies were also conducted to evaluate the effects of alpha-mangostin, the main component of panaxanthone, on BJMC3879 cells. RESULTS: In the in vivo study, tumor volumes were significantly suppressed in mice treated with 2,500 and 5,000 ppm panaxanthone in their diet. The multiplicity of lung metastasis was significantly lower in the 5,000 ppm group. Lymph node metastasis also tended to decrease in the 5,000 ppm group but not significantly. The antitumor effects of panaxanthone were associated with elevation of apoptotic cell death, antiproliferation (inhibition of PCNA) and antiangiogenesis (inhibition of microvessel density). The in vitro study demonstrated that alpha-mangostin induced apoptosis, as evidenced by increased numbers of TUNEL-positive cells, elevated activities of caspases and a decrease in mitochondrial membrane potential, cell cycle arrest in the G(1)-phase and decreases in the cell population in the S- and G(2)/M-phases. CONCLUSION: These results suggest that the observed antimetastatic activity of panaxanthone may be of clinical significance as adjuvant therapy in metastatic human breast cancer, and may also be useful as a chemopreventative of breast cancer development.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Divisão Celular/efeitos dos fármacos , Modelos Animais de Doenças , Garcinia mangostana/química , Neoplasias Mamárias Experimentais/patologia , Metástase Neoplásica/prevenção & controle , Xantonas/farmacologia , Animais , Antineoplásicos Fitogênicos/isolamento & purificação , Apoptose , Peso Corporal , Ciclo Celular , Comportamento Alimentar , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Xantonas/isolamento & purificação
16.
Med Mol Morphol ; 41(1): 34-43, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18470679

RESUMO

These experiments were conducted to investigate whether the antimetastatic effects of HSVtk/GCV therapy involve T-cell-mediated immune responses. In the first experiment, immunocompetent syngeneic mice were inoculated with metastatic mammary cancers, then given a direct intratumoral injection of a plasmid vector containing a suicide gene (pHSVtk) or control vector once a week for 8 weeks. Gene electrotransfer treatment was applied to the tumors, and mice were administered ganciclovir (GCV) using a mini-osmotic pump. At the end of the experiment, tumor volume was significantly lower in the pHSVtk/GCV group. Macrophage accumulations were frequently observed in the peripheries of the necrotic regions in pHSVtk-transfected mice. Levels of CD4 and CD8 proteins in tumors were higher in the pHSVtk/GCV group than in the control group. Interleukin (IL)-12 mRNA levels tended to be higher in tumors in the pHSVtk/GCV group, but there were large variations. Tumor microvessel density was significantly lower in the pHSVtk/GCV group. The numbers of dilated lymphatic vessels containing intraluminal tumor cells tended to be higher in the pHSVtk/GCV group. However, vascular endothelial growth factor (VEGF)-A and VEGF-C mRNA levels in tumors were similar in the control and pHSVtk/GCV groups. In the second experiment, tumor volume and metastatic parameters were compared for immunocompetent syngeneic mice and immunodeficient athymic mice (without an intact T-cell system) given pHSVtk/GCV therapy. Although tumor volumes were significantly smaller in both syngeneic and athymic mice given pHSVtk/GCV therapy, the inhibition ratios (relative to control mice) were much greater in syngeneic mice than in athymic mice. No suppression of metastasis to the lymph nodes and lungs was observed for athymic mice given pHSVtk/GCV therapy. Our data suggest that HSVtk/GCV suicide gene therapy exerts an antimetastatic effect via a T-cell-mediated immune response.


Assuntos
Terapia Genética , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Linfócitos T/imunologia , Animais , Antivirais/uso terapêutico , Sequência de Bases , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Primers do DNA/genética , Feminino , Ganciclovir/uso terapêutico , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Interleucina-12/genética , Interleucina-12/metabolismo , Neoplasias Pulmonares/secundário , Metástase Linfática , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Timidina Quinase/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo
17.
Med Mol Morphol ; 40(2): 103-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17572846

RESUMO

We report an easy and stable transfection technique using electrogene transfer with a nonviral Epstein-Barr (EB) virus-based vector. To achieve stable transfection of human breast cancer cells, we conducted electrogene transfer of an EB virus-based plasmid vector (reduced size of oriP) containing the enhanced green fluorescence protein (eGFP) gene. Because the EB virus-based vector exhibits high transfer efficiency and strong persistent transgene expression as a result of autonomous replication in human cells, and as Nucleofector electrogene transfer can achieve highly efficient gene transfection, this method is particularly suitable for generation of stably transfected cell lines.


Assuntos
Neoplasias da Mama/genética , Eletroporação/instrumentação , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Vetores Genéticos/genética , Neoplasias Pulmonares/secundário , Plasmídeos/genética , Transfecção/métodos , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/ultraestrutura , Linhagem Celular Tumoral , Antígenos Nucleares do Vírus Epstein-Barr/genética , Feminino , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Plasmídeos/metabolismo
18.
Breast Cancer Res Treat ; 98(1): 31-43, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16752227

RESUMO

Aberrant carbohydrate expression frequently occurs in breast cancer and may endow cells with metastatic potential. Here we first studied the relationship between expression of Vicia villosa agglutinin (lectin) (VVA)-binding carbohydrates and aggressive breast cancer. We then investigated the molecular characteristics of these glycoproteins and compared them with those of glycoproteins recognized by the mouse anti-Tn monoclonal antibody (MAb) HB-Tn1. Histochemical studies of samples from 322 cases of invasive ductal carcinoma demonstrated that VVA-binding carbohydrate expression correlated with tumor stage, lymphatic invasion, and lymph node metastasis (p=0.0385, p=0.0019, and p=0.0430. respectively). Western blotting analysis of frozen materials from 39 cases, under denaturing and reducing conditions, revealed that the major cancer cell-specific VVA-binding proteins were molecules of about 30, 33, and >200 kDa. Cases expressing the approximately 33 kDa molecule had significant lymphatic invasion more frequently than did cases not expressing this molecule (p=0.0076). Binding of VVA to the approximately 30 and approximately 33 kDa molecules was completely lost by preincubation of VVA with 1 mM Tn antigen (N-acetylgalactosamine alpha1-O-serine). The VVA-binding molecules appeared to react with VU-3C6 anti-MUC1 MAb. Expression of HB-Tn1 in breast cancer cells showed significant correlation with expression of VVA-binding carbohydrate(s) (p<0.0001) but HB-Tn1 reactivity was not clearly related to breast cancer aggressiveness. Because anti-Tn MAbs bound to Tn antigen clusters, we concluded that atypical MUC1 bearing the noncluster form of Tn antigen is implicated in aggressive growth of primary breast cancer cells, particularly in lymphatic metastasis.


Assuntos
Antígenos Glicosídicos Associados a Tumores/biossíntese , Neoplasias da Mama/metabolismo , Glicoproteínas/metabolismo , Mucina-1/biossíntese , Lectinas de Plantas/química , Adulto , Idoso , Anticorpos Monoclonais/química , Carboidratos/química , Proliferação de Células , Epitopos/química , Feminino , Humanos , Metástase Linfática , Pessoa de Meia-Idade , Lectinas de Plantas/metabolismo
19.
J Gene Med ; 8(3): 335-52, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16345101

RESUMO

BACKGROUND: Human breast cancer metastasizes mainly to lymph nodes, lungs, liver, and bone; in the majority of cases, it is the development of metastases which leads to death. In order to suppress mammary cancer metastasis, we applied in vivo electrogene transfer (non-viral method) as a means of interleukin-12 (IL-12) gene therapy on highly metastatic murine mammary cancer model. METHODS: Metastatic mammary tumors induced by inoculation in BALB/c female mice were treated by intratumoral injections of either a plasmid vector containing IL-12 or empty vector and then subjected to in vivo electrogene transfer once a week for 8 weeks. RESULTS: Treatment with IL-12 resulted in elevation of both IL-12 and IFNgamma levels in mammary tumors and in serum and intratumoral levels of CD4 and CD8 proteins were also increased. Tumor volumes and lymphatic and pulmonary metastases were significantly reduced. The histopathological changes induced by IL-12 characteristically included marked inflammation, increased apoptosis, decreased DNA synthesis, peripheral influx of significantly greater numbers of active macrophages, and reduced blood microvessel density, and apoptotic vascular endothelial cells were frequently seen. Western blotting showed decreases in VEGFR-3 of tumors exposed to IL-12 gene therapy. In adjuvant immunofluorescence studies, the CD31-positive endothelial cells of microvessels showed decreased VEGFR-3 expression in IL-12-treated tumors. However, apparent alterations in VEGFR-3 expression of podoplanin-positive lymphatic endothelial cells were not observed in IL-12-treated tumors. Although recombinant IL-12 did not inhibit tubular formation of human umbilical vein endothelial cells in a Matrigel assay, recombinant IFNgamma did completely suppress the tubular formation. CONCLUSIONS: In vivo electrogene transfer of IL-12 exerts strong anti-tumorigenic and anti-metastatic effects likely due to T-cell-mediated immune responses as well as anti-angiogenic action.


Assuntos
Técnicas de Transferência de Genes , Interleucina-12/genética , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/terapia , Animais , Eletrofisiologia , Feminino , Vetores Genéticos , Imuno-Histoquímica , Interleucina-12/biossíntese , Interleucina-12/uso terapêutico , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica/prevenção & controle , Neovascularização Patológica , Plasmídeos , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/biossíntese
20.
Int J Cancer ; 114(6): 926-35, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15645421

RESUMO

The role of CD95 ligand (FasL/Apo-1L)-expressing tumors in immunosuppression or immunopotentiation is controversial. CD95L-transfected tumors induce immunopotentiation after vigorous neutrophil infiltration. Thus, the induction of neutrophil infiltration by CD95L seems to play an important role in tumor rejection. The mechanism by which CD95L-expressing tumors cause neutrophil infiltration and antitumor immunity has not been well understood. CXC chemokine receptor 2 (CXCR2) knockout (KO) mice are a powerful tool for studying CXC chemokine-mediated neutrophil infiltration. We investigated the roles of CD95L and chemokines in CD95L-induced antitumor activity by using CXCR2 KO mice and CD95LcDNA-transfected MethA (MethA + CD95L) fibrosarcoma. MethA + CD95L cells were completely rejected in wild-type (WT) and even in KO mice. MethA + CD95L cells injected intraperitoneally (i.p.) induced the recruitment of both neutrophils and macrophages in WT but only macrophages in KO mice, although CXC and CC chemokines were released in both mice. Macrophages incubated with MethA + CD95L cells released CXC and CC chemokines. Macrophages derived from WT and KO but not neutrophils from WT mice induced the recruitment of neutrophils when adoptively i.p. transferred with MethA + CD95L cells into CD95L/CD95-deficient mice. The different recruitment of inflammatory cells between WT and KO mice was attributed to bone marrow (BM) cells by BM transfer experiment. Our results demonstrated that CXC chemokines are essential for neutrophil recruitment and that macrophages but not neutrophils play a critical role in the CD95L-induced infiltration of inflammatory cells and the eradication of CD95L-expressing tumor cells.


Assuntos
Quimiocinas CXC/imunologia , Quimiocinas CXC/farmacologia , Fibrossarcoma/patologia , Macrófagos/imunologia , Infiltração de Neutrófilos , Receptor fas/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Proteína Ligante Fas , Fibrossarcoma/veterinária , Citometria de Fluxo , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , RNA Mensageiro/análise
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa