Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Breast Cancer Res ; 7(6): R881-9, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16280035

RESUMEN

INTRODUCTION: Ductal carcinoma in situ (DCIS) is a noninvasive premalignant lesion and is considered a precursor to invasive carcinoma. DCIS accounts for nearly 20% of newly diagnosed breast cancer, but the lack of experimentally amenable in vivo DCIS models hinders the development of treatment strategies. Here, we demonstrate the utility of a mouse transplantation model of DCIS for chemoprevention studies using selective estrogen receptor modulators (SERMs). This model consists of a set of serially transplanted lines of genetically engineered mouse mammary intraepithelial neoplasia (MIN) outgrowth (MIN-O) tissue that have stable characteristics. We studied the ovarian-hormone-responsiveness of one of the lines with a particular focus on the effects of two related SERMs, tamoxifen and ospemifene. METHODS: The estrogen receptor (ER) status and ovarian-hormone-dependence of the mouse MIN outgrowth tissue were determined by immunohistochemistry and ovarian ablation. The effects of tamoxifen and ospemifene on the growth and tumorigenesis of MIN outgrowth were assessed at 3 and 10 weeks after transplantation. The effects on ER status, cell proliferation, and apoptosis were studied with immunohistochemistry. RESULTS: The MIN-O was ER-positive and ovarian ablation resulted in reduced MIN-O growth and tumor development. Likewise, tamoxifen and ospemifene treatments decreased the MIN growth and tumor incidence in comparison with the control (P < 0.01). Both SERMs significantly decreased cell proliferation. Between the two SERM treatment groups, there were no statistically significant differences in MIN-O size, tumor latency, or proliferation rate. In contrast, the ospemifene treatment significantly increased ER levels while tamoxifen significantly decreased them. CONCLUSION: Tamoxifen and ospemifene inhibit the growth of premalignant mammary lesions and the progression to invasive carcinoma in a transplantable mouse model of DCIS. The inhibitory effects of these two SERMs are similar except for their effects on ER modulation. These differences in ER modulation may suggest different mechanisms of action between the two related SERMs and may portend different long-term outcomes. These data demonstrate the value of this model system for preclinical testing of antiestrogen or other therapies designed to prevent or delay the malignant transformation of premalignant mammary lesions in chemoprevention.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Intraductal no Infiltrante/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Animales , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Quimioprevención , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Ratones , Invasividad Neoplásica , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/fisiología
2.
Clin Exp Metastasis ; 22(1): 47-59, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16132578

RESUMEN

Two cell lines, Met-1(fvb2) and DB-7(fvb2), with different metastatic potential, were derived from mammary carcinomas in FVB/N-Tg(MMTV-PyVmT) and FVB/N-Tg(MMTV-PyVmT ( Y315F/Y322F )) mice, transplanted into syngeneic FVB/N hosts and characterized. The lines maintain a stable morphological and biological phenotype after multiple rounds of in vitro culture and in vivo transplantation. The Met-1(fvb2) line derived from a FVB/N-Tg(MMTV-PyVmT) tumor exhibits invasive growth and 100% metastases when transplanted into the females FVB/N mammary fat pad. The DB-7(fvb2) line derived from the FVB/N-Tg(MMTV-PyVmT ( Y315F/Y322F )) with a "double base" modification at Y315F/Y322F exhibits more rapid growth when transplanted into the mammary fat pad, but a lower rate of metastasis (17%). The Met1(fvb2) cells show high activation of AKT, while DB-7(fvb2) cells show very low levels of AKT activation. The DNA content and gene expression levels of both cell lines are stable over multiple generations. Therefore, these two cell lines provide a stable, reproducible resource for the study of metastasis modulators, AKT molecular pathway interactions, and gene target and marker discovery.


Asunto(s)
Carcinoma/patología , Línea Celular Tumoral , Neoplasias Mamarias Experimentales/patología , Animales , Carcinoma/genética , Carcinoma/metabolismo , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Metástasis de la Neoplasia , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptores de Estrógenos/análisis , Receptores de Estrógenos/metabolismo
3.
Mol Cancer Ther ; 3(8): 941-53, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15299077

RESUMEN

The development of models to investigate the pathobiology of premalignant breast lesions is a critical prerequisite for development of breast cancer prevention and early intervention strategies. Using tissue transplantation techniques, we modified the widely used polyomavirus middle T (PyV-mT) transgenic mouse model of breast cancer to study the premalignant stages of tumorigenesis. Premalignant atypical lesions were isolated from PyV-mT transgenic mice and used to generate two sets of three mammary intraepithelial neoplasia (MIN) outgrowth lines. Investigation of these six unique lines, each of which fulfills the criteria for MIN, has provided new information regarding the biology of PyV-mT-induced neoplasia. Although expression of the PyV-mT transgene was the primary initiating event for all lines, they exhibited different tumor latencies, metastatic potentials, and morphologies. Six distinguishable morphologic patterns of differentiation were identified within the premalignant outgrowths that are likely to represent several tumorigenic pathways. Further, several tumor phenotypes developed from each line and the tumors developing from the six lines had different metastatic potentials. These observations are consistent with the hypothesis that distinct pathways of PyV-mT-initiated neoplastic progression lead to different outcomes with respect to latency and metastasis. The MIN outgrowth lines share several characteristics with precursors of human breast cancer including the observation that gene expression profiles of tumors are more similar to those of the MIN outgrowth line outgrowth from which they developed than to other tumors. These lines provide an opportunity to study the full range of events occurring secondary to PyV-mT expression in the mammary gland.


Asunto(s)
Antígenos Transformadores de Poliomavirus/química , Carcinoma in Situ/patología , Neoplasias Mamarias Animales/patología , Animales , Carcinoma in Situ/virología , Diferenciación Celular , Línea Celular Tumoral , ADN/metabolismo , Progresión de la Enfermedad , Evolución Molecular , Humanos , Inmunohistoquímica , Pulmón/patología , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/virología , Ratones , Ratones Transgénicos , Familia de Multigenes , Metástasis de la Neoplasia , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/química , Fenotipo , ARN/metabolismo , Receptores de Estrógenos/metabolismo , Factores de Tiempo , Transgenes
4.
Breast Cancer Res ; 6(3): R157-69, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15084239

RESUMEN

INTRODUCTION: In order to study metastatic disease, we employed the use of two related polyomavirus middle T transgenic mouse tumor transplant models of mammary carcinoma (termed Met and Db) that display significant differences in metastatic potential. METHODS: Through suppression subtractive hybridization coupled to the microarray, we found osteopontin (OPN) to be a highly expressed gene in the tumors of the metastatic mouse model, and a lowly expressed gene in the tumors of the lowly metastatic mouse model. We further analyzed the role of OPN in this model by examining sense and antisense constructs using in vitro and in vivo methods. RESULTS: With in vivo metastasis assays, the antisense Met cells showed no metastatic tumor formation to the lungs of recipient mice, while wild-type Met cells, with higher levels of OPN, showed significant amounts of metastasis. The Db cells showed a significantly reduced metastasis rate in the in vivo metastasis assay as compared with the Met cells. Db cells with enforced overexpression of OPN showed elevated levels of OPN but did not demonstrate an increase in the rate of metastasis compared with the wild-type Db cells. CONCLUSIONS: We conclude that OPN is an essential regulator of the metastatic phenotype seen in polyomavirus middle T-induced mammary tumors. Yet OPN expression alone is not sufficient to cause metastasis. These data suggest a link between metastasis and phosphatidylinositol-3-kinase-mediated transcriptional upregulation of OPN, but additional phosphatidylinositol-3-kinase-regulated genes may be essential in precipitating the metastasis phenotype in the polyomavirus middle T model.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Sialoglicoproteínas/fisiología , Animales , Antígenos Transformadores de Poliomavirus/fisiología , Transformación Celular Neoplásica/genética , Transformación Celular Viral/genética , Femenino , Perfilación de la Expresión Génica , Biblioteca de Genes , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Trasplante de Neoplasias , Oligodesoxirribonucleótidos Antisentido/genética , Osteopontina , Fosfatidilinositol 3-Quinasas/fisiología , ARN Complementario/genética , Proteínas Recombinantes de Fusión/fisiología , Sialoglicoproteínas/genética , Técnica de Sustracción , Transfección
5.
Proc Natl Acad Sci U S A ; 101(31): 11438-43, 2004 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-15277673

RESUMEN

Imaging mouse models of human cancer promises more effective analysis of tumor progression and reduction of the number of animals needed for statistical power in preclinical therapeutic intervention trials. This study utilizes positron emission tomography imaging of 2-[18F]-fluoro-deoxy-D-glucose to monitor longitudinal development of mammary intraepithelial neoplasia outgrowths in immunocompetent FVB/NJ mice. The mammary intraepithelial neoplasia outgrowth tissues mimic the progression of breast cancer from premalignant ductal carcinoma in situ to invasive carcinoma. Progression of disease is clearly evident in the positron emission tomography images, and tracer uptake correlates with histological evaluation. Furthermore, quantitative markers of disease extracted from the images can be used to track proliferation and progression in vivo over multiple time points.


Asunto(s)
Neoplasias Mamarias Animales/diagnóstico por imagen , Tomografía Computarizada de Emisión/métodos , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fluorodesoxiglucosa F18 , Estudios Longitudinales , Glándulas Mamarias Animales/diagnóstico por imagen , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/patología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA