Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neoplasia ; 21(10): 963-973, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31437536

RESUMEN

The transcriptional coactivator Amplified in Breast Cancer 1 (AIB1) plays a major role in the progression of hormone and HER2-dependent breast cancers but its role in triple negative breast cancer (TNBC) is undefined. Here, we report that established TNBC cell lines, as well as cells from a TNBC patient-derived xenograft (PDX) that survive chemotherapy treatment in vitro express lower levels of AIB1 protein. The surviving cell population has an impaired tube-formation phenotype when cultured onto basement membrane, a property shared with TNBC cells that survive shRNA-mediated depletion of AIB1 (AIB1LOW cells). DNA analysis by exome sequencing revealed that AIB1LOW cells represent a distinct subpopulation. Consistent with their in vitro phenotype AIB1LOW cells implanted orthotopically generated slower growing tumors with less capacity for pulmonary metastases. Gene expression analysis of cultured cells and tumors revealed that AIB1LOW cells display a distinct expression signature of genes in pro-inflammatory pathways, cell adhesion, proteolysis and tissue remodeling. Interestingly, the presence of this AIB1LOW expression signature in breast cancer specimens is associated with shorter disease free survival of chemotherapy treated patients. We concluded that TNBC cell lines contain heterogeneous populations with differential dependence on AIB1 and that the gene expression pattern of AIB1LOW cells may represent a signature indicative of poor response to chemotherapy in TNBC patients.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Coactivador 3 de Receptor Nuclear/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Línea Celular Tumoral , Evolución Clonal/genética , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Ratones , Fenotipo , ARN Interferente Pequeño/genética , Transducción de Señal , Transcriptoma , Neoplasias de la Mama Triple Negativas/mortalidad , Neoplasias de la Mama Triple Negativas/patología , Secuenciación del Exoma
2.
Oncogene ; 33(23): 3033-42, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23851504

RESUMEN

The key molecular events required for the formation of ductal carcinoma in situ (DCIS) and its progression to invasive breast carcinoma have not been defined. Here, we show that the nuclear receptor coactivator amplified in breast cancer 1 (AIB1) is expressed at low levels in normal breast but is highly expressed in DCIS lesions. This is of significance since reduction of AIB1 in human MCFDCIS cells restored a more normal three-dimensional mammary acinar structure. Reduction of AIB1 in MCFDCIS cells, both before DCIS development or in existing MCFDCIS lesions in vivo, inhibited tumor growth and led to smaller, necrotic lesions. AIB1 reduction in MCFDCIS cells was correlated with significant reduction in the CD24-/CD44+ breast cancer-initiating cell (BCIC) population, and a decrease in myoepithelial progenitor cells in the DCIS lesions in vitro and in vivo. The loss of AIB1 in MCFDCIS cells was also accompanied by a loss of expression of NOTCH 2, 3 and 4, JAG2, HES1, GATA3, human epidermal growth factor receptor 2 (HER2) and HER3 in vivo. These signaling molecules have been associated with differentiation of breast epithelial progenitor cells. These data indicate that AIB1 has a central role in the initiation and maintenance of DCIS and that reduction of AIB1 causes loss of BCIC, loss of components of the NOTCH, HER2 and HER3 signaling pathways and fewer DCIS myoepithelial progenitor cells in vivo. We propose that increased expression of AIB1, through the maintenance of BCIC, facilitates formation of DCIS, a necessary step before development of invasive disease.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Células Madre Neoplásicas/fisiología , Coactivador 3 de Receptor Nuclear/metabolismo , Animales , Diferenciación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Mamarias Experimentales , Ratones , Ratones Desnudos , Células Madre Neoplásicas/patología , Coactivador 3 de Receptor Nuclear/antagonistas & inhibidores , Coactivador 3 de Receptor Nuclear/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Oncogene ; 27(22): 3186-93, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18071314

RESUMEN

We have previously shown that increased and deregulated estrogen receptor alpha expression in the mammary gland leads to the development of proliferative disease and cancer. To address the importance of cyclin D1 in ERalpha-mediated mammary tumorigenesis, we crossed ERalpha-overexpressing mice with cyclin D1 knockout mice. Mammary gland morphogenesis was completely interrupted in the ERalpha-overexpressing cyclin D1-deficient triple transgenic mice. In addition to a highly significant reduction in mammary epithelial cell proliferation, cyclin E was upregulated resulting in DNA damage checkpoint activation and apoptosis. This imbalance between proliferative and apoptotic rates in conjunction with remarkable structural defects and cellular disorganization in the terminal end buds interrupted ductal morphogenesis. Interestingly, the structure of the mammary fat pad was fundamentally altered by the consequences of overexpressing ERalpha in the epithelial cells in the absence of cyclin D1 illustrating how alterations in the epithelial compartment can impact surrounding stromal composition. Transplantation of embryonic ERalpha-overexpressing and cyclin D1-deficient mammary epithelium into the cleared fat pad of wild-type mice did not rescue the aberrant mammary gland phenotype indicating that it was intrinsic to the mammary epithelial cells. In conclusion, although cyclin D1 is not essential for proliferation of normal mammary epithelial cells, ERalpha-overexpressing cells are absolutely dependent on cyclin D1 for proliferation. This differential requirement for cyclin D1 in normal vs abnormal mammary epithelial cells supports the application of cyclin D1 inhibitors as therapeutic interventions in ERalpha-overexpressing breast cancers.


Asunto(s)
Daño del ADN/genética , Receptor alfa de Estrógeno/genética , Eliminación de Gen , Genes bcl-1 , Glándulas Mamarias Animales/crecimiento & desarrollo , Morfogénesis/genética , Animales , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular , Transformación Celular Neoplásica/genética , Ciclina E/metabolismo , Células Epiteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Transgénicos , Regulación hacia Arriba
4.
Oncogene ; 27(6): 794-802, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-17653086

RESUMEN

BRCA1 can regulate estrogen receptor-alpha (ERalpha) activity. This study tested the hypotheses that Brca1 loss in mammary epithelium alters the estrogenic growth response and that exposure to increased estrogen or ERalpha collaborates with Brca1 deficiency to accelerate preneoplasia and cancer development. Longer ductal extension was found in mammary glands of Brca1(f/f;MMTV-Cre) mice during puberty as compared to wild-type mice. Terminal end bud differentiation was impaired in Brca1 mutant mice with preservation of prolactin-induced alveolar differentiation. Exogenous estrogen stimulated an abnormal sustained increase in mammary epithelial cell proliferation and the appearance of ERalpha-negative preneoplasia in postpubertal Brca1 mutant mice. Carcinogenesis was investigated using Brca1(f/f;MMTV-Cre) mice hemizygous for p53. Exogenous estrogen increased the percentage of mice with multiple hyperplastic alveolar nodules. Targeted conditional ERalpha overexpression in mammary epithelial cells of mice that were Brca1 mutant and hemizygous for p53 increased the percentage of mice exhibiting multiple hyperplastic nodules, invasive mammary cancers and cancer multiplicity. Significantly more than half of the preneoplasia and cancers were ERalpha negative even as their initiation was promoted by ERalpha overexpression.


Asunto(s)
Proteína BRCA1/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Neoplasias Mamarias Experimentales/patología , Lesiones Precancerosas/patología , Animales , Proliferación Celular , Receptor alfa de Estrógeno/análisis , Estrógenos/farmacología , Femenino , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Mutantes , Invasividad Neoplásica , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo
5.
Breast Cancer Online ; 8(8)2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24371431

RESUMEN

Available mouse models of ductal carcinoma in situ (DCIS) and BRCA1-mutation-related breast cancer are reviewed. The best validated mouse models of human DCIS are the conditional estrogen receptor α in mammary tissue (CERM) model initiated by deregulated estrogen receptor α and the serial explant mouse model initiated by p53 deficiency. At present the most useful and best validated mouse model of BRCA1-mutation-related breast cancer uses the cre-lox system to make a conditional Brca1 deletion targeted to mammary epithelial cells. The major shortcoming of the non-conditional Brca1 models is the high incidence of non-mammary tumor development. The use of mammary gland transplants or explants from these mice into nude hosts is one approach that could be used to circumvent this deficiency. Development and validation of a Brca1-mutation-related mouse model of basal cell breast cancer is an important next step.

6.
Br J Cancer ; 91(7): 1372-9, 2004 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-15354213

RESUMEN

The expression of the proto-oncogene c-myc is frequently deregulated, via multiple mechanisms, in human breast cancers. Deregulated expression of c-myc contributes to mammary epithelial cell transformation and is causally involved in mammary tumorigenesis in MMTV-c-myc transgenic mice. c-Myc is known to promote cellular proliferation, apoptosis, genomic instability and tumorigenesis in several distinct tissues, both in vivo and in vitro. Expression of the proapoptotic regulatory gene bax is reduced or absent in human breast cancers, and c-Myc has been shown to regulate the expression of Bax, as well as cooperate with Bax in controlling apoptosis in a fibroblast model. Additionally, loss of bax reduces c-Myc-induced apoptosis in lymphoid cells and increases c-Myc-mediated lymphomagenesis in vivo. In order to assess whether loss of bax could influence c-Myc-induced apoptosis and tumorigenesis in the mammary gland in vivo, we generated MMTV-c-myc transgenic mice in which neither, one, or both wild-type alleles of bax were eliminated. Haploid loss of bax in MMTV-c-myc transgenic mice resulted in significantly reduced mammary tumour apoptosis. As anticipated for an apoptosis-regulatory gene, loss of the wild-type bax alleles did not significantly alter cellular proliferation in either mammary adenocarcinomas or dysplastic mammary tissues. However, in contrast to c-Myc-mediated lymphomagenesis, loss of one or both alleles of bax in MMTV-c-myc transgenic mice did not significantly enhance mammary tumorigenesis, despite evidence that haploid loss of bax might modestly increase mammary tumour multiplicity. Our results demonstrate that Bax contributes significantly to c-Myc-induced apoptosis in mammary tumours. In addition, they suggest that in contrast to c-Myc-induced lymphomagenesis, mammary tumorigenesis induced by deregulated c-myc expression requires some amount of Bax expression.


Asunto(s)
Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , División Celular/genética , Transformación Celular Neoplásica , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/fisiopatología , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Proto-Oncogénicas/genética , Receptores Virales/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Regulación hacia Abajo , Genes myc , Humanos , Proteínas de la Membrana/farmacología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/farmacología , Proteína X Asociada a bcl-2
7.
Genomics ; 78(3): 129-34, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11735219

RESUMEN

The signal transducers and activators of transcription (STAT) 5 and 3 are critical for mammary alveolar development during pregnancy and remodeling during involution. In the mouse, STAT3, STAT5a, and STAT5b are encoded by adjacent genes on chromosome 11 (60.5 cM). To identify additional genes in the Stat3/5 locus that may participate in normal and neoplastic development of the mammary gland, we have cloned and sequenced 500 kb and searched for genes preferentially expressed in mammary tissue. We identified six known genes and cloned two new genes, termed D11Lgp1 and D11Lgp2. Both genes are most highly expressed in normal mammary tissue and mammary tumors from several transgenic mouse models. LGP1 consists of 532 and 530 amino acids in mouse and human, respectively (88% similarity). A region in the carboxy-terminal half of LGP1 has limited homology with Arabidopsis thaliana GH3-like proteins. Immunofluorescence studies demonstrated that LGP1 is located in the nuclear envelope and the endoplasmic reticulum. LGP2 is a cytoplasmic protein of 678 amino acids.


Asunto(s)
ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/metabolismo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/genética , Proteínas de la Membrana/genética , Proteínas de la Leche , Transactivadores/genética , Secuencia de Aminoácidos , Animales , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3 , Factor de Transcripción STAT5 , Homología de Secuencia de Aminoácido , Proteínas Supresoras de Tumor
8.
Nat Genet ; 28(3): 266-71, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11431698

RESUMEN

Breast cancer is a chief cause of cancer-related mortality that affects women worldwide. About 8% of cases are hereditary, and approximately half of these are associated with germline mutations of the breast tumor suppressor gene BRCA1 (refs. 1,2). We have previously reported a mouse model in which Brca1 exon 11 is eliminated in mammary epithelial cells through Cre-mediated excision. This mutation is often accompanied by alterations in transformation-related protein 53 (Trp53, encoding p53), which substantially accelerates mammary tumor formation. Here, we sought to elucidate the underlying mechanism(s) using mice deficient in the Brca1 exon 11 isoform (Brca1Delta11/Delta11). Brca1Delta11/Delta11 embryos died late in gestation because of widespread apoptosis. Unexpectedly, elimination of one Trp53 allele completely rescues this embryonic lethality and restores normal mammary gland development. However, most female Brca1Delta11/Delta11 Trp53+/- mice develop mammary tumors with loss of the remaining Trp53 allele within 6-12 months. Lymphoma and ovarian tumors also occur at lower frequencies. Heterozygous mutation of Trp53 decreases p53 and results in attenuated apoptosis and G1-S checkpoint control, allowing Brca1Delta11/Delta11 cells to proliferate. The p53 protein regulates Brca1 transcription both in vitro and in vivo, and Brca1 participates in p53 accumulation after gamma-irradiation through regulation of its phosphorylation and Mdm2 expression. These findings provide a mechanism for BRCA1-associated breast carcinogenesis.


Asunto(s)
Apoptosis/genética , Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Genes BRCA1 , Genes p53 , Neoplasias Mamarias Animales/genética , Animales , Exones , Femenino , Genes Letales , Linfoma/etiología , Linfoma/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Animales/etiología , Ratones , Ratones Mutantes , Mutación , Neoplasias Ováricas/etiología , Neoplasias Ováricas/genética , Isoformas de Proteínas , Eliminación de Secuencia
9.
Toxicol Sci ; 62(1): 46-53, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11399792

RESUMEN

These experiments tested whether in utero and lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) alters mammary gland differentiation, estrogen receptor alpha (ERalpha) expression levels, or the response to estrogen in the female postpubertal rat mammary gland. Pregnant Holtzman rats were administered a single oral dose of 1 microg/kg TCDD or vehicle on gestation-day 15. Exposed and non-exposed female offspring were weaned on postnatal day 21 and ovariectomized at 9 weeks of age. Two weeks later, both TCDD and control animals were divided into 3 groups, receiving treatment with placebo, 0.025, or 0.1 mg 17beta-estradiol pellet implants. After 48 h, mammary tissue was removed for analysis following euthanasia. TCDD-exposed mammary glands demonstrated impaired differentiation as measured by the distribution of terminal ductal structures and increased expression levels of ERalpha. The response to exogenous estrogen was tested in TCDD-exposed animals and compared to control non-exposed animals. Estrogen stimulation of the TCDD-exposed glands induced progesterone receptor expression and mammary gland differentiation as measured by a shift in distribution from terminal end buds and terminal ducts to Types I and II lobules. Control glands were better differentiated at baseline and did not exhibit any significant changes in the distribution of terminal ductal structures following estrogen stimulation. The increase in progesterone receptor-expression levels by exogenous estrogen in control glands was similar to the TCDD-exposed glands. These experiments demonstrate that in utero and lactational exposures to TCDD impair mammary gland differentiation but that TCDD-exposed mammary glands retain the ability to differentiate in response to estrogen.


Asunto(s)
Mama/efectos de los fármacos , Antagonistas de Estrógenos/toxicidad , Lactancia/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Efectos Tardíos de la Exposición Prenatal , Receptores de Estrógenos/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Animales Recién Nacidos , Mama/crecimiento & desarrollo , Mama/metabolismo , Diferenciación Celular/efectos de los fármacos , Cartilla de ADN/química , Receptor alfa de Estrógeno , Estrógenos/farmacología , Femenino , Técnicas para Inmunoenzimas , Antígeno Ki-67/metabolismo , Ovariectomía , Embarazo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Contemp Top Lab Anim Sci ; 40(2): 27-30, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11300684

RESUMEN

Polymerase chain reaction (PCR) amplification of DNA is the most widely used technique for screening of large numbers of genetically engineered transgenic or knockout mice (Mus musculus). In this report, we present a new DNA preparation procedure for running diagnostic PCR. In this procedure, mouse ear tissue was used directly for PCR after the tissue underwent brief digestion in a solution containing only proteinase K. Using this method, we have successfully screened several lines of single, double, and triple transgenic and knockout mice. The results are reliable and reproducible. The advantage of this new method is that DNA purification by organic extraction or isolation kit was omitted. DNA purification is the limiting factor in terms of time and money when screening transgenic and knockout mice by PCR. In addition, using ear instead of tail tissue can reduce distress of animals because the samples can be obtained when the mice are labeled by ear punch.


Asunto(s)
ADN/análisis , Pruebas Genéticas/métodos , Ratones Noqueados/genética , Ratones Transgénicos/genética , Reacción en Cadena de la Polimerasa/métodos , Sistemas de Identificación Animal , Animales , Oído Externo/química , Ratones , Ratones Endogámicos C57BL , Reproducibilidad de los Resultados , Moldes Genéticos
11.
Biol Reprod ; 64(4): 1153-9, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11259262

RESUMEN

Little is known about the embryonic factors that regulate the size of the primordial follicle endowment at birth. A few studies suggest that members of the B-cell lymphoma/leukemia-2 (bcl-2) family of protooncogenes may be important determinants. Thus, the purpose of this study was to test whether bcl-2 regulates the size of the primordial follicle pool at birth. To test this hypothesis, three lines of transgenic mice (c-kit/bcl-2 mice) were generated that overexpress human bcl-2 in an effort to reduce prenatal oocyte loss. The overexpression was targeted to the ovary and appropriate embryonic time period with the use of a 4.8-kilobase c-kit promoter. This promoter provided two to three times more expression of bcl-2 in the ovaries with minimal or no overexpression in most nongonadal tissues. On Postnatal Days 8-60, ovaries were collected from homozygous c-kit/bcl-2 and nontransgenic littermates (controls) and processed for histological evaluation of follicle numbers. All lines of c-kit/bcl-2 mice were born with significantly more primordial follicles than control mice (P < or = 0.05). By Postnatal Days 30-60, however, there were no significant differences in follicle numbers between c-kit/bcl-2 and control mice. These results indicate that bcl-2 overexpression increases the number of primordial follicles at birth, but that the surfeit of primordial follicles is not maintained in postnatal life. These data suggest that it is possible that the ovary may contain a census mechanism by which excess numbers of primordial follicles at birth are detected and removed from the ovary by adulthood.


Asunto(s)
Folículo Ovárico/fisiología , Ovario/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Animales , Animales Recién Nacidos , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Transgénicos , Oocitos/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-kit/genética
12.
Cancer Res ; 60(21): 5950-3, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11085510

RESUMEN

In the present study, we examined the role of prolactin and glucocorticoids in regulating bcl-x transcription in mammary epithelial cells. We report that dexamethasone, but not prolactin, induced native bcl-x gene expression in a dose-dependent manner in HC11 cells and enhanced serum-starved HC11 cell survival. This effect was mediated through the glucocorticoid receptor and independent of STAT-5 activity. We propose that the mechanism through which glucocorticoids enhance mammary epithelial cell survival is by increasing steady-state levels of bcl-xL, RNA.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Dexametasona/farmacología , Glucocorticoides/farmacología , Glándulas Mamarias Animales/metabolismo , Proteínas de la Leche , Prolactina/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Receptores de Glucocorticoides/fisiología , Transactivadores/fisiología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/fisiología , Mifepristona/farmacología , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN/biosíntesis , ARN/genética , Receptores de Glucocorticoides/antagonistas & inhibidores , Factor de Transcripción STAT5 , Regulación hacia Arriba/efectos de los fármacos , Proteína bcl-X
14.
Oncogene ; 19(8): 1010-9, 2000 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-10713684

RESUMEN

Understanding the process of carcinogenesis is key to developing therapies which might interrupt or reverse tumor onset and progression. Cell growth and death signals are dependent not only upon molecular mechanisms within a cell but also upon external stimuli such as hormones, cell - cell signaling, and extracellular matrix. Mouse models can be used to dissect these complex processes, to identify key signaling pathways operating at different stages of tumorigenesis, and to test the strength of specific interventions. In the WAP-TAg mouse model, carcinogenesis is initiated by expression of the Simian Virus 40 T antigen (TAg). TAg expression is triggered by hormonal stimulation, either during estrus or pregnancy. Breast adenocarcinomas (ranging from well to poorly differentiated) develop in 100% of the female mice by approximately 8 - 9 months of age. Three distinct stages of tumorigenesis are easily identified: an initial proliferation, hyperplasia, and adenocarcinoma. The mean time to first palpable tumor in mice which undergo at least one pregnancy is 6 months. The tumorigenic process is marked by a competition between proliferation and apoptosis and is characterized by cellular acquisition of genetic mutations and increased stromal fibrosis. Protein levels of cell cycle control genes cyclin D1, cdk2, and E2F-1 are increased in these adenocarcinomas. c-Fos protein levels are slightly increased in these cancers, while c-Jun levels do not change. Hormonal exposure alters progression. Estrogen plays a role during the early stages of oncogenesis although the growth of the resulting adenocarcinomas is estrogen-independent. Transient hormonal stimulation by glucocorticoids that temporarily increases the rate of cell proliferation results in tetraploidy, premature appearance of irreversible hyperplasia, and early tumor development. Tumor appearance also can be accelerated through over expression of the cell survival protein, Bcl-2. Bcl-2 over expression not only reduces apoptosis during the initial proliferative process but also decreases the total rate of cell proliferation. This block in cell proliferation is lost selectively as the cells transition to adenocarcinoma. The WAP-TAg model can be utilized to investigate how the basic processes of cell proliferation, apoptosis, DNA mutation, and DNA repair are modified by external and internal signals during mammary oncogenesis.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Hormonas/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Mutación , Proteínas Proto-Oncogénicas c-bcl-2/genética , Animales , Mama/crecimiento & desarrollo , Mama/metabolismo , División Celular/genética , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Estrógenos/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Humanos , Hiperplasia/inducido químicamente , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Transgénicos , Mitosis , Embarazo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2
15.
Clin Obstet Gynecol ; 43(4): 821-9, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11100299

RESUMEN

In conclusion, several studies indicate that there is an association between cigarette smoking and adverse reproductive outcomes in women as well as men. Some studies indicate that alcohol consumption impairs the reproductive capacity of women. Exposures to PCE in the dry cleaning industry, toluene in the printing business, ethylene oxide and mixed solvents have been associated with decreased fecundity. Abnormalities in sperm production have been found in men exposed to radiant heat or heavy metals. Environmental exposure to chlorinated hydrocarbons (e.g., DDT, PCB, pentachlorophenol, hexachlorocyclohexane) has been associated with an increase in rates of miscarriage and endometriosis. Clinicians should counsel patients who are trying to achieve a successful pregnancy to stop smoking and limit alcohol intake. Clinicians can additionally counsel patients who are in contact with potentially harmful occupational and environmental toxicants to limit their exposure. It is important to recognize, however, that many of the studies to date are limited by small sample size, poor exposure assessment, poor outcome measurements, recruitment bias, or recall bias. Additional studies will be necessary to clarify the magnitude of risk associated with these factors.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Infertilidad Femenina/etiología , Infertilidad Masculina/etiología , Consumo de Bebidas Alcohólicas/efectos adversos , Femenino , Humanos , Masculino , Exposición Profesional/efectos adversos , Embarazo , Fumar/efectos adversos
16.
Oncogene ; 19(52): 6053-8, 2000 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-11146557

RESUMEN

Maspin is a unique serpin involved in the suppression of tumor growth and metastasis. To investigate whether increased levels of maspin protect against tumor progression in vivo, we established a transgenic model in which maspin is targeted to mammary epithelial cells by the Whey Acidic Protein (WAP) promoter for overexpression. We crossed these WAP-maspin transgenic mice with the WAP-TAg mouse model of tumor progression. Maspin overexpression increased the rate of apoptosis of both preneoplastic and carcinomatous mammary epithelial cells. Maspin reduced tumor growth through a combination of reduced angiogenesis and increased apoptosis. The number of pulmonary metastases was reduced in the presence of maspin overexpression. These data demonstrate that targeted overexpression of maspin can inhibit tumor progression in vivo, likely through a combination of increased apoptosis, decreased angiogenesis, and inhibition of tumor cell migration.


Asunto(s)
Adenocarcinoma/patología , Antígenos Virales de Tumores/metabolismo , Progresión de la Enfermedad , Neoplasias Mamarias Animales/patología , Proteínas de la Leche/genética , Proteínas/metabolismo , Serpinas/metabolismo , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/genética , Animales , Antígenos Virales de Tumores/genética , Apoptosis , División Celular , Movimiento Celular , Supervivencia sin Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Expresión Génica , Genes Supresores de Tumor , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica , Ensayos de Protección de Nucleasas , Lesiones Precancerosas/irrigación sanguínea , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Regiones Promotoras Genéticas/genética , Proteínas/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Serpinas/genética , Transgenes/genética
17.
Oncogene ; 18(47): 6589-96, 1999 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-10597263

RESUMEN

Bcl-2 is an anti-apoptotic and anti-proliferative protein over-expressed in several different human cancers including breast. Gain of Bcl-2 function in mammary epithelial cells was superimposed on the WAP-TAg transgenic mouse model of breast cancer progression to determine its effect on epithelial cell survival and proliferation at three key stages in oncogenesis: the initial proliferative process, hyperplasia, and cancer. During the initial proliferative process, Bcl-2 strongly inhibited both apoptosis and mitotic activity. However as tumorigenesis progressed to hyperplasia and adenocarcinoma, the inhibitory effects on mitotic activity were lost. In contrast, anti-apoptotic activity persisted in both hyperplasias and adenocarcinomas. These results demonstrate that the inhibitory effect of Bcl-2 on epithelial cell proliferation and apoptosis can separate during cancer progression. In this model, retention of anti-apoptotic activity with loss of anti-proliferative action resulted in earlier tumor presentation.


Asunto(s)
Adenocarcinoma/patología , Neoplasias Mamarias Experimentales/patología , Mitosis/genética , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Adenocarcinoma/genética , Animales , Modelos Animales de Enfermedad , Humanos , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-bcl-2/genética
20.
J Mammary Gland Biol Neoplasia ; 4(2): 153-64, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10426394

RESUMEN

The Bcl-2 gene family regulates tissue development and tissue homeostasis through the interplay of survival and death factors. Family members are characterized as either pro-apoptotic or anti-apoptotic, depending on cellular context. In addition to its anti-apoptotic effect, Bcl-2 also inhibits progression through the cell cycle. Functional interactions between family members as well as binding to other cellular proteins modulate their activities. Mammary gland tissue, similar to many other tissues, expresses a number of different Bcl-2 relatives including bcl-x, bax, bak, bad, bcl-w, bfl-1, bcl-2 as well as the bcl-2 binding protein Bag-1. Bcl-2 is expressed in the nonpregnant mammary gland and early pregnancy. In contrast, expression of bcl-x and bax continues through late pregnancy, is down-regulated during lactation, and upregulated with the start of involution. Bak, bad, bcl-w, and bfl-1 are also up-regulated during involution. The specific roles of individual gene products are investigated using dominant gain of function and loss of function mice. Finally, different Bcl-2 family members are commonly over- or under-expressed in human breast cancers. Bcl-2 expression in human breast cancers has been associated with a good prognosis, while decreased Bax expression has been linked to poor clinical outcome. Understanding the role Bcl-2 family members play in regulating mammary epithelial cell survival is salient to both normal mammary gland physiology and the development of new therapeutic approaches to breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Mama/fisiología , Genes bcl-2 , Glándulas Mamarias Animales/fisiología , Familia de Multigenes , Animales , Apoptosis , Mama/citología , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Neoplasias de la Mama/terapia , Supervivencia Celular , Femenino , Regulación de la Expresión Génica , Humanos , Lactancia/fisiología , Glándulas Mamarias Animales/citología , Ratones , Embarazo , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteínas Proto-Oncogénicas c-bcl-2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA