Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-18540570

RESUMO

Estrogen and progesterone play a critical role in normal and neoplastic development of the mammary gland. A long duration of estrogen and progesterone exposure is associated with increased breast cancer risk, and a short duration of the same doses of these hormones is associated with a reduced breast cancer risk. The protective effects of estrogen and progesterone have been extensively studied in animal models. Several studies have demonstrated that these hormones induce persistent and long-lasting alterations in gene expression in the mammary epithelial cells. In the experiments discussed herein, the protective effect of estrogen and progesterone is shown to occur in genetically engineered mice (the p53-null mammary gland). The protective effect is associated with a decrease in cell proliferation. The effects of hormones seem to manifest as a delay in premalignant progression. In the nontumor-bearing glands of hormone-treated mice, premalignant foci are present at the time the control glands are actively developing mammary tumors. If the hormone-treated cells are transplanted from the treated host to the untreated host, the cells resume their predetermined tumorigenic potential. The protective effect reflects both host-mediated factors (either stroma-determined or systemic factors) and mammary epithelial intrinsic changes. If normal, untreated p53 cells are transplanted into a host that has been previously treated with a short dose of hormones, the cells exhibit a significant delay in tumorigenesis. The relative contributions of host-mediated factors and mammary cell intrinsic factors remain to be determined. Current studies are moving this research area from the biological to the molecular realm and from the rodent models to human studies and offer the potential for directing prevention efforts at specific molecular targets.


Assuntos
Estrogênios/fisiologia , Genes p53 , Engenharia Genética , Neoplasias Mamárias Experimentais/prevenção & controle , Progesterona/fisiologia , Animais , Perfilação da Expressão Gênica , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Knockout
2.
Mol Carcinog ; 45(7): 490-505, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16482517

RESUMO

The inappropriate activation of one or more members of the ErbB family of receptor tyrosine kinases [ErbB-1 (EGFR), ErbB-2, ErbB-3, ErbB-4] has been linked with oncogenesis. ErbB-2 is frequently coexpressed with ErbB-3 in breast cancer cells and in the presence of the ligand heregulin (HRG) the ErbB-2/ErbB-3 receptors form a signaling heterodimer that can affect cell proliferation and apoptosis. The major goal of the present study was to determine whether endogenous HRG causes autocrine/paracrine activation of ErbB-2/ErbB-3 and contributes to the proliferation of mammary epithelial tumor cells. Tyrosine-phosphorylated (activated) ErbB-2 and ErbB-3 receptors were detected in the majority of extracts from tumors that had formed spontaneously or as a result of oncogene expression. HRG-1 transcripts and protein were found in the epithelial cells of most of these mouse mammary tumors. Various mouse mammary cell lines also contained activated ErbB-2/ErbB-3 and HRG transcripts. A approximately 50 kDa C-terminal fragment of pro-HRG was detected, which indicates that the HRG-1 precursor is readily processed by these cells. It is likely that the secreted mature HRG activated the ErbB-2/3 receptors. Addition of an antiserum against HRG to the mammary epithelial tumor cell line TM-6 reduced ErbB-3 Tyr-phosphorylation. Treatment with HRG-1 siRNA oligonucleotides or infection with a retroviral construct to stably express HRG siRNA effectively reduced HRG protein levels, ErbB-2/ErbB-3 activation, and the rate of proliferation, which could be reversed by the addition of HRG. The cumulative findings from these experiments show that coexpression of the HRG ligand contributes to activation of ErbB-2/Erb-3 in mouse mammary tumor cells in an autocrine or paracrine fashion.


Assuntos
Neoplasias Mamárias Experimentais/genética , Neuregulina-1/genética , Receptor ErbB-2/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Primers do DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Camundongos , Neuregulina-1/metabolismo , Fragmentos de Peptídeos/química , Plasmídeos , Reação em Cadeia da Polimerase , RNA Interferente Pequeno/genética , Receptor ErbB-2/genética , Proteínas Recombinantes/metabolismo , Transfecção
3.
Oncogene ; 20(52): 7645-57, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11753642

RESUMO

Development of strategies for prevention of breast cancer development requires an understanding of the effects of mammary oncogenes on mammary cells at early stages in neoplastic transformation. As mammary oncogenes wnt-1 and int-2 affect different signal transduction pathways, we investigated their effects on established mouse mammary epithelial cell lines (MMECLs) reflecting early stages in tumorigenesis. Normal interactions between beta-catenin and E-cadherin were abrogated in all three immortalized MMECLs and the cells lacked beta-catenin-mediated transactivation activity, detectable using a reporter assay, suggesting that alterations in cell adhesion may be very early events in mammary tumorigenesis. Immortalized FSK4 and EL12 cells and hyperplastic TM3 cells were stably transfected with expression vectors encoding wnt-1 or int-2 or the control vector, and drug-selected pooled cells from each line were confirmed by reverse transcription-polymerase chain reaction to express the transfected oncogene; this expression persisted in the cells analysed in vitro and in vivo. Resultant phenotypic changes depended both on the oncogene and the target mammary cell line. In FSK4 cells, expression of wnt-1 or int-2 resulted in proliferative changes in vitro, including reduced contact inhibition, increased beta-catenin expression, and decreased p53 transcriptional activity, but neither oncogene conferred upon those cells the ability to produce tumors in vivo. EL12 cells were highly refractory to the effects of both oncogenes, with the only measurable changes being increased E-cadherin levels induced by both oncogenes and increased proliferation of the int-2-transfected cells in the absence of serum. Parental TM3 cells were phenotypically similar to wnt-1- or int-2-transfected FSK4 cells and displayed an increased rate of proliferation in vitro and markedly increased tumorigenicity in vivo following transfection with int-2 but not with wnt-1. These results suggest that wnt-1 signaling is redundant in the hyperplastic TM3 cells and indicate that wnt-1-induced effects in the immortalized FSK4 and EL12 cells were not sufficient to mediate a tumorigenic phenotype. This study showed that the wnt-1 and int-2 oncogenes have similar but distinguishable effects on immortalized MMECLs and that the genetic background of the mammary cells greatly influences the consequences of oncogene expression at early stages of cell transformation.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Transativadores , Proteínas de Peixe-Zebra , Animais , Caderinas/metabolismo , Adesão Celular , Divisão Celular , Linhagem Celular , Meios de Cultura Livres de Soro , Células Epiteliais/citologia , Feminino , Fator 3 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/genética , Expressão Gênica , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes , Transcrição Gênica , Ativação Transcricional , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Proteínas Wnt , Proteína Wnt1 , beta Catenina
4.
Cancer Res ; 61(18): 6945-51, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11559574

RESUMO

Maspin is a unique serine protease inhibitor of which the down-regulation is associated with the development of breast cancers. In vitro, recombinant maspin inhibits tumor cell migration and invasion. Overexpression of maspin in transgenic mice is protective against tumor progression. Additionally, maspin acts as an angiogenesis inhibitor in rat cornea model and in a xenograft tumor model. To additionally prove that maspin is directly involved in the suppression of tumor growth and metastasis, we tested maspin in a new syngeneic mammary tumor model, TM40D. This model involves the implantation of TM40D mammary tumor cells orthotopically to the mammary gland; tumors grew within the gland and then become invasive and metastatic to other organs. Here we demonstrate that TM40D cells in implanted mammary glands are highly invasive. Overall, a 75% rate of invasion and metastasis was observed in this model. However, both primary tumor growth and metastasis were significantly blocked in TM40D cells that overexpress maspin as a consequence of plasmid or retrovirus infection. Maspin-transfected tumors tended to have tumor encapsulation and less necrosis, which were associated with better prognosis and lower invasiveness. Thus, maspin can block primary tumor growth as well as invasion and metastasis. These data support the concept that maspin has a strong protective role against tumor progression.


Assuntos
Neoplasias Mamárias Experimentais/patologia , Proteínas/fisiologia , Serpinas/fisiologia , Animais , Divisão Celular/fisiologia , Modelos Animais de Doenças , Feminino , Genes Supressores de Tumor , Humanos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Biossíntese de Proteínas , Proteínas/genética , Serpinas/biossíntese , Serpinas/genética , Transfecção
5.
Cell Growth Differ ; 12(6): 285-95, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11432803

RESUMO

The level of circulating ovarian hormones (estrogen and progesterone) alone or in combination with pituitary hormones have a potent mitogenic impact in the normal mammary gland, and they also play a pivotal role in the development and progression of mammary carcinoma. The differential effects of hormones on the molecular components of cyclin-dependent kinase (cdk) complexes in mammary epithelium of the hormone-dependent ductal outgrowth line, EL11, and the hormone-independent alveolar outgrowth line, TM2L, were the focus of this study. The two outgrowth lines, which represent early stages in mammary hyperplasia, were compared with normal mammary gland at different hormonal conditions: control, hormone stimulated by pituitary isograft, and hormone depleted by ovariectomy. Hormonal stimulation by a pituitary isograft resulted in DNA synthesis and lobuloalveolar development of normal mammary ducts, DNA synthesis but no lobuloalveolar development in the EL11 ductal outgrowth, and no changes either in DNA synthesis or in lobuloalveolar morphology in the TM2L outgrowth. The levels of cdk4- and cyclin D1-associated kinase activities were correlated with cell proliferation in only the alveolar phenotypes (i.e., in only hormonally stimulated normal virgin gland and in alveolar mammary outgrowth), whereas cyclin D2-dependent kinase activity was correlated with cell proliferation in only the alveolar preneoplasia. p16(INK4a) and p21(Cip1) protein levels were decreased at the earliest stages of preneoplasia, i.e., at immortalization, and were independent from changes in cyclin D1, which occurred later in preneoplasia. Although all cdk inhibitors changed in concordance with hormonal status reflected by proliferation levels, p27(Kip1) was the only cdk inhibitor that was up-regulated at the earliest stages of preneoplasia and may have a unique role in blocking alveolar differentiation in response to the loss of one or more of the cell cycle-negative regulators. We hypothesize that up-regulation of p27(Kip1) prevents immortalized ductal outgrowths (EL11) from progressing to the neoplastic state, even under hormonal stimulation.


Assuntos
Mama/patologia , Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Lesões Pré-Cancerosas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Proteínas Supressoras de Tumor , Animais , Mama/crescimento & desenvolvimento , Mama/metabolismo , Ciclina D1/metabolismo , Ciclina D2 , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Células Epiteliais/metabolismo , Feminino , Hormônios/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ovariectomia , Lesões Pré-Cancerosas/metabolismo , Proteínas/metabolismo
6.
Oncogene ; 20(32): 4409-11, 2001 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-11466622

RESUMO

Ataxia-telangiectasia (AT) is a genetic syndrome resulting from the inheritance of two defective copies of the ATM gene that includes among its stigmata radiosensitivity and cancer susceptibility. Epidemiological studies have demonstrated that although women with a single defective copy of ATM (AT heterozygotes) appear clinically normal, they may never the less have an increased relative risk of developing breast cancer. Whether they are at increased risk for radiation-induced breast cancer from medical exposures to ionizing radiation is unknown. We have used a murine model of AT to investigate the effect of a single defective Atm allele, the murine homologue of ATM, on the susceptibility of mammary epithelial cells to radiation-induced transformation. Here we report that mammary epithelial cells from irradiated mice with one copy of Atm truncated in the PI-3 kinase domain were susceptible to radiation-induced genomic instability and generated a 10% incidence of dysplastic mammary ducts when transplanted into syngenic recipients, whereas cells from Atm(+/+) mice were stable and formed only normal ducts. Since radiation-induced ductal dysplasia is a precursor to mammary cancer, the results indicate that AT heterozygosity increases susceptibility to radiogenic breast cancer in this murine model system.


Assuntos
Neoplasias da Mama/genética , Carcinoma Intraductal não Infiltrante/genética , Proteínas Serina-Treonina Quinases/genética , Radiação Ionizante , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Mama/patologia , Mama/efeitos da radiação , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/metabolismo , Carcinoma Intraductal não Infiltrante/patologia , Proteínas de Ciclo Celular , Células Cultivadas , Quebra Cromossômica , Proteínas de Ligação a DNA , Células Epiteliais/efeitos da radiação , Feminino , Genoma , Heterozigoto , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/metabolismo , Tolerância a Radiação , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
7.
Am J Pathol ; 157(6): 2151-9, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11106587

RESUMO

Breast cancer is the most frequent tumor type among women in the United States and in individuals with Li-Fraumeni syndrome. The p53 tumor suppressor gene is altered in a large proportion of both spontaneous breast malignancies and Li-Fraumeni breast cancers. This suggests that loss of p53 can accelerate breast tumorigenesis, yet p53-deficient mice rarely develop mammary tumors. To evaluate the effect of p53 loss on mammary tumor formation, the p53(null) allele was back-crossed onto the BALB/c genetic background. Median survival was 15.4 weeks for BALB/c-p53(-/-) mice compared to 54 weeks for BALB/c-p53(+/-) mice. Sarcomas and lymphomas were the most frequent tumor types in BALB/c-p53(-/-) mice, whereas 55% of the female BALB/c-p53(+/-) mice developed mammary carcinomas. The mammary tumors were highly aneuploid, frequently lost the remaining wild-type p53 allele, but rarely lost BRCA1. Although mammary tumors were rarely detected in BALB/c-p53(-/-) female mice, when glands from BALB/c-p53(-/-) mice were transplanted into wild-type BALB/c hosts, 75% developed mammary tumors. The high rate of mammary tumor development in the BALB/c background, but not C57Bl/6 or 129/Sv, suggests a genetic predisposition toward mammary tumorigenesis. Therefore, the BALB/c-p53(+/-) mice provide a unique model for the study of breast cancer in Li-Fraumeni syndrome. These results demonstrate the critical role that the p53 tumor suppressor gene plays in preventing tumorigenesis in the mammary gland.


Assuntos
Heterozigoto , Síndrome de Li-Fraumeni/genética , Neoplasias Mamárias Animais/genética , Camundongos Endogâmicos BALB C/genética , Proteína Supressora de Tumor p53/genética , Animais , Modelos Animais de Doenças , Feminino , Deleção de Genes , Genes BRCA1/genética , Incidência , Masculino , Neoplasias Mamárias Animais/epidemiologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos , Fenótipo , Análise de Sobrevida , Proteína Supressora de Tumor p53/deficiência
8.
FASEB J ; 14(14): 2221-9, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11053243

RESUMO

Mammary epithelial cells from p53 null mice have been shown recently to exhibit an increased risk for tumor development. Hormonal stimulation markedly increased tumor development in p53 null mammary cells. Here we demonstrate that mammary tumors arising in p53 null mammary cells are highly aneuploid, with greater than 70% of the tumor cells containing altered chromosome number and a mean chromosome number of 56. Normal mammary cells of p53 null genotype and aged less than 14 wk do not exhibit aneuploidy in primary cell culture. Significantly, the hormone progesterone, but not estrogen, increases the incidence of aneuploidy in morphologically normal p53 null mammary epithelial cells. Such cells exhibited 40% aneuploidy and a mean chromosome number of 54. The increase in aneuploidy measured in p53 null tumor cells or hormonally stimulated normal p53 null cells was not accompanied by centrosome amplification. These results suggest that normal levels of progesterone can facilitate chromosomal instability in the absence of the tumor suppressor gene, p53. The results support the emerging hypothesis based both on human epidemiological and animal model studies that progesterone markedly enhances mammary tumorigenesis.


Assuntos
Aneuploidia , Células Epiteliais/efeitos dos fármacos , Glândulas Mamárias Animais/efeitos dos fármacos , Progesterona/farmacologia , Proteína Supressora de Tumor p53/genética , Animais , Células Cultivadas , Centrossomo/efeitos dos fármacos , Centrossomo/metabolismo , Células Epiteliais/metabolismo , Feminino , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Mutação , Células Tumorais Cultivadas
9.
Oncogene ; 19(8): 1052-8, 2000 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-10713689

RESUMO

Although alterations in the p53 tumor suppressor gene are detected frequently in human breast cancers, mammary tumors are observed infrequently in p53(null) mice. This has led to the suggestion that absence of p53 alone is not sufficient for induction of mammary tumors. However, early death of p53(null) mice from thymic lymphomas may obscure tumor phenotypes that would develop later. Therefore, p53(null) mammary epithelium was transplanted into cleared mammary fat pads of wild type p53 BALB/c hosts to allow long-term analysis of mammary tumor phenotypes. Five treatments were compared for their effects on tumor incidence in hosts bearing transplants of p53(null) and p53wt mammary epithelium. The treatment groups were: (1) untreated; (2) continuous hormone stimulation with pituitary isografts; (3) multiple pregnancies; (4) DMBA alone; and (5) DMBA+pituitary isografts. The tumor incidences in p53(null) vs p53wt mammary transplants for each treatment group were 62% vs 0%, 100% vs 0%, 68% vs 0%, 60% vs 4% and 91% vs 14%, respectively. The mammary tumors that developed in the p53(null) mammary epithelium were all adenocarcinomas and were frequently aneuploid. These data demonstrate that the absence of p53 is sufficient to cause development of mammary tumors and that hormonal stimulation enhances the tumorigenicity of p53(null) mammary epithelium to a greater extent than DMBA exposure alone. This model provides an in situ approach to examine the molecular basis for the role of p53 in the regulation of mammary tumorigenesis.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/patologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Proteína Supressora de Tumor p53/genética , 9,10-Dimetil-1,2-benzantraceno , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/epidemiologia , Aneuploidia , Animais , Carcinógenos , Modelos Animais de Doenças , Células Epiteliais/patologia , Células Epiteliais/transplante , Feminino , Genes Supressores de Tumor , Incidência , Neoplasias Mamárias Experimentais/epidemiologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Hipófise/metabolismo , Gravidez
10.
Oncogene ; 18(47): 6597-604, 1999 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-10597264

RESUMO

Bcl-2 is known to have dual antiproliferative and antiapoptotic roles. Overexpression of Bcl-2 in the mammary gland using a whey acidic protein (WAP) promoter-driven Bcl-2 transgene inhibits apoptosis in the mammary gland during pregnancy, lactation, and involution, and also counteracts apoptosis induced by overexpression of a mutant p53 transgene (WAP-p53 172 R-L). WAP-Bcl-2 mice and nontransgenic controls were treated with the carcinogen dimethylbenz(a)anthracene (DMBA). Surprisingly, the nontransgenic mice developed mammary tumors with decreased latency. Tumors arising in WAP-Bcl-2 mice displayed substantially reduced levels of proliferation relative to those seen in nontransgenic mice (P < 0.015), perhaps resulting in the observed increase in tumor latency following carcinogen treatment. This WAP-Bcl-2 mouse tumor model reflects the situation seen in some human breast cancers overexpressing Bcl-2, where expression of Bcl-2 has been shown to correlate with a lower proliferative index in tumors.


Assuntos
9,10-Dimetil-1,2-benzantraceno/toxicidade , Carcinógenos/toxicidade , Neoplasias Mamárias Experimentais/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Animais , Apoptose , Divisão Celular/genética , Genes p53 , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Proteínas do Leite/genética , Transgenes
11.
Carcinogenesis ; 20(9): 1715-20, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10469615

RESUMO

We have developed an in vivo model system of mouse mammary preneoplasias in order to examine the cell and molecular changes that occur during tumorigenesis. Most of these preneoplasias are characterized by an alveolar hyperplasia morphologically similar to that present in normal pregnant mammary gland, but have tumor forming capabilities ranging from very low to high. One of these hyperplasias, the TM3 HOG (transformed mammary hyperplastic outgrowth), forms tumors infrequently and has the unusual characteristic of spontaneous regression. We have observed that 7-8 months post-transplantation into the cleared mammary fat pad of a BALB/c mouse, the TM3 hyperplasia will begin to regress, leaving only a sparse ductal tree with remnant alveolar structures by 10-12 months post-transplantation. We have sought to elucidate the mechanism of this regression by determining the apoptotic and proliferative rates of the alveolar cells during TM3 HOG development. Studies show that apoptotic rates in the TM3 HOG are consistently high (4-7%) at all times after transplantation. This apoptotic rate is higher than the rates found in other preneoplasias in our system and approach the rates observed in the normal involuting gland. An unusual p53 mutation, a serine insertion at codon 233, may be causally related to the high spontaneous apoptotic frequencies as well as elevated inducible apoptotic frequencies in TM3. In addition, a sudden decrease ( approximately 63%) in proliferation occurs around 8 months post-transplantation. Furthermore, transplantation experiments indicate that the ability of the 8-month-old host and/or mammary gland to support growth of preneoplastic mammary tissues is markedly diminished compared with 3- or 6-month-old hosts. The results presented here suggest that the persistent high apoptotic rates, concomitant with decreased proliferation rates, may be responsible for TM3's regression and implicate a unique mutant p53 as a causal factor. Additionally, the results suggest that host determinants can interact with specific molecular changes in the preneoplastic cells to influence growth and progression of the preneoplastic populations.


Assuntos
Apoptose/genética , Genes p53 , Glândulas Mamárias Animais/patologia , Lesões Pré-Cancerosas/genética , Fatores Etários , Animais , Apoptose/efeitos da radiação , Divisão Celular , Códon/genética , Feminino , Raios gama , Hiperplasia , Glândulas Mamárias Animais/transplante , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Lesões Pré-Cancerosas/patologia , Remissão Espontânea , Proteína Supressora de Tumor p53/fisiologia
12.
Cancer Res ; 59(17): 4276-84, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10485472

RESUMO

To define the functional relevance of progesterone-initiated intracellular signaling in mammary gland tumorigenesis, the progesterone receptor knockout (PRKO) mouse model was used in the context of an established carcinogen-induced mammary tumorigenesis system. In carcinogen-treated, 7,12-dimethylbenz(a)anthracene (DMBA), pituitary-isografted mice, there was a marked reduction in mammary tumor incidence in PRKO mice as compared with isogenic wild types (WT). Mammary tumors arose in 12 (60%) of 20 WT mice compared with 3 (15%) of 20 PRKO mice by 44 weeks after the initial DMBA treatment. In the absence of a pituitary isograft, mammary tumors developed in 4 (20%) of 20 WT mice versus 4 (20%) of 20 PRKO mice by 47 weeks. At the time of carcinogen administration, the proliferative index of the pituitary-stimulated WT gland was at least 4-fold higher than similarly treated PRKO glands, supporting the importance of PR-mediated proliferative pathways in the genesis of this tumor type. Unlike the WT gland, the PRKO gland was unable to exhibit alveologenesis in response to pituitary isograft stimulation; thus, DMBA-initiated mammary tumors observed in the PRKO were assumed to be exclusively of ductal origin. Compared with previous tested strains, by 47 weeks, a higher incidence of DMBA-induced ovarian tumors was observed in this mouse strain: (a) 4 (20%) of 20 WT mice and 9 (45%) of 20 PRKO mice with a pituitary isograft; and (b) 10 (50%) of 20 WT mice and 10 (50%) of 20 PRKO mice without a pituitary isograft. Despite the host-strain's underlying propensity for DMBA-induced ovarian neoplasms, our studies underscore the specific importance of the PR (as distinct from the estrogen receptor) as a mandatory mediator for those intracellular signaling pathways that are essential for the initiation of the majority of murine mammary tumors induced by DMBA. Apart from providing strong support for progesterone's role in mammary gland tumorigenesis as well as furthering our fundamental understanding of breast cancer etiology, these studies may have implications for the routine use of progestins.


Assuntos
Neoplasias Mamárias Experimentais/etiologia , Receptores de Progesterona/fisiologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Progesterona/genética
13.
Mol Carcinog ; 14(2): 75-83, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7576102

RESUMO

Murine p53 containing an Arg-->Leu substitution at amino acid 172 possesses many properties characteristic of wild-type p53, including the ability to induce p21/WAF/Cip1 and apoptosis. To determine if p53-dependent apoptosis plays a critical role in mammary tumorigenesis, transgenic mice were generated in which the expression of this mutant p53 protein was targeted to the mammary gland by using the rat whey acidic protein gene promoter. Mice bearing pituitary isografts were treated with 7,12-dimethylbenz[a]anthracene (DMBA) and examined for mammary tumor development. Mice overexpressing the p53 transgene exhibited a statistically significant increase in apoptosis in the mammary gland and a statistically significant decrease in the incidence of DMBA-induced mammary tumors. No difference in tumor incidence was observed in mice without pituitary isografts who were treated with DMBA, because the transgene is not overexpressed in the absence of hormone stimulation provided by the pituitary isograft. The unexpected wild-type properties of the 172Arg-->Leu mutant p53, including its ability to stimulate apoptosis, make it a possible candidate for use in gene therapy protocols.


Assuntos
Apoptose , Genes p53 , Neoplasias Mamárias Experimentais/genética , Proteína Supressora de Tumor p53/genética , 9,10-Dimetil-1,2-benzantraceno , Animais , Replicação do DNA , Feminino , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Gravidez , Regiões Promotoras Genéticas , Ratos , Ativação Transcricional , Proteína Supressora de Tumor p53/fisiologia
14.
Cancer Res ; 53(14): 3374-81, 1993 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8324748

RESUMO

Preneoplastic lesions in the mammary gland represent a population of cells at increased risk of progression to tumors. Because p53 is the most commonly mutated gene in human breast cancer, we sought to determine whether mutations in p53 were present in preneoplastic lesions or were acquired during progression to overt tumors. In the mouse mammary gland, hyperplastic alveolar nodules (HAN) are the most common preneoplastic lesion. Analysis of the TM series of transplantable murine HAN outgrowths and tumors allowed the status of p53 to be determined at distinct stages of mammary tumorigenesis. Alterations in the p53 gene or in the pattern of p53 protein expression were observed in all five HAN outgrowth lines examined. Altered expression of p53 protein was detected in 3 of 5 TM HAN outgrowth lines as determined by immunohistochemistry. Overexpression of nuclear p53 was detected in only a fraction of the cells (10-50%) in TM3 and TM4 HAN outgrowths, whereas in tumors that arose from TM4 HAN outgrowths, the proportion of cells overexpressing p53 protein approached approximately 100%. Despite overexpression of p53 in TM3 HAN outgrowths, no tumors have developed in this line. The TM9 outgrowth line exhibited a different pattern of p53 expression by immunohistochemistry: p53 protein was overexpressed in the cytoplasm of virtually all cells in the HAN outgrowths but was localized to the nuclei of TM9 tumor cells. Direct sequencing of p53 transcripts from tumors and cell lines revealed various genetic changes: point mutations in exons 4 and 5 (TM2H, nonsense; TM4, missense); a deletion in exon 5 (TM4); and an insertion in exon 7 (TM3). Although p53 protein was overexpressed in TM9 tumors, it was shown to be wild-type both by immunoprecipitation and direct sequencing of the entire coding region of the cDNA. TM4 cells were homogeneous with respect to mutant p53 genotype and uniformly expressed p53 by immunohistochemical staining in vitro, but transplantation of TM4 cells to fat pads of BALB/c hosts resulted in HAN outgrowths in situ in which < 50% of the cells expressed the mutant p53 at detectable levels. In summary, mutation of the p53 gene and overexpression of p53 protein can occur in preneoplastic mammary epithelial cells, and those mutations are maintained in tumors that arise from the HAN. Conversely, expression of mutant p53 was decreased when cells were grown in situ, implicating the presence of cellular factors that can suppress p53 expression in vivo. These observations demonstrate that the p53 pathway may be a common target for mutation in murine mammary tumorigenesis.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Genes p53/genética , Neoplasias Mamárias Animais/genética , Mutação/genética , Lesões Pré-Cancerosas/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Sequência de Bases , Núcleo Celular/metabolismo , Feminino , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Lesões Pré-Cancerosas/patologia , Células Tumorais Cultivadas
15.
Cancer Res ; 53(7): 1646-52, 1993 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8453637

RESUMO

Breast cancer is a consequence of multiple alterations occurring over a long period of time. Genetic changes in early stages of tumorigenesis have not been defined. A recently developed murine system permits the study of mammary preneoplastic cells in vivo and in vitro (F. S. Kittrell et al., Cancer Res., 52: 1924-1932, 1992). To assess the potential role of p53 mutations in early stages of breast cancer, the status of p53 was determined in a series of mouse mammary epithelial cell lines which give rise to preneoplastic outgrowths (hyperplastic alveolar nodules) when transplanted into cleared mammary fat pads of syngeneic mice. Protein stability and conformation were analyzed using immunoprecipitations and immunochemical assays; p53 transcripts were sequenced using a polymerase chain reaction approach. The parental cell lines (FSK lines) showed no evidence of p53 alterations at either the protein or the nucleic acid level, indicating that p53 mutations are not essential for the establishment of mammary epithelial cell lines in vitro. In contrast, cell lines (TM lines) derived from hyperplastic alveolar nodule outgrowths induced by FSK cells expressed only mutant p53 genes. The mutation in one outgrowth cell line (TM-2H) resulted in the loss of p53 protein synthesis, whereas two other outgrowth lines (TM-3, TM-4) overexpressed mutant p53 protein. Mutation of p53 appears to correlate with preneoplastic growth in vivo. Although it is not clear if the mutations occur before or after transplantation of cells in vivo, there appears to be a pronounced growth advantage in the mammary gland for cells expressing mutant p53.


Assuntos
Sequência Conservada , Genes p53/genética , Neoplasias Mamárias Animais/genética , Mutação/genética , Lesões Pré-Cancerosas/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Sequência de Bases , Meia-Vida , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Dados de Sequência Molecular , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética
16.
Cancer Res ; 53(3): 668-74, 1993 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8425201

RESUMO

The TM preneoplastic mammary outgrowth lines were established in vivo from mammary epithelial cell lines and have been characterized with respect to their tumorigenic, morphological, and functional properties. The TM outgrowth lines were then established as in vitro cell lines. A comparison of the growth factor dependencies of the TM preneoplastic lines and their progenitor cell lines grown in monolayer cell culture indicated that the TM preneoplastic cell lines exhibited a decreased dependence on epidermal growth factor for growth in vitro. The exception to this result was the TM3 cell line which still exhibited a marked dependence on epidermal growth factor for growth. An examination of several genes for mRNA levels indicated that the expression of c-neu, c-H-ras, c-myc, and retinoblastoma was not elevated in those TM preneoplasias which exhibited a decreased dependence on epidermal growth factor. Additionally, there was no evidence that c-H-ras was activated in the preneoplastic outgrowths or tumors. In contrast, mouse mammary tumor virus long terminal repeat mRNA was increased in preneoplasias and tumors, whereas gelsolin mRNA was decreased in tumors but not in preneoplasias. The down-regulation of gelsolin mRNA is consistent with recent reports in human breast cancers. These results together with those reported in another paper (D. Medina et al., Cancer Res., 53: 663-667, 1993) indicate that the TM3 outgrowth line is a minimally deviated preneoplasia which does not share many of the molecular alterations exhibited in tumorigenic TM preneoplastic outgrowth lines. These data, along with other recent data, are interpreted in a hypothesis which views the three essential characteristics of the mammary preneoplastic state as independent and dissociable genetic alterations. Importantly, each of the three characteristics is independently isolated in one or more of the in vivo outgrowth populations. These outgrowth lines should allow identification of the nature and function of the molecular alterations associated with each stage of mammary preneoplasia.


Assuntos
Substâncias de Crescimento/farmacologia , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/fisiologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Proto-Oncogenes/genética , Animais , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Epitélio/patologia , Epitélio/fisiologia , Feminino , Genes myc/genética , Genes ras/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Células Tumorais Cultivadas
17.
Cancer Res ; 53(3): 663-7, 1993 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-7678778

RESUMO

The TM series of preneoplastic mammary outgrowth lines was derived from the transplantation of the FSK mammary cell lines into the cleared mammary fat pads of syngeneic female BALB/c mice. The tumor-producing capabilities of the 6 TM outgrowth lines varied from high (TM2, -4, -6) to low (TM9, -10) to nil (TM3). Outgrowth lines 2, 4, and 6 each segregated into sublines of high and low tumor potential. The majority of the outgrowth lines exhibited a moderate to dense alveolar hyperplasia typical of mouse mammary hyperplasias. The exceptions to this picture were lines TM2H and TM10 which exhibited a unique ductular morphology. The ductular morphology was not correlated with tumor potential of the outgrowth lines but was correlated with the expression of K6 and K14 keratins in luminal epithelial cells. In an examination of the growth and hormonal responsiveness properties of the TM outgrowth lines, the TM3 line stands as distinct from the other lines and from any other lines previously characterized in BALB/c mice. The TM3 line grew very slowly and failed to fill the fat pad by 12 months of age. At 12 months of age, the alveolar hyperplasia had regressed so that only bare ducts were present. The TM3 outgrowth was ovarian hormone dependent for growth and alveolar differentiation. TM3 outgrowth represents a minimally deviated mammary hyperplasia which has acquired the immortal phenotype but not the other phenotypic characteristics of mammary preneoplasias. This outgrowth line will be useful for examining the essential molecular changes important for the preneoplastic state, some of which are reported in an accompanying paper (D. Medina et al., Cancer Res., 53: 668-674, 1993).


Assuntos
Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/patologia , Lesões Pré-Cancerosas/patologia , Animais , Caseínas/biossíntese , Caseínas/genética , Feminino , Hormônios/farmacologia , Hormônios/fisiologia , Queratinas/fisiologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Ovário/fisiologia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/fisiopatologia , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas
18.
Carcinogenesis ; 14(1): 25-8, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8425268

RESUMO

Mouse mammary epithelial cells (MMEC) isolated from normal virgin BALB/c female mice and grown in cell culture for various lengths of time were injected into the mammary fat pads of syngenic mice. Of the ductal outgrowths which resulted from the injected MMEC, four gave rise to outgrowths that were serially transplanted beyond the lifetime of normal ductal outgrowths. The lifetime of normal ducts is five or six transplant generations. The four ductal outgrowth lines, termed EL for 'extended life', have been serially transplanted for 7, 9, 13 and 14 transplant generations. The outgrowths are predominantly ductal in morphology, do not exhibit intraductal epitheliosis characteristic of ductal hyperplasias, are ovarian dependent for growth and are responsive to prolactin-mediated alveolar differentiation. Three of the EL lines, EL5, 7 and 11 have not produced any tumors spontaneously (0/64) and only one tumor after dimethylbenz[a]anthracene (DMBA) treatment (1/30). The fourth line, EL12, differs from the other three in the presence of a limited degree of alveolar differentiation. The EL12 line has not produced any spontaneous tumors (0/23) but is somewhat more responsive to DMBA (3/10). We interpret the EL lines (at least EL11 and EL12) to represent cell populations where the immortalized phenotype is dissociated from the hyperplastic phenotype which is characteristic of mouse mammary preneoplastic populations. The tumor suppressor gene, p53, is not overexpressed in the EL ductal outgrowths. To our knowledge, this is the first example of cell populations in vivo that are immortalized but otherwise normal. As such, they may represent the earliest stage observable in the genesis of mouse mammary tumors and provide unique cell populations to examine molecular alterations associated with the property of immortality.


Assuntos
Divisão Celular , Transformação Celular Neoplásica , Neoplasias Mamárias Experimentais/patologia , Lesões Pré-Cancerosas/patologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Linhagem Celular , Epitélio/patologia , Epitélio/transplante , Feminino , Genes p53 , Neoplasias Mamárias Experimentais/etiologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Ovariectomia , Fenótipo
19.
Cancer Res ; 52(9): 2482-8, 1992 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-1568218

RESUMO

Methyl-p-hydroxyphenyllactate (MeHPLA) is a bioflavonoid and/or tyrosine metabolite which may regulate cellular growth and proliferation through interactions with nuclear type II sites. Our current studies suggest that type II sites may function as MeHPLA receptors which are localized on the nuclear matrix, and occupancy of this binding site by MeHPLA directly correlates with the inhibition of normal and malignant cell proliferation. This ligand is inactivated by MeHPLA esterase in mammary tumors, resulting in a deficiency in MeHPLA, high quantities of unoccupied type II sites, and uncontrolled cellular proliferation. For these reasons we synthesized 2,6-bis((3,4-dihydroxyphenyl)methylene)-cyclohexanone (BDHPC) and 2,6-bis((3-methoxy-4-hydroxyphenyl)-methylene)cyclohexanone (BMHPC) for assessment as nuclear type II site and cell growth antagonists. These two esterase stable cyclohexanone derivatives, which bind to nuclear type II sites with high affinity (Kd 1-7 nM), mimic MeHPLA as cell growth-regulating agents. Dose-dependent occupancy of type II sites in MCF-7 human cells by BDHPC and BMHPC directly correlated with the inhibition of cell proliferation, and administration of BDHPC by silastic implant inhibited mouse mammary tumor growth in vivo. These findings demonstrate that esterase-stable type II antagonists such as BDHPC and BMHPC inhibit mammary cancer cell proliferation in vitro and in vivo and support earlier studies demonstrating that MeHPLA and functionally related compounds may regulate malignant cell proliferation at the level of this binding site.


Assuntos
Neoplasias da Mama/metabolismo , Catecóis/metabolismo , Núcleo Celular/metabolismo , Cicloexanonas/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Neoplasias da Mama/patologia , Catecóis/farmacologia , Divisão Celular/efeitos dos fármacos , Cicloexanonas/farmacologia , Estradiol/metabolismo , Feminino , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Endogâmicos
20.
Cancer Res ; 52(7): 1924-32, 1992 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1372532

RESUMO

A series of mouse mammary epithelial cell lines has been established by a protocol that gives highly reproducible results. The mammary epithelial cell lines, designated as FSK lines, were judged to be epithelial based on positive immunostaining for keratin-intermediate filaments, negative immunostaining for vimentin-intermediate filaments, hormonal induction of casein, and the ability to exhibit ductal and alveolar mammary morphogenesis in vivo. The FSK cell lines are dependent on epidermal growth factor and insulin in a low serum (1%) medium. Conditioned medium from spindle cell cultures replaced the requirement for serum and increased the growth of FSK3 and FSK4 4-5 times in collagen gels and 12-14 times in monolayer culture, respectively. Following injection into the mammary fat pad at passages 2-11, the FSK cell lines generated stable transplantable hyperplastic alveolar outgrowth lines. The in vivo outgrowth lines were judged as preneoplastic based on their stable alveolar morphology in vivo and an increased susceptibility for tumorigenesis. The FSK cell lines and their derivative in vivo outgrowth lines provide a new and potentially productive system to examine critical molecular alterations involved in the development of mammary preneoplasias. Furthermore, the reproducibility of the in vitro culture system provides the assurance that stable cell lines of mouse mammary epithelial cells can be generated easily and at will.


Assuntos
Linhagem Celular , Glândulas Mamárias Animais/citologia , Neoplasias Mamárias Experimentais/patologia , Lesões Pré-Cancerosas/patologia , Adenocarcinoma Bronquioloalveolar/patologia , Animais , Carcinoma Intraductal não Infiltrante/patologia , Divisão Celular/efeitos dos fármacos , Técnicas de Cultura , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais , Feminino , Imunofluorescência , Queratinas/análise , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA