Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Proteome Res ; 2023 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-37497607

RESUMO

The lipid metabolism adaptations of estrogen and progesterone receptor-positive breast cancer tumors from a mouse syngeneic model are investigated in relation to differences across the transition from hormone-dependent (HD) to hormone-independent (HI) tumor growth and the acquisition of endocrine therapy (ET) resistance (HIR tumors). Results are articulated with reported polar metabolome results to complete a metabolic picture of the above transitions and suggest markers of tumor progression and aggressiveness. Untargeted nuclear magnetic resonance metabolomics was used to analyze tumor and mammary tissue lipid extracts. Tumor progression (HD-HI-HIR) was accompanied by increased nonesterified cholesterol forms and phospholipids (phosphatidylcholine, phosphatidylethanolamine, sphingomyelins, and plasmalogens) and decreased relative contents of triglycerides and fatty acids. Predominating fatty acids became shorter and more saturated on average. These results were consistent with gradually more activated cholesterol synthesis, ß-oxidation, and phospholipid biosynthesis to sustain tumor growth, as well as an increase in cholesterol (possibly oxysterol) forms. Particular compound levels and ratios were identified as potential endocrine tumor HD-HI-HIR progression markers, supporting new hypotheses to explain acquired ET resistance.

3.
Clin Cancer Res ; 29(5): 866-877, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36269797

RESUMO

PURPOSE: Preclinical data suggest that antiprogestins inhibit the growth of luminal breast carcinomas that express higher levels of progesterone receptor isoform A (PRA) than isoform B (PRB). Thus, we designed a presurgical window of opportunity trial to determine the therapeutic effects of mifepristone in patients with breast cancer, based on their high PRA/PRB isoform ratio (MIPRA; NCT02651844). PATIENTS AND METHODS: Twenty patients with luminal breast carcinomas with PRA/PRB > 1.5 (determined by Western blots), and PR ≥ 50%, naïve from previous treatment, were included for mifepristone treatment (200 mg/day orally; 14 days). Core needle biopsies and surgical samples were formalin fixed for IHC studies, while others were snap-frozen to perform RNA sequencing (RNA-seq), proteomics, and/or Western blot studies. Plasma mifepristone levels were determined using mass spectrometry. The primary endpoint was the comparison of Ki67 expression pretreatment and posttreatment. RESULTS: A 49.62% decrease in Ki67 staining was observed in all surgical specimens compared with baseline (P = 0.0003). Using the prespecified response parameter (30% relative reduction), we identified 14 of 20 responders. Mifepristone induced an increase in tumor-infiltrating lymphocytes; a decrease in hormone receptor and pSer118ER expression; and an increase in calregulin, p21, p15, and activated caspase 3 expression. RNA-seq and proteomic studies identified downregulated pathways related to cell proliferation and upregulated pathways related to immune bioprocesses and extracellular matrix remodeling. CONCLUSIONS: Our results support the use of mifepristone in patients with luminal breast cancer with high PRA/PRB ratios. The combined effects of mifepristone and estrogen receptor modulators warrant clinical evaluation to improve endocrine treatment responsiveness in these patients. See related commentary by Ronchi and Brisken, p. 833.


Assuntos
Neoplasias da Mama , Mifepristona , Humanos , Feminino , Mifepristona/farmacologia , Mifepristona/uso terapêutico , Receptores de Progesterona/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteômica , Antígeno Ki-67 , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
4.
Front Oncol ; 12: 786931, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35299741

RESUMO

Breast cancer (BC) is the most common type of cancer in women and, in most cases, it is hormone-dependent (HD), thus relying on ovarian hormone activation of intracellular receptors to stimulate tumor growth. Endocrine therapy (ET) aimed at preventing hormone receptor activation is the primary treatment strategy, however, about half of the patients, develop resistance in time. This involves the development of hormone independent tumors that initially are ET-responsive (HI), which may subsequently become resistant (HIR). The mechanisms that promote the conversion of HI to HIR tumors are varied and not completely understood. The aim of this work was to characterize the metabolic adaptations accompanying this conversion through the analysis of the polar metabolomes of tumor tissue and non-compromised mammary gland from mice implanted subcutaneously with HD, HI and HIR tumors from a medroxyprogesterone acetate (MPA)-induced BC mouse model. This was carried out by nuclear magnetic resonance (NMR) spectroscopy of tissue polar extracts and data mining through multivariate and univariate statistical analysis. Initial results unveiled marked changes between global tumor profiles and non-compromised mammary gland tissues, as expected. More importantly, specific metabolic signatures were found to accompany progression from HD, through HI and to HIR tumors, impacting on amino acids, nucleotides, membrane percursors and metabolites related to oxidative stress protection mechanisms. For each transition, sets of polar metabolites are advanced as potential markers of progression, including acquisition of resistance to ET. Putative biochemical interpretation of such signatures are proposed and discussed.

5.
Biomolecules ; 10(9)2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32867141

RESUMO

The metabolic characteristics of metastatic and non-metastatic breast carcinomas remain poorly studied. In this work, untargeted Nuclear Magnetic Resonance (NMR) metabolomics was used to compare two medroxyprogesterone acetate (MPA)-induced mammary carcinomas lines with different metastatic abilities. Different metabolic signatures distinguished the non-metastatic (59-2-HI) and the metastatic (C7-2-HI) lines, with glucose, amino acid metabolism, nucleotide metabolism and lipid metabolism as the major affected pathways. Non-metastatic tumours appeared to be characterised by: (a) reduced glycolysis and tricarboxylic acid cycle (TCA) activities, possibly resulting in slower NADH biosynthesis and reduced mitochondrial transport chain activity and ATP synthesis; (b) glutamate accumulation possibly related to reduced glutathione activity and reduced mTORC1 activity; and (c) a clear shift to lower phosphoscholine/glycerophosphocholine ratios and sphingomyelin levels. Within each tumour line, metabolic profiles also differed significantly between tumours (i.e., mice). Metastatic tumours exhibited marked inter-tumour changes in polar compounds, some suggesting different glycolytic capacities. Such tumours also showed larger intra-tumour variations in metabolites involved in nucleotide and cholesterol/fatty acid metabolism, in tandem with less changes in TCA and phospholipid metabolism, compared to non-metastatic tumours. This study shows the valuable contribution of untargeted NMR metabolomics to characterise tumour metabolism, thus opening enticing opportunities to find metabolic markers related to metastatic ability in endocrine breast cancer.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico , Feminino , Glucose/metabolismo , Glicólise , Humanos , Metabolismo dos Lipídeos , Espectroscopia de Ressonância Magnética , Neoplasias Mamárias Experimentais/induzido quimicamente , Acetato de Medroxiprogesterona , Metaboloma , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica/patologia , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia
6.
Cancer Invest ; 38(5): 300-309, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32378982

RESUMO

Centrosome amplification leads to aberrant mitosis, giving rise to aneuploidy and it has been associated with poor prognosis in human cancers. This study aimed to evaluate the relationship between polyploidy, centrosome abnormalities, and response to endocrine treatment in progestin-induced mouse mammary carcinomas. We found cells with three or more centrosomes in the polyploid tumors. The endocrine unresponsive tumors showed a higher average number of centrosomes per cell than the responsive tumors. The results suggest an association between polyploidy and centrosome amplification with the resistance to endocrine therapy in this luminal breast cancer model.


Assuntos
Neoplasias da Mama/patologia , Carcinoma/patologia , Centrossomo/patologia , Hormônios/metabolismo , Aneuploidia , Animais , Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Linhagem Celular Tumoral , Centrossomo/metabolismo , Feminino , Amplificação de Genes/fisiologia , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Mitose/fisiologia , Poliploidia
7.
Artigo em Inglês | MEDLINE | ID: mdl-32349923

RESUMO

Progesterone (Pg) is a pregnancy-related hormone that prepares the endometrium for the implantation of the fertilized zygote and suppresses myometrial contractility for the maintenance of pregnancy. At high concentrations, it acts as a natural immunosuppressant avoiding the rejection of a half allogeneic foetus. It is the precursor of many other related steroid hormones, but what is its role in the human breast? In this chapter, we will discuss some aspects related to Pg and its role in breast development and in the neoplastic disease. Understanding the mechanisms related to Pg-induced effects in the normal and neoplastic mammary gland will light the way to exploit this hormone signalling pathway therapeutically. We will introduce some aspects of the effects of progestins in normal breast development, breast cancer risk and in neoplastic growth, and we will describe ongoing clinical trials in breast cancer using progestins or antiprogestins.


Assuntos
Neoplasias da Mama , Progesterona , Mama , Neoplasias da Mama/tratamento farmacológico , Feminino , Hormônios , Humanos , Gravidez , Progestinas , Receptores de Progesterona
8.
Endocr Relat Cancer ; 2018 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-29991638

RESUMO

There is a consensus that progestins and thus their cognate receptor molecules, the progesterone receptors (PR), are essential in the development of the adult mammary gland and regulators of proliferation and lactation. However, a role for natural progestins in breast carcinogenesis remains poorly understood. A hint to that possible role came from studies in which the synthetic progestin medroxyprogesterone acetate was associated with an increased breast cancer risk in women under hormone replacement therapy. However, progestins have been also used for breast cancer treatment and to inhibit the growth of several experimental breast cancer models. More recently, PR have been shown to be regulators of estrogen receptor signaling. With all this information, the question is how can we target PR, and if so, which patients may benefit from such an approach? PR are not single unique molecules. Two main PR isoforms have been characterized, PRA and PRB, that exert different functions and the relative abundance of one isoform respect to the other determines the response of PR agonists and antagonists. Immunohistochemistry with standard antibodies against PR do not discriminate between isoforms. In this review, we summarize the current knowledge on the expression of both PR isoforms in mammary glands, in experimental models of breast cancer and in breast cancer patients, to better understand how the PRA/PRB ratio can be exploited therapeutically to design personalized therapeutic strategies.

9.
J Pathol Clin Res ; 3(4): 227-233, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29085663

RESUMO

Progesterone receptors (PR) are prognostic and predictive biomarkers in hormone-dependent cancers. Two main PR isoforms have been described, PRB and PRA, that differ only in that PRB has 164 extra N-terminal amino acids. It has been reported that several antibodies empirically exclusively recognize PRA in formalin-fixed paraffin-embedded (FFPE) tissues. To confirm these findings, we used human breast cancer xenograft models, T47D-YA and -YB cells expressing PRA or PRB, respectively, MDA-MB-231 cells modified to synthesize PRB, and MDA-MB-231/iPRAB cells which can bi-inducibly express either PRA or PRB. Cells were injected into immunocompromised mice to generate tumours exclusively expressing PRA or PRB. PR isoform expression was verified using immunoblots. FFPE samples from the same tumours were studied by immunohistochemistry using H-190, clone 636, clone 16, and Ab-6 anti-PR antibodies, the latter exclusively recognizing PRB. Except for Ab-6, all antibodies displayed a similar staining pattern. Our results indicate that clones 16, 636, and the H-190 antibody recognize both PR isoforms. They point to the need for more stringency in evaluating the true specificity of purported PRA-specific antibodies as the PRA/PRB ratio may have prognostic and predictive value in breast cancer.

10.
Cancer Genet ; 207(6): 233-46, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25176624

RESUMO

Cytogenetic studies of breast cancer cells have identified numerous chromosomal imbalances, including gains in human chromosome regions 1q, 4p, 8q, and 20q and losses in regions 1p, 3p, 6q, 11q, 16q, 17p, and 22q. Mouse models have been developed to study the mechanisms of mammary carcinogenesis, and in most cases, the corresponding karyotypes have been reported. Here, I summarize the cytogenetic findings and the candidate genes that are involved in mammary tumorigenesis. The most commonly altered chromosomes in mouse breast cancer models are chromosomes 4 and 11, which are orthologous to human chromosomes that are also affected by chromosomal abnormalities in human breast cancer. The genes that are affected by chromosomal imbalances in mouse models have also been found to participate in human breast cancer. In addition, the amplification and overexpression of several new genes in mouse models have subsequently been confirmed in human breast cancer. In this review, I compile information on the available karyotypes for mouse breast cancer models.


Assuntos
Neoplasias da Mama/genética , Aberrações Cromossômicas , Modelos Animais de Doenças , Neoplasias Mamárias Experimentais/genética , Animais , Citogenética , Feminino , Humanos , Camundongos
11.
Cancer Genet ; 205(4): 168-76, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22559978

RESUMO

Bladder cancer is frequently associated with chromosomal abnormalities, and the complexity of karyotypes increases with tumor progression. The murine model MB49 is one of the most widely studied models of bladder cancer. We developed the invasive cell line MB49-I by successive in vivo passages of MB49 primary tumors. Because little is known about the chromosomal alterations of this model, our goal was to perform cytogenetic analyses of the MB49 and MB49-I lines. The karyotypes of both lines were analyzed by G-banding and fluorescence in situ hybridization techniques. Both lines were composed of two cell subpopulations, a diploid population, which was found mainly in the MB49 line, and the tetraploid population, which was found mainly in the MB49-I line. A translocation between chromosomes 5 and 9 and an isochromosome of chromosome 19 were observed in the subpopulations of both lines. New structural abnormalities and additional chromosomal imbalances were detected in the MB49-I line. Tumor progression in the MB49/MB49-I model was associated with a selection of polyploid cells with accompanying chromosomal abnormalities. This model may be advantageous for the study of the genetic changes associated with the progression of bladder cancer.


Assuntos
Linhagem Celular Tumoral , Aberrações Cromossômicas , Modelos Animais de Doenças , Neoplasias da Bexiga Urinária/genética , Animais , Bandeamento Cromossômico , Cromossomos de Mamíferos/genética , Análise Citogenética , Progressão da Doença , Hibridização in Situ Fluorescente , Cariotipagem , Camundongos , Deleção de Sequência , Translocação Genética , Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/patologia
12.
BMC Cancer ; 10: 293, 2010 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-20553594

RESUMO

BACKGROUND: Increasing evidence has underscored the role of carcinoma associated fibroblasts (CAF) in tumor growth. However, there are controversial data regarding the persistence of inoculated CAF within the tumors. We have developed a model in which murine metastatic ductal mammary carcinomas expressing estrogen and progesterone receptors transit through different stages of hormone dependency. Hormone dependent (HD) tumors grow only in the presence of progestins, whereas hormone independent (HI) variants grow without hormone supply. We demonstrated previously that CAF from HI tumors (CAF-HI) express high levels of FGF-2 and that FGF-2 induced HD tumor growth in vivo. Our main goal was to investigate whether inoculated CAF-HI combined with purified epithelial (EPI) HD cells can induce HD tumor growth. METHODS: Purified EPI cells of HD and HI tumors were inoculated alone, or together with CAF-HI, into female BALB/c mice and tumor growth was evaluated. In another set of experiments, purified EPI-HI alone or combined with CAF-HI or CAF-HI-GFP were inoculated into BALB/c or BALB/c-GFP mice. We assessed whether inoculated CAF-HI persisted within the tumors by analyzing inoculated or host CAF in frozen sections of tumors growing in BALB/c or BALB/c-GFP mice. The same model was used to evaluate early stages of tumor development and animals were euthanized at 2, 7, 12 and 17 days after EPI-HI or EPI-HI+CAF-HI inoculation. In angiogenesis studies, tumor vessels were quantified 5 days after intradermal inoculation. RESULTS: We found that admixed CAF-HI failed to induce epithelial HD tumor growth, but instead, enhanced HI tumor growth (p < 0.001). Moreover, inoculated CAF-HI did not persist within the tumors. Immunofluorescence studies showed that inoculated CAF-HI disappeared after 13 days. We studied the mechanisms by which CAF-HI increased HI tumor growth, and found a significant increase in angiogenesis (p < 0.05) in the co-injected mice at early time points. CONCLUSIONS: Inoculated CAF-HI do not persist within the tumor mass although they play a role during the first stages of tumor formation promoting angiogenesis. This angiogenic environment is unable to replace the hormone requirement of HD tumors that still need the hormone to recruit the stroma from the host.


Assuntos
Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Comunicação Celular , Proliferação de Células , Células Epiteliais/patologia , Fibroblastos/patologia , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/irrigação sanguínea , Carcinoma Ductal de Mama/imunologia , Carcinoma Ductal de Mama/metabolismo , Sobrevivência Celular , Células Epiteliais/metabolismo , Feminino , Fibroblastos/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Transplante de Neoplasias , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fatores de Tempo , Carga Tumoral , Células Tumorais Cultivadas
13.
Endocr Relat Cancer ; 16(2): 333-50, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19190078

RESUMO

More than 60% of all breast neoplasias are ductal carcinomas expressing estrogen (ER) and progesterone receptors (PR). By contrast, most of the spontaneous, chemically or mouse mammary tumor virus induced tumors, as well as tumors arising in genetically modified mice do not express hormone receptors. We developed a model of breast cancer in which the administration of medroxyprogesterone acetate to BALB/c female mice induces mammary ductal carcinomas with a mean latency of 52 weeks and an incidence of about 80%. These tumors are hormone-dependent (HD), metastatic, express both ER and PR, and are maintained by syngeneic transplants. The model has been further refined to include mammary carcinomas that evolve through different stages of hormone dependence, as well as several hormone-responsive cell lines. In this review, we describe the main features of this tumor model, highlighting the role of PR as a trigger of key signaling pathways mediating tumor growth. In addition, we discuss the relevance of this model in comparison with other presently used breast cancer models pointing out its advantages and limitations and how, this model may be suitable to unravel key questions in breast cancer.


Assuntos
Carcinoma Ductal/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Receptores de Progesterona/fisiologia , Animais , Antineoplásicos Hormonais/toxicidade , Feminino , Neoplasias Mamárias Experimentais/patologia , Acetato de Medroxiprogesterona/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Estrogênio/metabolismo
14.
Breast Cancer Res Treat ; 116(2): 247-55, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18622696

RESUMO

Currently, to our knowledge, there are no continuous cell lines derived from estrogen dependent, tamoxifen sensitive spontaneous mouse mammary carcinomas. We describe here the establishment and characterization of a cell line derived from the M05 mouse mammary tumor, LM05-Mix, composed of both an epithelial and a fibroblastic component. From it the respective epithelial LM05-E and fibroblastic LM05-F cell lines were generated by limiting dilution. Immunofluorescence studies confirmed that the epithelial cells were positive for E-cadherin, cytokeratins and vimentin whereas the fibroblastic cells were negative for the epithelial markers and positive for alpha-smooth muscle actin and vimentin. Both cell types expressed estrogen and progesterone receptors, although only the epithelial LM05-E cells were stimulated by estradiol and inhibited by tamoxifen. In the bicellular LM05-Mix cell line estradiol proved to stimulate cell proliferation whereas the response to tamoxifen was dependent on confluency and the degree of epithelial-fibroblastic interactions. The presence of membrane estrogen receptors in both cell types was suggested by the achievement of non-genomic responses to short treatments with estradiol, leading to the phosphorylation of ERK1/2. Finally, cytogenetic studies suggest that these two cell types represent independent cell populations within the tumor and would not be the result of an epithelial-mesenchymal transition. This model presents itself as a valuable alternative for the study of estrogen responsiveness and tamoxifen resistance in the context of epithelial-stromal interactions.


Assuntos
Linhagem Celular Tumoral/citologia , Células Epiteliais/citologia , Fibroblastos/citologia , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Animais , Apoptose/fisiologia , Western Blotting , Linhagem Celular Tumoral/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Células Epiteliais/metabolismo , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Fibroblastos/metabolismo , Imunofluorescência , Hibridização in Situ Fluorescente , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Neoplasias Mamárias Experimentais/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Translocação Genética
15.
Int J Cancer ; 123(11): 2518-31, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18767044

RESUMO

The mechanisms by which mammary carcinomas acquire hormone independence are still unknown. To study the role of cancer-associated fibroblasts (CAF) in the acquisition of hormone-independence we used a hormone-dependent (HD) mouse mammary tumor and its hormone-independent (HI) variant, which grows in vivo without hormone supply. HI tumors express higher levels of FGFR-2 than HD tumors. In spite of their in vivo differences, both tumors have the same hormone requirement in primary cultures. We demonstrated that CAF from HI tumors (CAF-HI) growing in vitro, express higher levels of FGF-2 than HD counterparts (CAF-HD). FGF-2 activated the progesterone receptors (PR) in the tumor cells, thus increasing cell proliferation in both HI and HD tumors. CAF-HI induced a higher proliferative rate on the tumor cells and in PR activation than CAF-HD. The blockage of FGF-2 in the co-cultures or the genetic or pharmacological inhibition of FGFR-2 inhibited PR activation and tumor cell proliferation. Moreover, in vivo, the FGFR inhibitor decreased C4-HI tumor growth, whereas FGF-2 was able to stimulate C4-HD tumor growth as MPA. T47D human breast cancer cells were also stimulated by progestins, FGF-2 or CAF-HI, and this stimulation was abrogated by antiprogestins, suggesting that the murine C4-HI cells respond as the human T47D cells. In summary, this is the first study reporting differences between CAF from HD and HI tumors suggesting that CAF-HI actively participate in driving HI tumor growth.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Progesterona/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Progesterona/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Feminino , Fibroblastos , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais/efeitos dos fármacos
16.
Cancer Res ; 67(9): 4104-12, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17483321

RESUMO

Serial analysis of gene expression from aggressive mammary tumors derived from transplantable p53 null mouse mammary outgrowth lines revealed significant up-regulation of Tfdp1 (transcription factor Dp1), Lamp1 (lysosomal membrane glycoprotein 1) and Gas6 (growth arrest specific 6) transcripts. All of these genes belong to the same linkage cluster, mapping to mouse chromosome band 8A1. BAC-array comparative genomic hybridization and fluorescence in situ hybridization analyses revealed genomic amplification at mouse region ch8A1.1. The minimal region of amplification contained genes Cul4a, Lamp1, Tfdp1, and Gas6, highly overexpressed in the p53 null mammary outgrowth lines at preneoplastic stages, and in all its derived tumors. The same amplification was also observed in spontaneous p53 null mammary tumors. Interestingly, this region is homologous to human chromosome 13q34, and some of the same genes were previously observed amplified in human carcinomas. Thus, we further investigated the occurrence and frequency of gene amplification affecting genes mapping to ch13q34 in human breast cancer. TFDP1 showed the highest frequency of amplification affecting 31% of 74 breast carcinomas analyzed. Statistically significant positive correlation was observed for the amplification of CUL4A, LAMP1, TFDP1, and GAS6 genes (P < 0.001). Meta-analysis of publicly available gene expression data sets showed a strong association between the high expression of TFDP1 and decreased overall survival (P = 0.00004), relapse-free survival (P = 0.0119), and metastasis-free interval (P = 0.0064). In conclusion, our findings suggest that CUL4A, LAMP1, TFDP1, and GAS6 are targets for overexpression and amplification in breast cancers. Therefore, overexpression of these genes and, in particular, TFDP1 might be of relevance in the development and/or progression in a significant subset of human breast carcinomas.


Assuntos
Neoplasias da Mama/genética , Cromossomos Humanos Par 13/genética , Amplificação de Genes , Neoplasias Mamárias Experimentais/genética , Animais , Northern Blotting , Mapeamento Cromossômico , DNA de Neoplasias/genética , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Hibridização in Situ Fluorescente , Camundongos , Família Multigênica , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Cancer Genet Cytogenet ; 161(2): 130-9, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16102583

RESUMO

Medroxyprogesterone acetate (MPA)-induced mammary carcinomas express high levels of estrogen (ER) and progesterone receptors (PR) and when transplanted in syngeneic mice they show a progestin-dependent (PD) growth pattern. By successive transplantation, progestin-independent (PI) variants were generated and showed a different response to antihormone therapy. A diploid chromosome number (2n=40) was found in three of five PD tumors, with numbers in the triploid to tetraploid range in the other two. Some PI tumors were diploid, but most were aneuploid (8 of 12 tumors). The most frequent alterations found in PD and PI tumors were gains of chromosomes 3, 4, and 6 and losses of chromosomes 16 and X. Chromosomes 4 and 7 were involved in translocations in three of the four tumor families studied. single-strand conformation polymorphism analysis revealed a point mutation on the Trp53 gene in one of the PD tumors; this showed a stable diploid karyotype, suggesting that mutated Trp53 is not uniquely involved in chromosome instability. We have shown that hormone independence may be acquired without changes in ploidy, suggesting that the increase in ploidy is favored by successive transplantation. In our model, diploid tumors responded to hormone treatment but aneuploid tumors were either responsive or not.


Assuntos
Aneuploidia , Genes p53 , Neoplasias Mamárias Experimentais/genética , Neoplasias Hormônio-Dependentes/genética , Mutação Puntual , Animais , Bandeamento Cromossômico , Feminino , Hibridização in Situ Fluorescente , Acetato de Medroxiprogesterona , Camundongos , Camundongos Endogâmicos BALB C , Triptofano
18.
J Cell Physiol ; 202(3): 672-82, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15389583

RESUMO

We have developed a murine mammary tumor cell line, MC4-L4, and after 15 passages, a spindle-shaped population became evident. The cuboidal cells, MC4-L4E, cloned by limit dilution, proved to be epithelial tumor cells. When inoculated in syngeneic mice, they gave rise to invasive metastatic carcinomas expressing estrogen and progesterone receptors. These tumors regressed after anti-progestin treatment and stopped growing after 17-beta-estradiol administration. In vitro, they were insensitive to medroxyprogesterone acetate (MPA), 17-beta-estradiol, and EGF and were inhibited by TGFbeta1. They expressed mutated p53 and estrogen receptors alpha; progesterone receptors were undetectable. Cells were polyploid and shared the same four common marker chromosomes present in the parental tumor in addition to an exclusive marker. Spindle-shaped cells, MC4-L4F, were selected by differential attachment and detachment and proved to be non-epithelial non-tumorigenic cells. They were cytokeratin negative, showed mesenchymal features by electron microscopy, differentiated to adipocytes when treated with an adipogenic cocktail, were stimulated by TGFbeta1 and EGF, showed a wild-type p53, and did not exhibit the marker chromosomes of the parental tumor. Although they expressed estrogen receptors alpha, they were insensitive to 17-beta-estradiol in proliferation assays. Co-cultures of both cell types had a synergic effect on progesterone receptors expression and on cell proliferation, being the epithelial cells, the most responsive ones, and 17-beta-estradiol increased cell proliferation only in co-cultures. Cytogenetic studies and data on p53 mutations rule out the possibility of an epithelial mesenchymal transition. Their unique characteristics make them an excellent model to be used in studies of epithelial-stromal interactions in the context of hormone responsiveness in hormone related tumors.


Assuntos
Neoplasias da Mama , Linhagem Celular Tumoral , Glândulas Mamárias Animais/citologia , Células Estromais , Animais , Antineoplásicos Hormonais/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Linhagem Celular Tumoral/citologia , Linhagem Celular Tumoral/fisiologia , Forma Celular , Técnicas de Cocultura , Fator de Crescimento Epidérmico/metabolismo , Estradiol/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Marcadores Genéticos , Antagonistas de Hormônios/metabolismo , Humanos , Queratinas/metabolismo , Acetato de Medroxiprogesterona/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mifepristona/metabolismo , Mutação , Transplante de Neoplasias , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1 , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
19.
Breast Cancer Res Treat ; 83(3): 233-44, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14758093

RESUMO

We report the establishment of two mouse mammary cancer cell lines, MC7-2A and MC7-2B obtained from a mouse mammary carcinoma induced by medroxyprogesterone acetate (MPA) and maintained by syngeneic transplantation in BALB/c mice. They are epithelial (express cytokeratins) and express both estrogen receptors alpha (ERalpha) and progesterone receptors (PRs) isoforms A and B (western blots). In vitro, MPA inhibited 3H-thymidine uptake, starting from concentrations as low as 10(-13) M in MC7-2A and 10(10) M in MC7-2B; the antiprogestin RU 486 exerted a stimulatory effect at 10(-14) M in both cell lines; 17-beta-estradiol (E2) also exerted a stimulatory effect starting at 10(-10) M in MC7-2A and at 10(-13) M in MC7-2B. When transplanted in syngeneic mice, both cell lines originated adenocarcinomas that gave rise to lung metastases within 3 months. In in vivo studies, in MC7-2A, the antiprogestin inhibited completely tumor growth, E2 induced a slight although significant ( p < 0.05) stimulatory effect and MPA stimulated tumor growth while MC7-2B cells were unresponsive to all treatments. ER and PR were also expressed in tumors as assessed by immunohistochemistry. Two marker chromosomes were identified by FISH as translocations between chromosomes 4 and 7, and between chromosomes X and 2; the third marker chromosome remains unidentified. All these markers were also present in the parental tumor. A new marker, a centric fusion of chromosomes 2, was acquired in both cell lines. Considering that there are very few murine breast carcinoma responsive cell lines, these cells represent new tools in which the regulatory effect of hormones can be studied.


Assuntos
Adenocarcinoma/patologia , Marcadores Genéticos , Neoplasias Mamárias Animais/patologia , Metástase Neoplásica/fisiopatologia , Receptores de Estrogênio/biossíntese , Receptores de Progesterona/biossíntese , Células Tumorais Cultivadas , Animais , Antineoplásicos Hormonais/farmacologia , Western Blotting , Estradiol/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/veterinária , Acetato de Medroxiprogesterona/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Estrogênio/análise , Receptores de Progesterona/análise
20.
Breast Cancer Res Treat ; 79(1): 25-35, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12779079

RESUMO

The purpose of this study was to evaluate the effect of the selective estrogen receptor modulators raloxifene and tamoxifen and of the pure antiestrogen fulvestrant on tumor growth and progesterone receptor (PR) expression in an experimental model of breast cancer. The effects of these compounds on cell proliferation were studied in primary cultures of a progestin-dependent mammary carcinoma tumor line, in the presence of medroxyprogesterone acetate (MPA) or 17-beta-estradiol (E2). In in vivo studies the tumor was inoculated subcutaneously in BALB/c female mice treated with 20 mg MPA depot. Raloxifene (12.5 mg/kg) or tamoxifen (5 mg/kg) were administered in daily doses or E2 silastic pellets (5 mg) were implanted. When the tumors reached about 25-50 mm2 MPA was removed in half of the animals. E2 induced complete tumor regressions, tamoxifen inhibited tumor growth in vivo while raloxifene disclosed proliferative effects in animals in which MPA had been removed. In vitro, E2 inhibited cell proliferation at concentrations higher than 10(-14)M. Raloxifene and fulvestrant, but not tamoxifen, partially reverted E2-induced inhibition. Fulvestrant and tamoxifen inhibited MPA-induced cell proliferation while raloxifene had a stimulatory effect. Tamoxifen and E2 increased, raloxifene induced no effect, and fulvestrant significantly decreased PR expression. In this study we provide evidence for differential effects of tamoxifen and raloxifene on experimental mammary tumors. Since raloxifene is under evaluation for use in breast cancer prevention, these results may have important clinical implications.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Carcinoma Ductal de Mama/tratamento farmacológico , Gonadotropina Coriônica Humana Subunidade beta/farmacologia , Estradiol/farmacologia , Moduladores de Receptor Estrogênico/farmacologia , Meliteno/farmacologia , Cloridrato de Raloxifeno/farmacologia , Tamoxifeno/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Divisão Celular/efeitos dos fármacos , Estradiol/análogos & derivados , Feminino , Acetato de Medroxiprogesterona/farmacologia , Meliteno/análogos & derivados , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Progesterona/efeitos dos fármacos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA