Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Cancer Invest ; 38(5): 300-309, 2020 May.
Article in English | MEDLINE | ID: mdl-32378982

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Centrosome/pathology , Hormones/metabolism , Aneuploidy , Animals , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Centrosome/metabolism , Female , Gene Amplification/physiology , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mitosis/physiology , Polyploidy
3.
Article in English | MEDLINE | ID: mdl-32349923

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Progesterone , Breast , Breast Neoplasms/drug therapy , Female , Hormones , Humans , Pregnancy , Progestins , Receptors, Progesterone
4.
Endocr Relat Cancer ; 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-29991638

ABSTRACT

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.

5.
Cancer Genet ; 207(6): 233-46, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25176624

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Chromosome Aberrations , Disease Models, Animal , Mammary Neoplasms, Experimental/genetics , Animals , Cytogenetics , Female , Humans , Mice
6.
Cancer Genet ; 205(4): 168-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22559978

ABSTRACT

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.


Subject(s)
Cell Line, Tumor , Chromosome Aberrations , Disease Models, Animal , Urinary Bladder Neoplasms/genetics , Animals , Chromosome Banding , Chromosomes, Mammalian/genetics , Cytogenetic Analysis , Disease Progression , In Situ Hybridization, Fluorescence , Karyotyping , Mice , Sequence Deletion , Translocation, Genetic , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
7.
BMC Cancer ; 10: 293, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20553594

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cell Communication , Cell Proliferation , Epithelial Cells/pathology , Fibroblasts/pathology , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/blood supply , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/metabolism , Cell Survival , Epithelial Cells/metabolism , Female , Fibroblasts/metabolism , Gonadal Steroid Hormones/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Time Factors , Tumor Burden , Tumor Cells, Cultured
8.
Endocr Relat Cancer ; 16(2): 333-50, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19190078

ABSTRACT

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.


Subject(s)
Carcinoma, Ductal/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasms, Hormone-Dependent/metabolism , Receptors, Progesterone/physiology , Animals , Antineoplastic Agents, Hormonal/toxicity , Female , Mammary Neoplasms, Experimental/pathology , Medroxyprogesterone Acetate/toxicity , Mice , Mice, Inbred BALB C , Receptors, Estrogen/metabolism
9.
Int J Cancer ; 123(11): 2518-31, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18767044

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fibroblast Growth Factor 2/pharmacology , Progesterone/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, Progesterone/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Female , Fibroblasts , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Protein Binding , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction/drug effects
10.
Cancer Res ; 67(9): 4104-12, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483321

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 13/genetics , Gene Amplification , Mammary Neoplasms, Experimental/genetics , Animals , Blotting, Northern , Chromosome Mapping , DNA, Neoplasm/genetics , Gene Expression , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Mice , Multigene Family , Nucleic Acid Hybridization , Reverse Transcriptase Polymerase Chain Reaction
11.
Cancer Genet Cytogenet ; 161(2): 130-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16102583

ABSTRACT

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.


Subject(s)
Aneuploidy , Genes, p53 , Mammary Neoplasms, Experimental/genetics , Neoplasms, Hormone-Dependent/genetics , Point Mutation , Animals , Chromosome Banding , Female , In Situ Hybridization, Fluorescence , Medroxyprogesterone Acetate , Mice , Mice, Inbred BALB C , Tryptophan
SELECTION OF CITATIONS
SEARCH DETAIL
...