Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Genome Biol ; 14(11): R125, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24220145

ABSTRACT

BACKGROUND: Human breast cancer is a heterogeneous disease consisting of multiple molecular subtypes. Genetically engineered mouse models are a useful resource for studying mammary cancers in vivo under genetically controlled and immune competent conditions. Identifying murine models with conserved human tumor features will facilitate etiology determinations, highlight the effects of mutations on pathway activation, and should improve preclinical drug testing. RESULTS: Transcriptomic profiles of 27 murine models of mammary carcinoma and normal mammary tissue were determined using gene expression microarrays. Hierarchical clustering analysis identified 17 distinct murine subtypes. Cross-species analyses using three independent human breast cancer datasets identified eight murine classes that resemble specific human breast cancer subtypes. Multiple models were associated with human basal-like tumors including TgC3(1)-Tag, TgWAP-Myc and Trp53-/-. Interestingly, the TgWAPCre-Etv6 model mimicked the HER2-enriched subtype, a group of human tumors without a murine counterpart in previous comparative studies. Gene signature analysis identified hundreds of commonly expressed pathway signatures between linked mouse and human subtypes, highlighting potentially common genetic drivers of tumorigenesis. CONCLUSIONS: This study of murine models of breast carcinoma encompasses the largest comprehensive genomic dataset to date to identify human-to-mouse disease subtype counterparts. Our approach illustrates the value of comparisons between species to identify murine models that faithfully mimic the human condition and indicates that multiple genetically engineered mouse models are needed to represent the diversity of human breast cancers. The reported trans-species associations should guide model selection during preclinical study design to ensure appropriate representatives of human disease subtypes are used.


Subject(s)
Breast Neoplasms/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Transcriptome/genetics , Animals , Breast Neoplasms/classification , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cluster Analysis , Female , Humans , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA
2.
EMBO Mol Med ; 5(9): 1335-50, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868506

ABSTRACT

We show that elevated levels of Ret receptor are found in different sub-types of human breast cancers and that high Ret correlates with decreased metastasis-free survival. The role of Ret in ER+ breast cancer models was explored combining in vitro and in vivo approaches. Our analyses revealed that ligand-induced Ret activation: (i) stimulates migration of breast cancer cells; (ii) rescues cells from anti-proliferative effects of endocrine treatment and (iii) stimulates expression of cytokines in the presence of endocrine agents. Indeed, we uncovered a positive feed-forward loop between the inflammatory cytokine IL6 and Ret that links them at the expression and the functional level. In vivo inhibition of Ret in a metastatic breast cancer model inhibits tumour outgrowth and metastatic potential. Ret inhibition blocks the feed-forward loop by down-regulating Ret levels, as well as decreasing activity of Fak, an integrator of IL6-Ret signalling. Our results suggest that Ret kinase should be considered as a novel therapeutic target in subsets of breast cancer.


Subject(s)
Breast Neoplasms/physiopathology , Cell Movement , Cell Proliferation , Neoplasm Metastasis/physiopathology , Proto-Oncogene Proteins c-ret/biosynthesis , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/mortality , Disease Models, Animal , Female , Humans , Interleukin-6/metabolism , Mice , Microarray Analysis , Survival Analysis
3.
Cancer Res ; 70(10): 4102-11, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20442283

ABSTRACT

We have previously reported the oncogenic properties of the gene amplified in breast cancer 1 (AIB1), a member of the p160 family of hormone receptor coactivators. In a transgenic mouse model, AIB1 overexpression resulted in a high incidence of tumors in various tissues, including mammary gland, uterus, lung, and pituitary. To determine whether the AIB1 oncogenicity in this model depended on its function as an estrogen receptor (ER) coactivator, we abolished ER signaling through two independent approaches, by performing ovariectomy on AIB1 transgenic (AIB1-tg) mice to prevent gonadal estrogen production and by crossing AIB1-tg mice with ERalpha-null mutant mice. Ovariectomized (ovx) mice, but not AIB1 x ERalpha-/- mice, still developed mammary gland hyperplasia and ductal carcinoma in situ. Both approaches, however, completely prevented the development of invasive mammary tumors, indicating that invasive mammary tumor formation is strictly estrogen dependent. Once developed, AIB1-induced mammary tumors can subsequently lose their dependence on estrogen: Injection of ERalpha(+) tumor cell lines derived from such tumors into ovx or untreated wild-type mice resulted in a similar rate of tumor growth in both groups. Surprisingly, however, ovx mice had an approximately 4-fold higher rate of metastasis formation, suggesting that estrogen provided some protection from metastasis formation. Lastly, our experiments identified oncogenic functions of AIB1 that are independent of its ER coactivation, as both approaches, ovariectomy and ER-/- crosses, still resulted in a high incidence of tumors in the lung and pituitary. We therefore conclude that AIB1 can exert its oncogenicity through tissue-specific estrogen-dependent and estrogen-independent functions.


Subject(s)
Carcinoma, Intraductal, Noninfiltrating/pathology , Estrogens/physiology , Mammary Neoplasms, Experimental/pathology , Neoplasms, Hormone-Dependent/pathology , Nuclear Receptor Coactivator 3/metabolism , Animals , Blotting, Western , Carcinoma, Intraductal, Noninfiltrating/metabolism , Estrogen Receptor alpha/metabolism , Female , Incidence , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms, Hormone-Dependent/metabolism , Nuclear Receptor Coactivator 3/antagonists & inhibitors , Ovariectomy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
4.
Cancer Res ; 68(9): 3204-13, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451146

ABSTRACT

CD151, a master regulator of laminin-binding integrins (alpha(6)beta(4), alpha(6)beta(1), and alpha(3)beta(1)), assembles these integrins into complexes called tetraspanin-enriched microdomains. CD151 protein expression is elevated in 31% of human breast cancers and is even more elevated in high-grade (40%) and estrogen receptor-negative (45%) subtypes. The latter includes triple-negative (estrogen receptor, progesterone receptor, and HER2 negative) basal-like tumors. CD151 ablation markedly reduced basal-like mammary cell migration, invasion, spreading, and signaling (through FAK, Rac1, and lck) while disrupting epidermal growth factor receptor (EGFR)-alpha(6) integrin collaboration. Underlying these defects, CD151 ablation redistributed alpha(6)beta(4) integrins subcellularly and severed molecular links between integrins and tetraspanin-enriched microdomains. In a prototypical basal-like mammary tumor line, CD151 ablation notably delayed tumor progression in ectopic and orthotopic xenograft models. These results (a) establish that CD151-alpha(6) integrin complexes play a functional role in basal-like mammary tumor progression; (b) emphasize that alpha(6) integrins function via CD151 linkage in the context of tetraspanin-enriched microdomains; and (c) point to potential relevance of CD151 as a high-priority therapeutic target, with relative selectivity (compared with laminin-binding integrins) for pathologic rather than normal physiology.


Subject(s)
Antigens, CD/physiology , Breast Neoplasms/genetics , Integrin alpha6/genetics , Integrin alpha6/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion/genetics , Cell Movement/genetics , Disease Progression , Epidermal Growth Factor/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha6/physiology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Protein Binding , Signal Transduction/genetics , Tetraspanin 24 , Tissue Distribution , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Cancer Res ; 66(23): 11381-8, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17145884

ABSTRACT

Amplified in breast cancer 1 (AIB1), an estrogen receptor (ER) coactivator, is frequently amplified or overexpressed in human breast cancer. We previously developed a transgenic mouse model in which AIB1 can act as an oncogene, giving rise to a premalignant hyperplastic mammary phenotype as well as to a high incidence of mammary tumors that are primarily ER(+). In this model, the AIB1 transgene is responsible for continued activation of the insulin-like growth factor-I receptor, suggesting a role for the activation of the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (mTOR) pathway in the premalignant phenotype and tumor development. Here we show that treatment of AIB1 transgenic mice with the mTOR inhibitor RAD001 reverts the premalignant phenotype. Furthermore, treatment of cell lines derived from AIB1-dependent mammary tumors with RAD001 in culture leads to a G(1) cell cycle arrest. Lastly, tumor growth after injection of ER(+) AIB1 tumor cell lines into wild-type animals is inhibited by RAD001 treatment. In this ER(+) model, inhibition of tumor growth by RAD001 was significantly better than inhibition by the antiestrogen 4-hydroxytamoxifen alone, whereas a combination of both RAD001 and 4-hydroxytamoxifen was most effective. Based on these results, we propose that the combination of mTOR inhibition and ER-targeted endocrine therapy may improve the outcome of the subset of ER(+) breast cancers overexpressing AIB1. These studies provide preclinical support for the clinical development of RAD001 and suggest that AIB1 may be a predictive factor of RAD001 response.


Subject(s)
Histone Acetyltransferases/genetics , Mammary Neoplasms, Experimental/genetics , Protein Kinases/physiology , Trans-Activators/genetics , Animals , Blotting, Western , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Dose-Response Relationship, Drug , Endometrial Hyperplasia/chemically induced , Endometrial Hyperplasia/prevention & control , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Everolimus , Female , G1 Phase/drug effects , G1 Phase/genetics , G1 Phase/physiology , Histone Acetyltransferases/physiology , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Transgenic , Nuclear Receptor Coactivator 3 , Oncogenes/genetics , Oncogenes/physiology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Precancerous Conditions/prevention & control , Receptors, Estrogen/agonists , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases , Tamoxifen/adverse effects , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Trans-Activators/physiology , Tumor Cells, Cultured
6.
Genes Dev ; 20(18): 2513-26, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16980581

ABSTRACT

Estrogen stimulates the proliferation of the most common type of human breast cancer that expresses estrogen receptor alpha (ERalpha) through the activation of the cyclin D1 (CCND1) oncogene. However, our knowledge of ERalpha transcriptional mechanisms remains limited. Hence, it is still elusive why ERalpha ectopically expressed in ER-negative breast cancer cells (BCC) is functional on ectopic reporter constructs but lacks activity on many endogenous target genes, including CCND1. Here, we show that estradiol (E2) stimulation of CCND1 expression in BCC depends on a novel cell-type-specific enhancer downstream from the CCND1 coding region, which is the primary ERalpha recruitment site in estrogen-responsive cells. The pioneer factor FoxA1 is specifically required for the active chromatin state of this enhancer and as such is crucial for both CCND1 expression and subsequent cell cycle progression. Interestingly, even in BCC, CCND1 levels and proliferation are tightly controlled by E2 through the establishment of a negative feedforward loop involving the induction of NFIC, a putative tumor suppressor capable of directly repressing CCND1 transcription. Taken together, our results reveal an estrogen-regulated combinatorial network including cell-specific cis- and trans-regulators of CCND1 expression where ERalpha collaborates with other transcription factors associated with the ER-positive breast cancer phenotype, including FoxA1 and NFIC.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cyclin D1/genetics , Estradiol/metabolism , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , DNA Polymerase II/metabolism , Enhancer Elements, Genetic , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , LIM Domain Proteins , Microfilament Proteins/genetics , Models, Biological , NFI Transcription Factors/genetics , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Transcription, Genetic/drug effects
7.
Cancer Cell ; 6(3): 263-74, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15380517

ABSTRACT

The gene encoding AIB1, an estrogen receptor coactivator, is amplified in a subset of human breast cancers. Here we show that overexpression of AIB1 in transgenic mice (AIB1-tg) leads to mammary hypertrophy, hyperplasia, abnormal postweaning involution, and the development of malignant mammary tumors. Tumors are also increased in other organs, including the pituitary and uterus. AIB1 overexpression increases mammary IGF-I mRNA and serum IGF-I protein levels. In addition, IGF-I receptor and downstream signaling molecules are activated in primary mammary epithelial cells and mammary tumor cells derived from AIB1-tg mice. Knockdown of AIB1 expression in cultured AIB1-tg mammary tumor cells leads to reduced IGF-I mRNA levels and increased apoptosis, suggesting that an autocrine IGF-I loop underlies the mechanism of AIB1-induced oncogenesis.


Subject(s)
Adenocarcinoma/genetics , Mammary Neoplasms, Experimental/genetics , Oncogenes , Trans-Activators/genetics , Acetyltransferases , Animals , Cell Differentiation , Female , Gene Silencing , Glycogen Synthase Kinase 3/metabolism , Histone Acetyltransferases , Insulin-Like Growth Factor I/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Mice, Transgenic , Nuclear Receptor Coactivator 3 , Oncogene Proteins , Pregnancy , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...