Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(7)2022 Mar 26.
Article in English | MEDLINE | ID: mdl-35409002

ABSTRACT

Oral Squamous Cell Carcinoma (OSCC) is the most common malignant cancer affecting the oral cavity. It is characterized by high morbidity and very few therapeutic options. Angiotensin (Ang)-(1-7) is a biologically active heptapeptide, generated predominantly from AngII (Ang-(1-8)) by the enzymatic activity of angiotensin-converting enzyme 2 (ACE 2). Previous studies have shown that Ang-(1-7) counterbalances AngII pro-tumorigenic actions in different pathophysiological settings, exhibiting antiproliferative and anti-angiogenic properties in cancer cells. However, the prevailing effects of Ang-(1-7) in the oral epithelium have not been established in vivo. Here, we used an inducible oral-specific mouse model, where the expression of a tamoxifen-inducible Cre recombinase (CreERtam), which is under the control of the cytokeratin 14 promoter (K14-CreERtam), induces the expression of the K-ras oncogenic variant KrasG12D (LSLK-rasG12D). These mice develop highly proliferative squamous papilloma in the oral cavity and hyperplasia exclusively in oral mucosa within one month after tamoxifen treatment. Ang-(1-7) treated mice showed a reduced papilloma development accompanied by a significant reduction in cell proliferation and a decrease in pS6 positivity, the most downstream target of the PI3K/Akt/mTOR signaling route in oral papilloma. These results suggest that Ang-(1-7) may be a novel therapeutic target for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Papilloma , Papillomavirus Infections , Angiotensin I/pharmacology , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Mice , Mice, Transgenic , Mouth Neoplasms/drug therapy , Papilloma/drug therapy , Papilloma/pathology , Papilloma/prevention & control , Papillomavirus Infections/drug therapy , Peptide Fragments , Phosphatidylinositol 3-Kinases/metabolism , Tamoxifen/therapeutic use
2.
Dis Model Mech ; 15(3)2022 03 01.
Article in English | MEDLINE | ID: mdl-35044452

ABSTRACT

RET is a receptor tyrosine kinase with oncogenic potential in the mammary epithelium. Several receptors with oncogenic activity in the breast are known to participate in specific developmental stages. We found that RET is differentially expressed during mouse mammary gland development: RET is present in lactation and its expression dramatically decreases in involution, the period during which the lactating gland returns to a quiescent state after weaning. Based on epidemiological and pre-clinical findings, involution has been described as tumor promoting. Using the Ret/MTB doxycycline-inducible mouse transgenic system, we show that sustained expression of RET in the mammary epithelium during the post-lactation transition to involution is accompanied by alterations in tissue remodeling and an enhancement of cancer potential. Following constitutive Ret expression, we observed a significant increase in neoplastic lesions in the post-involuting versus the virgin mammary gland. Furthermore, we show that abnormal RET overexpression during lactation promotes factors that prime involution, including premature activation of Stat3 signaling and, using RNA sequencing, an acute-phase inflammatory signature. Our results demonstrate that RET overexpression negatively affects the normal post-lactation transition.


Subject(s)
Mammary Glands, Human , Neoplasms , Animals , Female , Humans , Lactation/physiology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Human/metabolism , Mice , Neoplasms/pathology , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , STAT3 Transcription Factor/metabolism
3.
J Mammary Gland Biol Neoplasia ; 25(1): 13-26, 2020 03.
Article in English | MEDLINE | ID: mdl-32080788

ABSTRACT

Ret receptor tyrosine kinase is a proto-oncogene that participates in development of various cancers. Several independent studies have recently identified Ret as a key player in breast cancer. Although Ret overexpression and function have been under investigation, mainly in estrogen receptor positive breast cancer, a more comprehensive analysis of the impact of recurring Ret alterations in breast cancer is needed. This review consolidates the current knowledge of Ret alterations and their potential effects in breast cancer. We discuss and integrate data on Ret changes in different breast cancer subtypes and potential function in progression, as well as the participation of distinct Ret network signaling partners in these processes. We propose that it will be essential to define a shared molecular feature of tumors with alteration in Ret receptor, be this at the genetic level or via overexpression in order to design effective therapies to target the Ret pathway. Here we review experimental evidence from basic research and pre-clinical studies concentrating on Ret alterations as potential biomarkers for recurrence, and we discuss the possibility that targeting the Ret pathway might in the future become a treatment for breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Mutation , Proto-Oncogene Proteins c-ret/metabolism , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/genetics
4.
Oncogene ; 37(29): 4046-4054, 2018 07.
Article in English | MEDLINE | ID: mdl-29695833

ABSTRACT

The receptor tyrosine kinase Ret, a key gain-of-function mutated oncoprotein in thyroid carcinomas, has recently been implicated in other cancer types. While Ret copy number gains and mutations have been reported at low frequencies in breast tumors, we and others have reported that Ret is overexpressed in about 40% of human tumors and this correlates with poor patient prognosis. Ret activation regulates numerous intracellular pathways related to proliferation and inflammation, but it is not known whether abnormal Ret expression is sufficient to induce mammary carcinomas. Using a novel doxycycline-inducible transgenic mouse model with the MMTV promoter controlling Ret expression, we show that overexpression of wild-type Ret in the mammary epithelium produces mammary tumors, displaying a morphology that recapitulates characteristics of human luminal breast tumors. Ret-evoked tumors are estrogen receptor positive and negative for progesterone receptor. Moreover, tumors rapidly regress after doxycycline withdrawal, indicating that Ret is the driving oncoprotein. Using next-generation sequencing, we examined the levels of transcripts in these tumors, confirming a luminal signature. Ret-evoked tumors have been passaged in mice and used to test novel therapeutic approaches. Importantly, we have determined that tumors are resistant to endocrine therapy, but respond successfully to treatment with a Ret kinase inhibitor. Our data provide the first compelling evidence for an oncogenic role of non-mutated Ret in the mammary gland and are an incentive for clinical development of Ret as a cancer biomarker and therapeutic target.


Subject(s)
Breast Neoplasms/metabolism , Mammary Neoplasms, Animal/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Animals , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mammary Glands, Human/metabolism , Mice , Mice, Transgenic/metabolism , Receptors, Progesterone/metabolism
5.
Oncotarget ; 9(9): 8278-8289, 2018 Feb 02.
Article in English | MEDLINE | ID: mdl-29492194

ABSTRACT

Tristetraprolin (TTP), an mRNA-binding protein that negatively controls levels of inflammatory factors, is highly expressed in the lactating mouse mammary gland. To determine the biological relevance of this expression profile, we developed bi-transgenic mice in which this protein is specifically down-regulated in the secretory mammary epithelium in the secretory mammary epithelium during lactation. Our data show that TTP conditional KO mice produced underweight litters, possibly due to massive mammary cell death induced during lactation without the requirement of additional stimuli. This effect was linked to overexpression of inflammatory cytokines, activation of STAT3 and down-regulation of AKT phosphorylation. Importantly, blocking TNFα activity in the lactating conditional TTP KO mice inhibited cell death and similar effects were observed when this treatment was applied to wild-type animals during 48 h after weaning. Therefore, our results demonstrate that during lactation TTP wards off early involution by preventing the increase of local inflammatory factors. In addition, our data reveal the relevance of locally secreted TNFα for triggering programmed cell death after weaning.

6.
Nat Commun ; 7: 12258, 2016 07 13.
Article in English | MEDLINE | ID: mdl-27406745

ABSTRACT

The JAK/STAT pathway is an attractive target for breast cancer therapy due to its frequent activation, and clinical trials evaluating JAK inhibitors (JAKi) in advanced breast cancer are ongoing. Using patient biopsies and preclinical models of breast cancer, we demonstrate that the JAK/STAT pathway is active in metastasis. Unexpectedly, blocking the pathway with JAKi enhances the metastatic burden in experimental and orthotopic models of breast cancer metastasis. We demonstrate that this prometastatic effect is due to the immunosuppressive activity of JAKi with ensuing impairment of NK-cell-mediated anti-tumour immunity. Furthermore, we show that immunostimulation with IL-15 overcomes the enhancing effect of JAKi on metastasis formation. Our findings highlight the importance of evaluating the effect of targeted therapy on the tumour environment. The impact of JAKi on NK cells and the potential value of immunostimulators to overcome the weakened tumour immunosurveillance, are worthwhile considering in the clinical setting of breast cancer.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Immunologic Surveillance , Janus Kinase Inhibitors/pharmacology , Killer Cells, Natural/immunology , Models, Biological , Animals , Breast Neoplasms/drug therapy , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Enzyme Activation/drug effects , Female , Humans , Immunologic Surveillance/drug effects , Immunomodulation/drug effects , Interleukin-15/pharmacology , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/metabolism , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Mice , STAT Transcription Factors/metabolism
7.
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
8.
Biochem J ; 454(2): 345-57, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23789592

ABSTRACT

Integrins are heterodimeric cell-surface adhesion receptors that play a critical role in tissue development. Characterization of the full-length mRNA encoding the ß1 subunit (Itgb1) revealed an alternative functional cleavage and polyadenylation site that yields a new Itgb1 mRNA isoform 578 bp shorter than that previously reported. Using a variety of experimental and bioinformatic approaches, we found that the two Itgb1 isoforms are expressed at different levels in a variety of mouse tissues, including the mammary gland, where they are differentially regulated at successive developmental stages. The longer mRNA species is prevelant during lactation, whereas the shorter is induced after weaning. In 3D cultures, where expression of integrin ß1 protein is required for normal formation of acini, experimental blockade of the longer isoform induced enhanced expression of the shorter species which allowed normal morphological mammary differentiation. The short isoform lacks AU-rich motifs and miRNA target sequences that are potentially implicated in the regulation of mRNA stability and translation efficiency. We further determined that the AU-binding protein HuR appears to selectively stabilize the longer isoform in the mammary gland. In summary, the results of the present study identify a new regulatory instance involved in the fine-tuning of Itgb1 expression during mammary gland development and function.


Subject(s)
Integrin beta1/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , RNA Isoforms/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Messenger/metabolism , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Data Mining , Female , Gene Expression Regulation, Developmental , Integrin beta1/chemistry , Integrin beta1/genetics , Lactation/metabolism , Mammary Glands, Animal/cytology , Mice , Mice, Inbred BALB C , Polyadenylation , Pregnancy , RNA Isoforms/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , RNA, Small Interfering , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Weaning
9.
Breast Cancer Res Treat ; 135(3): 749-58, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22968621

ABSTRACT

Tristetraprolin (TTP) is a RNA-binding protein that inhibits the expression of pro-inflammatory cytokines and invasiveness-associated genes. TTP levels are decreased in many different cancer types and it has been proposed that this protein could be used as a prognostic factor in breast cancer. Here, using publicly available DNA microarray datasets, "serial analysis of gene expression" libraries and qRT-PCR analysis, we determined that TTP mRNA is present in normal breast cells and its levels are significantly decreased in all breast cancer subtypes. In addition, by immunostaining, we found that TTP expression is higher in normal breast tissue and benign lesions than in infiltrating carcinomas. Among these, lower grade tumors showed increased TTP expression compared to higher grade cancers. Therefore, these data indicate that TTP protein levels would provide a better negative correlation with breast cancer invasiveness than TTP transcript levels. In mice, we found that TTP mRNA and protein expression is also diminished in mammary tumors. Interestingly, a strong positive association of TTP expression and mammary differentiation was identified in normal and tumor cells. In fact, TTP expression is highly increased during lactation, showing good correlation with various mammary differentiation factors. TTP expression was also induced in mammary HC11 cells treated with lactogenic hormones, mainly by prolactin, through Stat5A activation. The effect of this hormone was highly dependent on mammary differentiation status, as prolactin was unable to elicit a similar response in proliferating or neoplastic mammary cells. In summary, these studies show that TTP expression is strongly linked to the mammary differentiation program in human and mice, suggesting that this protein might play specific and relevant roles in the normal physiology of the gland.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Tristetraprolin/genetics , Animals , Base Sequence , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Differentiation , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lactation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pregnancy , Prolactin/pharmacology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Tristetraprolin/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Mol Cell ; 41(1): 5-7, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21211718

ABSTRACT

In a recent issue of Molecular Cell, Kazanietz and colleagues (Sosa et al., 2010) show that P-Rex1, a Rac1 GEF, is overexpressed in ER+ and/or ErbB2+ breast cancers, suggesting that P-Rex1 might be a convergence node downstream of these receptors and an attractive therapeutic target.

11.
J Cell Biochem ; 110(4): 857-65, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20564184

ABSTRACT

It has been reported that expression of tumor necrosis factor superfamily members occur at the onset of the mammary gland post-lactational involution. One of these proteins, tumor necrosis factor alpha (TNFalpha), is a major mediator of inflammation that is able to induce expression of several cytokines. Leukemia inhibitory factor (LIF) is an inflammatory cytokine that is induced and plays a fundamental role during post-lactational involution of the mammary gland. Therefore, our goal was to determine whether TNFalpha activity in the mammary epithelium might include regulation of LIF expression. This biological role would increase the significance of TNFalpha expression at the end of lactation. Our results show that TNFalpha was able to induce LIF transcription through ERK1/2 activation in a non-tumorigenic mouse mammary epithelial cell line, SCp2. We found that activation of TNFalpha receptor-2 (TNFR2) was specifically involved in triggering this signaling pathway. In addition, our data suggest the participation of AP-1 transcription factor family members in this pathway. We determined that TNFalpha treatment induced c-fos transcription, and blocking AP-1 activity resulted in a significant inhibition of TNFalpha-induced LIF expression. Finally, we found that TNFalpha was also able to trigger LIF expression and ERK1/2 activation in the mouse mammary gland in vivo. Therefore, our data suggest that TNFalpha may contribute to mammary gland involution by, among other activities, eliciting LIF expression through ERK1/2 and AP1 activation.


Subject(s)
Leukemia Inhibitory Factor/metabolism , Mammary Glands, Human/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Blotting, Western , Cell Line , Electrophoretic Mobility Shift Assay , Enzyme Activation , Humans , Immunohistochemistry , Mammary Glands, Human/cytology , Mice , Mice, Inbred BALB C , Transcription Factor AP-1/metabolism
12.
J Virol ; 80(22): 11409-15, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16971449

ABSTRACT

Mice harboring three mouse mammary tumor virus (MMTV) variants develop pregnancy-dependent (PD) tumors that progress to pregnancy-independent (PI) behavior through successive passages. Herein, we identified 10 predominant insertions in PI transplants from 8 independent tumor lines. These mutations were also detected in small cell populations in the early PD passages. In addition, we identified a new viral insertion upstream of the gene Rspo3, which is overexpressed in three of the eight independent tumor lines and codes for a protein very similar to the recently described protein encoded by Int7. This study suggests that during progression towards hormone independence, clonal expansion of cells with specific mutations might be more relevant than the occurrence of new MMTV insertions.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/genetics , Mutation , Retroviridae Infections/virology , Selection, Genetic , Tumor Virus Infections/virology , Animals , Disease Models, Animal , Female , Mice , Recombination, Genetic
13.
Cancer Res ; 64(15): 5193-9, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15289324

ABSTRACT

Mouse mammary tumor virus (LA) induces pregnancy-dependent mammary tumors that progress toward autonomy. Here we show that in virgin females, pregnancy-dependent tumor transplants are able to remain dormant for up to 300 days. During that period, these tumors synthesize DNA, express high levels of estrogen and progesterone receptors (ER+PR+) and are able to resume growth after hormone stimulation. Surprisingly, in a subsequent transplant generation, all these tumors are fully able to grow in virgin females, they express low levels of ER and PR (ER-PR-) and have a monoclonal origin; i.e., show all of the features we have described previously in pregnancy-independent tumors. Histologically, mouse mammary tumor virus (LA)-induced tumors are morphologically similar to genetically engineered mouse (GEM) mammary tumors that overexpress genes belonging to the Wnt pathway. Interestingly, in the virus-induced neoplasias, pregnancy-independent passages arising after a dormant phase usually display a lower level of glandular differentiation together with epithelial cell trans-differentiation, a specific feature associated to Wnt pathway activation. In addition, dormancy can lead to the specific selection of Int2/Fgf3 mutated and overexpressing cells. Therefore, our results indicate that during hormone-dependent tumor dormancy, relevant changes in cell population occur, allowing rapid progression after changes in the animal internal milieu.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Pregnancy Complications, Neoplastic/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , Base Sequence , Cell Differentiation , Cell Division , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Fibroblast Growth Factor 3 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/pathogenicity , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Neoplasm Transplantation , Neoplasms, Hormone-Dependent , Pregnancy , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Time Factors , Wnt2 Protein
14.
Exp Cell Res ; 282(1): 35-47, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12490192

ABSTRACT

Leukemia inhibitory factor (LIF) is a multifunctional glycoprotein that displays multiple biological activities in different cell types, but to date there has been no report on its expression in the normal mammary gland. In this study we found that LIF is expressed at low but detectable levels in postpubertal, adult virgin, and pregnant mouse mammary glands. However, LIF expression drops after parturition to become almost undetectable in lactating glands. Interestingly, LIF expression shows a steep increase shortly after weaning that is maintained for the following 3 days. During this period, known as the first stage of mammary gland involution, the lack of suckling induces local factors that cause extensive epithelial cell death. It has been shown that Stat3 is the main factor in signaling the initiation of apoptosis, but the mechanism of its activation remains unclear. Herein, we show that LIF expression in the gland is induced by milk stasis and not by the decrease of circulating lactogenic hormones after weaning. Implantation of LIF containing pellets in lactating glands results in a significant increase in epithelium apoptosis. In addition, this treatment also induces Stat3 phosphorylation. We conclude that LIF regulated expression in the mouse mammary gland may play a relevant role during the first stage of mammary gland involution. Our results also show that LIF-induced mammary epithelium apoptosis could be mediated, at least partially, by Stat3 activation.


Subject(s)
Apoptosis/physiology , Epithelial Cells/metabolism , Growth Inhibitors/metabolism , Interleukin-6 , Lactation/physiology , Lymphokines/metabolism , Mammary Glands, Animal/metabolism , Animals , Apoptosis/drug effects , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Estrous Cycle/physiology , Female , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , Growth Inhibitors/genetics , Growth Inhibitors/pharmacology , Lactation/drug effects , Leukemia Inhibitory Factor , Leukemia Inhibitory Factor Receptor alpha Subunit , Lymphokines/genetics , Lymphokines/pharmacology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/drug effects , Mice , Mice, Inbred BALB C , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Cytokine/genetics , Receptors, OSM-LIF , STAT3 Transcription Factor , Trans-Activators/metabolism
15.
Breast Cancer Res Treat ; 75(3): 191-202, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12353808

ABSTRACT

In order to study mechanisms of progression of mouse mammary tumor virus (MMTV)-induced pregnancy-dependent mammary lesions, we removed and serially transplanted 17 small tumors detected in MMTV-infected pregnant females. This gave rise to the same number of 'in vivo' tumor lines. Hormone-dependency of the passages was determined by comparing tumor development in multiparous versus virgin hosts. We found that the first passages of most of these lesions (11/17) required pregnancy to grow. However, all these tumor lines lost their hormone-dependence through successive passages. The original pregnancy-dependent lesions were mostly multiclonal and showed high levels of estrogen and progesterone receptors. Alternatively, pregnancy-independent tumors arose as clonal dominant populations exhibiting a lower hormone receptor content. Our data show that the progression of hormone-dependent MMTV-induced mammary tumors is an irreversible process associated with the appearance of additional MMTV insertional events as well as alterations in the composition of the tumor cell population.


Subject(s)
Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/pathogenicity , Neoplasms, Hormone-Dependent/virology , Pregnancy, Animal , Animals , DNA, Neoplasm/metabolism , Disease Progression , Estrogens/metabolism , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Pregnancy , Progesterone/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...