Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
PLoS Genet ; 15(3): e1008002, 2019 03.
Article in English | MEDLINE | ID: mdl-30893315

ABSTRACT

Mammary epithelial progenitors are the normal cell-of-origin of breast cancer. We previously defined a population of p27+ quiescent hormone-responsive progenitor cells in the normal human breast whose frequency associates with breast cancer risk. Here, we describe that deletion of the Cdkn1b gene encoding the p27 cyclin-dependent kinase inhibitor in the estrogen-induced mammary tumor-susceptible ACI rat strain leads to a decrease in the relative frequencies of Cd49b+ mammary luminal epithelial progenitors and pregnancy-related differentiation. We show by comprehensive gene expression profiling of purified progenitor and differentiated mammary epithelial cell populations that p27 deletion has the most pronounced effects on luminal progenitors. Cdkn1b-/- females have decreased fertility, but rats that are able to get pregnant had normal litter size and were able to nurse their pups implying that loss of p27 in ACI rats does not completely abrogate ovarian function and lactation. Reciprocal mammary gland transplantation experiments indicate that the p27-loss-induced changes in mammary epithelial cells are not only caused by alterations in their intrinsic properties, but are likely due to altered hormonal signaling triggered by the perturbed systemic endocrine environment observed in Cdkn1b-/- females. We also observed a decrease in the frequency of mammary epithelial cells positive for progesterone receptor (Pr) and FoxA1, known direct transcriptional targets of the estrogen receptor (Erα), and an increase in phospho-Stat5 positive cells commonly induced by prolactin (Prl). Characterization of genome-wide Pr chromatin binding revealed distinct binding patterns in mammary epithelial cells of Cdkn1b+/+ and Cdkn1b-/- females and enrichment in genes with known roles in Notch, ErbB, leptin, and Erα signaling and regulation of G1-S transition. Our data support a role for p27 in regulating the pool size of hormone-responsive luminal progenitors that could impact breast cancer risk.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/physiology , Animals , Animals, Genetically Modified/genetics , Breast Neoplasms/genetics , Cell Differentiation , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endocrine Cells/physiology , Epithelial Cells , Estrogen Receptor alpha , Estrogens , Female , Genetic Predisposition to Disease/genetics , Humans , Integrin alpha1 , Mammary Glands, Animal , Mammary Glands, Human/growth & development , Pregnancy , Progesterone , Rats , Rats, Inbred ACI , Rats, Sprague-Dawley , Receptors, Estrogen , Receptors, Progesterone , Risk Factors , Signal Transduction , Stem Cells
2.
Physiol Genomics ; 50(3): 215-234, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29373076

ABSTRACT

Numerous laboratory and epidemiologic studies strongly implicate endogenous and exogenous estrogens in the etiology of breast cancer. Data summarized herein suggest that the ACI rat model of 17ß-estradiol (E2)-induced mammary cancer is unique among rodent models in the extent to which it faithfully reflects the etiology and biology of luminal types of breast cancer, which together constitute ~70% of all breast cancers. E2 drives cancer development in this model through mechanisms that are largely dependent upon estrogen receptors and require progesterone and its receptors. Moreover, mammary cancer development appears to be associated with generation of oxidative stress and can be modified by multiple dietary factors, several of which may attenuate the actions of reactive oxygen species. Studies of susceptible ACI rats and resistant COP or BN rats provide novel insights into the genetic bases of susceptibility and the biological processes regulated by genetic determinants of susceptibility. This review summarizes research progress resulting from use of these physiologically relevant rat models to advance understanding of breast cancer etiology and prevention.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/prevention & control , Estradiol/adverse effects , Mammary Neoplasms, Animal/pathology , Animals , Breast Neoplasms/genetics , Carcinogenesis/pathology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans
3.
Mamm Genome ; 29(1-2): 24-37, 2018 02.
Article in English | MEDLINE | ID: mdl-29487996

ABSTRACT

Breast cancer risk is intimately intertwined with exposure to estrogens. While more than 160 breast cancer risk loci have been identified in humans, genetic interactions with estrogen exposure remain to be established. Strains of rodents exhibit striking differences in their responses to endogenous ovarian estrogens (primarily 17ß-estradiol). Similar genetic variation has been observed for synthetic estrogen agonists (ethinyl estradiol) and environmental chemicals that mimic the actions of estrogens (xenoestrogens). This review of literature highlights the extent of variation in responses to estrogens among strains of rodents and compiles the genetic loci underlying pathogenic effects of excessive estrogen signaling. Genetic linkage studies have identified a total of the 35 quantitative trait loci (QTL) affecting responses to 17ß-estradiol or diethylstilbestrol in five different tissues. However, the QTL appear to act in a tissue-specific manner with 9 QTL affecting the incidence or latency of mammary tumors induced by 17ß-estradiol or diethylstilbestrol. Mammary gland development during puberty is also exquisitely sensitive to the actions of endogenous estrogens. Analysis of mammary ductal growth and branching in 43 strains of inbred mice identified 20 QTL. Regions in the human genome orthologous to the mammary development QTL harbor loci associated with breast cancer risk or mammographic density. The data demonstrate extensive genetic variation in regulation of estrogen signaling in rodent mammary tissues that alters susceptibility to tumors. Genetic variants in these pathways may identify a subset of women who are especially sensitive to either endogenous estrogens or environmental xenoestrogens and render them at increased risk of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Estrogens/genetics , Mammary Neoplasms, Animal/genetics , Quantitative Trait Loci/genetics , Animals , Breast Neoplasms/pathology , Estradiol/genetics , Estradiol/metabolism , Female , Genetic Predisposition to Disease , Genetic Variation , Humans , Mammary Glands, Human/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Risk Factors
4.
Mamm Genome ; 25(5-6): 244-52, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24448715

ABSTRACT

Estrogens control many aspects of pituitary gland biology, including regulation of lactotroph homeostasis and synthesis and secretion of prolactin. In rat models, these actions are strain specific and heritable, and multiple quantitative trait loci (QTL) have been mapped that impact the responsiveness of the lactotroph to estrogens. One such QTL, Ept7, was mapped to RNO7 in female progeny generated in an intercross between BN rats, in which the lactotroph population is insensitive to estrogens, and ACI rats, which develop lactotroph hyperplasia/adenoma and associated hyperprolactinemia in response to estrogen treatment. The primary objective of this study was to confirm the existence of Ept7 and to quantify the impact of this QTL on responsiveness of the pituitary gland of female and male rats to 17ß-estradiol (E2) and diethylstilbestrol (DES), respectively. Secondary objectives were to determine if Ept7 influences the responsiveness of the male reproductive tract to DES and to identify other discernible phenotypes influenced by Ept7. To achieve these objectives, a congenic rat strain that harbors BN alleles across the Ept7 interval on the genetic background of the ACI strain was generated and characterized to define the effect of administered estrogens on the anterior pituitary gland and male reproductive tissues. Data presented herein indicate Ept7 exerts a marked effect on development of lactotroph hyperplasia in response to estrogen treatment, but does not affect atrophy of the male reproductive tissues in response to hormone treatment. Ept7 was also observed to exert gender specific effects on body weight in young adult rats.


Subject(s)
Body Weight , Estrogens/metabolism , Pituitary Gland/metabolism , Rats/genetics , Alleles , Animals , Female , Male , Quantitative Trait Loci , Rats/growth & development , Rats/metabolism , Rats, Inbred ACI , Rats, Inbred BN
5.
BMC Cancer ; 13: 573, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24304664

ABSTRACT

BACKGROUND: We are using ACI and BN rats, which differ markedly in their susceptibility to 17ß-estradiol (E2)-induced mammary cancer, to identify genetic variants and environmental factors that determine mammary cancer susceptibility. The objective of this study was to characterize the cellular and molecular responses to E2 in the mammary glands of ACI and BN rats to identify qualitative and quantitative phenotypes that associate with and/or may confer differences in susceptibility to mammary cancer. METHODS: Female ACI and BN rats were treated with E2 for 1, 3 or 12 weeks. Mammary gland morphology and histology were examined by whole mount and hematoxylin and eosin (H&E) staining. Cell proliferation and epithelial density were evaluated by quantitative immunohistochemistry. Apoptosis was evaluated by quantitative western blotting and flow cytometry. Mammary gland differentiation was examined by immunohistochemistry. Gene expression was evaluated by microarray, qRT-PCR and quantitative western blotting assays. Extracellular matrix (ECM) associated collagen was evaluated by Picrosirius Red staining and Second Harmonic Generation (SHG) microscopy. RESULTS: The luminal epithelium of ACI rats exhibited a rapid and sustained proliferative response to E2. By contrast, the proliferative response exhibited by the mammary epithelium of BN rats was restrained and transitory. Moreover, the epithelium of BN rats appeared to undergo differentiation in response to E2, as evidenced by production of milk proteins as well as luminal ectasia and associated changes in the ECM. Marked differences in expression of genes that encode proteins with well-defined roles in mammary gland development (Pgr, Wnt4, Tnfsf11, Prlr, Stat5a, Areg, Gata3), differentiation and milk production (Lcn2, Spp1), regulation of extracellular environment (Mmp7, Mmp9), and cell-cell or cell-ECM interactions (Cd44, Cd24, Cd52) were observed. CONCLUSIONS: We propose that these cellular and molecular phenotypes are heritable and may underlie, at least in part, the differences in mammary cancer susceptibility exhibited by ACI and BN rats.


Subject(s)
Disease Susceptibility , Estradiol/administration & dosage , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Matrix/drug effects , Female , Gene Expression Regulation/drug effects , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/etiology , Phenotype , Rats , Species Specificity
6.
Curr Epidemiol Rep ; 9(1): 1-9, 2022.
Article in English | MEDLINE | ID: mdl-35223371

ABSTRACT

Purposeof Review: This review highlights six "best practices" for cancer epidemiology coordinating centers to facilitate the success of a research consortium. Recent Findings: Evidence from emerging literature regarding the Science of Team Science suggests that coordinating centers can more effectively foster collaborative cancer epidemiology research in consortia by (1) establishing collaboration as a shared goal at the start, (2) providing scientific expertise complementary to the research sites that adapts over the course of the project, (3) enacting anti-racist and inclusive approaches in all consortium decisions and activities, (4) fostering early-stage investigator career development, (5) engaging stakeholders including cancer survivors as peers, and (6) delivering reliable logistical support and technology tools with planned process evaluation so that researchers can collaboratively focus on the science. Summary: By drawing on the Science of Team Science, coordinating centers can accelerate research progress and increase the impact of cancer epidemiology consortia.

7.
Breast Cancer Res Treat ; 117(3): 517-24, 2009 Oct.
Article in English | MEDLINE | ID: mdl-18830694

ABSTRACT

The ACI rat is a unique model of human breast cancer in that mammary cancers are induced by estrogen without carcinogens, irradiation, xenografts or transgenic manipulations. We sought to characterize mammary cancers in a congenic variant of the ACI rat, the ACI.COP-Ept2. All rats with estradiol implants developed mammary cancers in 5-7 months. Rats bearing estradiol-induced mammary cancers were treated with tamoxifen for three weeks. Tamoxifen reduced tumor mass, measured by magnetic resonance imaging, by 89%. Tumors expressed estrogen receptors (ER), progesterone receptor (PR), and Erbb2. ERalpha and PR were overexpressed in tumor compared to adjacent non-tumor mammary gland. Thus, this model is highly relevant to hormone responsive human breast cancers.


Subject(s)
Estradiol/toxicity , Estrogens/toxicity , Mammary Neoplasms, Animal/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Animals , Animals, Congenic , Disease Models, Animal , Female , Humans , Immunohistochemistry , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/metabolism , Rats , Receptor, ErbB-2/biosynthesis , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis
8.
Endocrinology ; 149(8): 3850-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18420736

ABSTRACT

Ept1, Ept2, Ept6, and Ept9 are quantitative trait loci mapped in crosses between the ACI and Copenhagen (COP) rat strains as genetic determinants of responsiveness of the pituitary gland to estrogens. We have developed four congenic rat strains, each of which carries, on the genetic background of the ACI rat strain, alleles from the COP rat strain that span one of these quantitative trait loci. Relative to the female ACI rats, female ACI.COP-Ept1 rats exhibited reduced responsiveness to 17beta-estradiol (E2) in the pituitary gland, as evidenced by quantification of pituitary mass and circulating prolactin, and in the mammary gland, as evidenced by reduced susceptibility to E2-induced mammary cancer. The ACI.COP-Ept2 rat strain exhibited reduced responsiveness to E2 in the pituitary gland but did not differ from the ACI strain in regard to susceptibility to E2-induced mammary cancer. Interestingly, female Ept2 congenic rats exhibited increased responsiveness to E2 in the thymus, as evidenced by enhanced thymic atrophy. The ACI.COP-Ept6 rat strain exhibited increased responsiveness to E2 in the pituitary gland, which was associated with a qualitative phenotype suggestive of enhanced pituitary vascularization. The ACI.COP-Ept9 rat strain exhibited reduced responsiveness to E2 in the anterior pituitary gland, relative to the ACI rat strain. Neither Ept6 nor Ept9 impacted responsiveness to E2 in the mammary gland or thymus. These data indicate that each of these Ept genetic determinants of estrogen action is unique in regard to the tissues in which it exerts its effects and/or the direction of its effect on estrogen responsiveness.


Subject(s)
Drug Resistance/genetics , Estrogens/pharmacology , Pituitary Gland/drug effects , Quantitative Trait Loci/physiology , Animals , Animals, Congenic , Atrophy/genetics , Female , Genetic Markers/physiology , Genetic Predisposition to Disease , Incidence , Mammary Neoplasms, Animal/epidemiology , Mammary Neoplasms, Animal/genetics , Organ Size/drug effects , Organ Size/genetics , Organ Specificity/drug effects , Organ Specificity/genetics , Pituitary Gland/growth & development , Pituitary Gland/metabolism , Prolactin/blood , Rats , Thymus Gland/pathology
9.
Cancer Res ; 66(15): 7793-800, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885383

ABSTRACT

Exposure to estrogens is associated with an increased risk of breast cancer. Our laboratory has shown that the ACI rat is uniquely susceptible to 17beta-estradiol (E2)-induced mammary cancer. We previously mapped two loci, Emca1 and Emca2 (estrogen-induced mammary cancer), that act independently to determine susceptibility to E2-induced mammary cancer in crosses between the susceptible ACI rat strain and the genetically related, but resistant, Copenhagen (COP) rat strain. In this study, we evaluate susceptibility to E2-induced mammary cancer in a cross between the ACI strain and the unrelated Brown Norway (BN) rat strain. Whereas nearly 100% of the ACI rats developed mammary cancer when treated continuously with E2, BN rats did not develop palpable mammary cancer during the 196-day course of E2 treatment. Susceptibility to E2-induced mammary cancer segregated as a dominant or incompletely dominant trait in a cross between BN females and ACI males. In a population of 251 female (BN x ACI)F(2) rats, we observed evidence for a total of five genetic determinants of susceptibility. Two loci, Emca4 and Emca5, were identified when mammary cancer status at sacrifice was evaluated as the phenotype, and three additional loci, Emca6, Emca7, and Emca8, were identified when mammary cancer number was evaluated as the phenotype. A total of three genetic interactions were identified. These data indicate that susceptibility to E2-induced mammary cancer in the BN x ACI cross behaves as a complex trait controlled by at least five loci and multiple gene-gene interactions.


Subject(s)
Cocarcinogenesis , Estradiol/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/genetics , Animals , Chromosome Mapping , Female , Genetic Predisposition to Disease , Pituitary Neoplasms/genetics , Rats , Rats, Inbred ACI , Rats, Inbred BN
10.
PLoS One ; 13(9): e0204727, 2018.
Article in English | MEDLINE | ID: mdl-30261014

ABSTRACT

Pituitary adenoma is a common intracranial neoplasm that is observed in approximately 10% of unselected individuals at autopsy. Prolactin-producing adenomas, i.e., prolactinomas, comprise approximately 50% of all pituitary adenomas and represent the most common class of pituitary tumor. Multiple observations suggest that estrogens may contribute to development of prolactinoma; however, direct evidence for a causal role of estrogens in prolactinoma etiology is lacking. Rat models of estrogen-induced prolactinoma have been utilized extensively to identify the factors, pathways and processes that are involved in pituitary tumor development. The objective of this study was to localize to high resolution Ept7 (Estrogen-induced pituitary tumor), a quantitative trait locus (QTL) that controls lactotroph responsiveness to estrogens and was mapped to rat chromosome 7 (RNO7) in an intercross between BN and ACI rats. Data presented and discussed herein localize the Ept7 causal variant(s) to a 1.91 Mb interval of RNO7 that contains two protein coding genes, A1bg and Myc, and Pvt1, which yields multiple non-protein coding transcripts of unknown function. The Ept7 orthologous region in humans is located at 8q24.21 and has been linked in genome wide association studies to risk of 8 distinct epithelial cancers, including breast, ovarian, and endometrial cancers; 3 distinct types of B cell lymphoma; multiple inflammatory and autoimmune diseases; and orofacial cleft defects. In addition, the Ept7 locus in humans has been associated with variation in normal hematologic and development phenotypes, including height. Functional characterization of Ept7 should ultimately enhance our understanding of the genetic etiology of prolactinoma and these other diseases.


Subject(s)
Adenoma , Chromosomes, Mammalian/genetics , Estrogens , Lactotrophs/metabolism , Pituitary Neoplasms , Quantitative Trait Loci , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Estrogens/genetics , Estrogens/metabolism , Female , Genome-Wide Association Study , Humans , Hyperplasia , Lactotrophs/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Rats
11.
Proc Int Conf Mach Learn Appl ; 2018: 40-47, 2018 Dec.
Article in English | MEDLINE | ID: mdl-31799516

ABSTRACT

There is great interest in methods to improve human insight into trained non-linear models. Leading approaches include producing a ranking of the most relevant features, a non-trivial task for non-linear models. We show theoretically and empirically the benefit of a novel version of recursive feature elimination (RFE) as often used with SVMs; the key idea is a simple twist on the kinds of sensitivity testing employed in computational learning theory with membership queries (e.g., [1]). With membership queries, one can check whether changing the value of a feature in an example changes the label. In the real-world, we usually cannot get answers to such queries, so our approach instead makes these queries to a trained (imperfect) non-linear model. Because SVMs are widely used in bioinformatics, our empirical results use a real-world cancer genomics problem; because ground truth is not known for this task, we discuss the potential insights provided. We also evaluate on synthetic data where ground truth is known.

12.
Breast Dis ; 28: 69-86, 2007.
Article in English | MEDLINE | ID: mdl-18057545

ABSTRACT

The Norway rat has for many years been widely used as an experimental model for the study of breast cancer etiology and prevention. Mammary cancer can be induced in rats by a variety of agents. The mammary cancers that develop in the various rat models resemble in many respects the breast cancers that develop in humans. It is now clear that significant differences exist between different rat models with respect to the genetic bases of susceptibility to mammary cancer as well as the somatic genetic events that are associated with development of mammary cancer. In this review I summarize our current understanding of the genetic and genomics of mammary cancer in the rat, compare and contrast the genetic/genomic features of different rat mammary cancer models and discuss the relevance of these models to breast cancer in humans.


Subject(s)
Genetic Predisposition to Disease , Genomics , Mammary Neoplasms, Experimental/genetics , Animals , Animals, Outbred Strains , Chromosome Mapping , Female , Genomic Instability , Quantitative Trait Loci , Rats , Rats, Inbred Strains
13.
Genetics ; 169(4): 2189-97, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15687265

ABSTRACT

Estrogens stimulate proliferation and enhance survival of the prolactin (PRL)-producing lactotroph of the anterior pituitary gland and induce development of PRL-producing pituitary tumors in certain inbred rat strains but not others. The goal of this study was to elucidate the genetic bases of estrogen-induced pituitary tumorigenesis in reciprocal intercrosses between the genetically related ACI and Copenhagen (COP) rat strains. Following 12 weeks of treatment with the synthetic estrogen diethylstilbestrol (DES), pituitary mass, an accurate surrogate marker of absolute lactotroph number, was increased 10.6-fold in ACI rats and 4.5-fold in COP rats. Composite interval mapping analyses of the phenotypically defined F(2) progeny from the reciprocal crosses identified six quantitative trait loci (QTL) that determine the pituitary growth response to DES. These loci reside on chromosome 6 [Estrogen-induced pituitary tumor (Ept)1], chromosome 3 (Ept2 and Ept6), chromosome 10 (Ept9), and chromosome 1 (Ept10 and Ept13). Together, these six Ept loci and one additional suggestive locus on chromosome 4 account for an estimated 40% of the phenotypic variance exhibited by the combined F(2) population, while 34% of the phenotypic variance was estimated to result from environmental factors. These data indicate that DES-induced pituitary mass behaves as a quantitative trait and provide information that will facilitate identification of genes that determine the tumorigenic response of the pituitary gland to estrogens.


Subject(s)
Estrogens/pharmacology , Pituitary Gland/drug effects , Pituitary Neoplasms/chemically induced , Pituitary Neoplasms/genetics , Animals , Biomarkers , Chromosome Mapping , Crosses, Genetic , Epistasis, Genetic , Estrogens/metabolism , Female , Gene Expression Regulation, Neoplastic , Genetic Markers , Genotype , Male , Models, Genetic , Models, Statistical , Organ Size , Phenotype , Pituitary Gland/pathology , Prolactin/metabolism , Quantitative Trait Loci , Rats , Sensitivity and Specificity , Sex Factors , Species Specificity
14.
Genetics ; 168(4): 2113-25, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15611180

ABSTRACT

Hormonal, genetic, and environmental factors play major roles in the complex etiology of breast cancer. When treated continuously with 17beta-estradiol (E2), the ACI rat exhibits a genetically conferred propensity to develop mammary cancer. The susceptibility of the ACI rat to E2-induced mammary cancer appears to segregate as an incompletely dominant trait in crosses to the resistant Copenhagen (COP) strain. In both (ACI x COP)F(2) and (COP x ACI)F(2) populations, we find strong evidence for a major genetic determinant of susceptibility to E2-induced mammary cancer on distal rat chromosome 5. Our data are most consistent with a model in which the ACI allele of this locus, termed Emca1 (estrogen-induced mammary cancer 1), acts in an incompletely dominant manner to increase both tumor incidence and tumor multiplicity as well as to reduce tumor latency in these populations. We also find evidence suggestive of a second locus, Emca2, on chromosome 18 in the (ACI x COP)F(2) population. The ACI allele of Emca2 acts in a dominant manner to increase incidence and decrease latency. Together, Emca1 and Emca2 act independently to modify susceptibility to E2-induced mammary cancer.


Subject(s)
Chromosome Mapping , Estradiol/pharmacology , Genetic Predisposition to Disease , Mammary Neoplasms, Experimental/genetics , Animals , Female , Genetic Linkage , Genetic Markers , Mammary Neoplasms, Experimental/chemically induced , Rats , Rats, Inbred ACI
15.
Endocr Relat Cancer ; 22(2): 239-48, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25800038

ABSTRACT

The ACI rat model of 17ß-estradiol (E2)-induced mammary cancer is highly relevant for use in establishing the endocrine, genetic, and environmental bases of breast cancer etiology and identifying novel agents and strategies for preventing breast cancer. E2 treatment rapidly induces mammary cancer in female ACI rats and simultaneously induces pituitary lactotroph hyperplasia and adenoma. The pituitary tumors can result in undesired morbidity, which compromises long-term studies focused on mammary cancer etiology and prevention. We have defined the genetic bases of susceptibility to E2-induced mammary cancers and pituitary tumors and have utilized the knowledge gained in these studies to develop a novel inbred rat strain, designated ACWi, that retains the high degree of susceptibility to E2-induced mammary cancer exhibited by ACI rats, but lacks the treatment-related morbidity associated with pituitary lactotroph hyperplasia/adenoma. When treated with E2, female ACWi rats developed palpable mammary cancer at a median latency of 116 days, an incidence of 100% by 161 days and exhibited an average of 15.6 mammary tumors per rat following 196 days of treatment. These parameters did not differ from those observed for contemporaneously treated ACI rats. None of the E2-treated ACWi rats were killed before the intended experimental end point due to any treatment-related morbidity other than mammary cancer burden, whereas 20% of contemporaneously treated ACI rats exhibited treatment-related morbidity that necessitated premature killing. The ACWi rat strain is well suited for use by those in the research community, focusing on breast cancer etiology and prevention.


Subject(s)
Mammary Neoplasms, Experimental , Rats, Inbred Strains , Animals , Estradiol , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Phenotype , Pituitary Gland/pathology
16.
Cancer Prev Res (Phila) ; 8(9): 807-16, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26130252

ABSTRACT

This study evaluated the anticancer activity and mechanism of action of a γ-tocopherol-rich tocopherol mixture, γ-TmT, in two different animal models of estrogen-induced breast cancer. The chemopreventive effect of γ-TmT at early (6 weeks), intermediate (18 weeks), and late (31 weeks) stages of mammary tumorigenesis was determined using the August-Copenhagen Irish rat model. Female rats receiving 17ß-estradiol (E2) implants were administered with different doses (0%, 0.05%, 0.1%, 0.3%, and 0.5%) of γ-TmT diet. Treatment with 0.3% and 0.5% γ-TmT decreased tumor volume and multiplicity. At 31 weeks, serum concentrations of E2 were significantly decreased by γ-TmT. γ-TmT preferentially induced expression of the E2-metabolizing enzyme CYP1A1, over CYP1B1 in the rat mammary tissues. Nrf2-dependent antioxidant response was stimulated by γ-TmT, as evident from enhanced expression of its downstream targets, NQO1, GCLM, and HMOX1. Serum concentrations of the oxidative stress marker, 8-isoprostane, were also decreased in the γ-TmT-treated groups. Treatment with γ-TmT increased expression of PPARγ and its downstream genes, PTEN and p27, whereas the cell proliferation marker, PCNA, was significantly reduced in γ-TmT-treated mammary tumors. In an orthotopic model in which human MCF-7 breast cancer cells were injected into the mammary fat pad of immunodeficient mice, γ-TmT inhibited E2-dependent tumor growth at all the doses tested. In conclusion, γ-TmT reduced mammary tumor development, in part through decreased E2 availability and reduced oxidative stress in mammary tissues; γ-TmT could thus be an effective agent for the prevention and treatment of E2-induced breast cancer.


Subject(s)
Antioxidants/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Estrogens/metabolism , PPAR gamma/metabolism , PTEN Phosphohydrolase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , gamma-Tocopherol/therapeutic use , Animals , Cell Line, Tumor , Dinoprost/analogs & derivatives , Dinoprost/chemistry , Estradiol/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Immunohistochemistry , MCF-7 Cells , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Nude , Neoplasm Transplantation , Oxidative Stress , RNA, Messenger/metabolism , Rats , Time Factors
17.
PLoS One ; 10(2): e0118147, 2015.
Article in English | MEDLINE | ID: mdl-25693193

ABSTRACT

Congenital anomalies of the kidney and urogenital tract (CAKUT) occur in approximately 0.5% of live births and represent the most frequent cause of end-stage renal disease in neonates and children. The genetic basis of CAKUT is not well defined. To understand more fully the genetic basis of one type of CAKUT, unilateral renal agenesis (URA), we are studying inbred ACI rats, which spontaneously exhibit URA and associated urogenital anomalies at an incidence of approximately 10%. URA is inherited as an incompletely dominant trait with incomplete penetrance in crosses between ACI and Brown Norway (BN) rats and a single responsible genetic locus, designated Renag1, was previously mapped to rat chromosome 14 (RNO14). The goals of this study were to fine map Renag1, identify the causal genetic variant responsible for URA, confirm that the Renag1 variant is the sole determinant of URA in the ACI rat, and define the embryologic basis of URA in this rat model. Data presented herein localize Renag1 to a 379 kilobase (kb) interval that contains a single protein coding gene, Kit (v-kit Hardy-Zukerman 4 feline sarcoma viral oncogene homolog); identify an endogenous retrovirus-derived long terminal repeat located within Kit intron 1 as the probable causal variant; demonstrate aberrant development of the nephric duct in the anticipated number of ACI rat embryos; and demonstrate expression of Kit and Kit ligand (Kitlg) in the nephric duct. Congenic rats that harbor ACI alleles at Renag1 on the BN genetic background exhibit the same spectrum of urogenital anomalies as ACI rats, indicating that Renag1 is necessary and sufficient to elicit URA and associated urogenital anomalies. These data reveal the first genetic link between Kit and URA and illustrate the value of the ACI rat as a model for defining the mechanisms and cell types in which Kit functions during urogenital development.


Subject(s)
Chromosomes, Mammalian/genetics , Congenital Abnormalities/genetics , Congenital Abnormalities/pathology , Kidney Diseases/congenital , Kidney/abnormalities , Physical Chromosome Mapping/methods , Proto-Oncogene Proteins c-kit/genetics , Animals , Disease Models, Animal , Genetic Association Studies , Genetic Loci , Genetic Predisposition to Disease , Genetic Variation , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Rats , Rats, Inbred ACI , Stem Cell Factor/genetics
18.
G3 (Bethesda) ; 4(8): 1385-94, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24875630

ABSTRACT

When treated with 17ß-estradiol, female ACI rats (Rattus norvegicus) rapidly develop mammary cancers that share multiple phenotypes with luminal breast cancers. Seven distinct quantitative trait loci that harbor genetic determinants of susceptibility to 17ß-estradiol-induced mammary cancer have been mapped in reciprocal intercrosses between susceptible ACI rats and resistant Brown Norway (BN) rats. A panel of unique congenic rat strains has now been generated and characterized to confirm the existence of these quantitative trait loci, designated Emca3 through Emca9, and to quantify their individual effects on susceptibility to 17ß-estradiol-induced mammary cancer. Each congenic strain carries BN alleles spanning an individual Emca locus, introgressed onto the ACI genetic background. Data presented herein indicate that BN alleles at Emca3, Emca4, Emca5, Emca6, and Emca9 reduce susceptibility to 17ß-estradiol-induced mammary cancer, whereas BN alleles at Emca7 increase susceptibility, thereby confirming the previous interval mapping data. All of these Emca loci are orthologous to regions of the human genome that have been demonstrated in genome-wide association studies to harbor genetic variants that influence breast cancer risk. Moreover, four of the Emca loci are orthologous to loci in humans that have been associated with mammographic breast density, a biomarker of breast cancer risk. This study further establishes the relevance of the ACI and derived congenic rat models of 17ß-estradiol-induced mammary cancer for defining the genetic bases of breast cancer susceptibility and elucidating the mechanisms through which 17ß-estradiol contributes to breast cancer development.


Subject(s)
Genetic Predisposition to Disease , Mammary Neoplasms, Animal/genetics , Quantitative Trait Loci , Animals , Animals, Congenic , Estradiol , Estrogens , Female , Humans , Mammary Neoplasms, Animal/chemically induced , Phenotype , Rats, Inbred BN , Risk
19.
Cancer Res ; 74(22): 6419-29, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25172839

ABSTRACT

The majority of causative variants in familial breast cancer remain unknown. Of the known risk variants, most are tumor cell autonomous, and little attention has been paid yet to germline variants that may affect the tumor microenvironment. In this study, we developed a system called the Consomic Xenograft Model (CXM) to map germline variants that affect only the tumor microenvironment. In CXM, human breast cancer cells are orthotopically implanted into immunodeficient consomic strains and tumor metrics are quantified (e.g., growth, vasculogenesis, and metastasis). Because the strain backgrounds vary, whereas the malignant tumor cells do not, any observed changes in tumor progression are due to genetic differences in the nonmalignant microenvironment. Using CXM, we defined genetic variants on rat chromosome 3 that reduced relative tumor growth and hematogenous metastasis in the SS.BN3(IL2Rγ) consomic model compared with the SS(IL2Rγ) parental strain. Paradoxically, these effects occurred despite an increase in the density of tumor-associated blood vessels. In contrast, lymphatic vasculature and lymphogenous metastasis were unaffected by the SS.BN3(IL2Rγ) background. Through comparative mapping and whole-genome sequence analysis, we narrowed candidate variants on rat chromosome 3 to six genes with a priority for future analysis. Collectively, our results establish the utility of CXM to localize genetic variants affecting the tumor microenvironment that underlie differences in breast cancer risk.


Subject(s)
Breast Neoplasms/etiology , Tumor Microenvironment , 9,10-Dimethyl-1,2-benzanthracene , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Lymphangiogenesis , Male , Neoplasm Transplantation , Quantitative Trait Loci , Rats , Risk , Transplantation, Heterologous
20.
Cancer Prev Res (Phila) ; 6(1): 59-69, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23151807

ABSTRACT

The ACI rat model of 17ß-estradiol (E2)-induced mammary cancer has gained wide use in the study of breast cancer etiology, prevention, and genetics. Emca8, a QTL that determines susceptibility to E2-induced mammary cancer, was previously mapped to rat chromosome 5 (RNO5) in an intercross between resistant Brown Norway (BN) and susceptible ACI rats. In this study, a panel of congenic rat strains, each of which carries BN alleles across a defined segment of RNO5 on the ACI genetic background, was generated and used to map more precisely the Emca8 determinants of mammary cancer susceptibility. Three distinct genetic determinants were localized within Emca8, and two of these were mapped to intervals of less than 15 megabases. Emca8.1 harbors Cdkn2a, Cdkn2b, and other genes and is orthologous to the 9p21 breast cancer locus identified in genome-wide and candidate gene association studies. Emca8.2 harbors Cdkn2c and other genes and is orthologous to the 1p32 locus in humans that is frequently deleted in breast cancers. Both Emca8.1 and Emca8.2 harbor copy number variants that are orthologous to copy number variant regions in humans. Gene expression profiles were defined for mammary tissues from E2-treated ACI and ACI.BN-Emca8 rats to define the impact of Emca8 on gene expression and identify differentially expressed genes residing within Emca8.1 and Emca8.2. This study further illustrates the relevance of the ACI rat model of E2-induced mammary cancer for identifying novel genetic determinants of breast cancer susceptibility and defining the mechanisms through which estrogens contribute to breast cancer development. Cancer Prev Res; 6(1); 59-69. ©2012 AACR.


Subject(s)
Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Mammary Neoplasms, Animal/genetics , Alleles , Animals , Animals, Congenic , Chromosome Mapping/methods , Comparative Genomic Hybridization , Crosses, Genetic , Estradiol/metabolism , Female , Gene Dosage , Genetic Variation , Genotype , Phenotype , Quantitative Trait Loci , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL