Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Sci Rep ; 11(1): 6351, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33737566

RESUMO

Perillyl alcohol (POH) has been extensively studied for the treatment of peripheral and primary brain tumors. The intranasal route of administration has been preferred for dosing POH in early-stage clinical trials associated with promising outcomes in primary brain cancer. However, it is unclear how intranasal POH targets brain tumors in these patients. Multiple studies indicate that intranasally applied large molecules may enter the brain and cerebrospinal fluid (CSF) through direct olfactory and trigeminal nerve-associated pathways originating in the nasal mucosa that bypass the blood-brain barrier. It is unknown whether POH, a small molecule subject to extensive nasal metabolism and systemic absorption, may also undergo direct transport to brain or CSF from the nasal mucosa. Here, we compared CSF and plasma concentrations of POH and its metabolite, perillic acid (PA), following intranasal or intravascular POH application. Samples were collected over 70 min and assayed by high-performance liquid chromatography. Intranasal administration resulted in tenfold higher CSF-to-plasma ratios for POH and tenfold higher CSF levels for PA compared to equal dose intravascular administration. Our preclinical results demonstrate POH undergoes direct transport from the nasal mucosa to the CSF, a finding with potential significance for its efficacy as an intranasal chemotherapeutic for brain cancer.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Monoterpenos/farmacologia , Mucosa Nasal/efeitos dos fármacos , Administração Intranasal , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/patologia , Neoplasias Encefálicas/líquido cefalorraquidiano , Neoplasias Encefálicas/patologia , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Humanos , Ratos , Nervo Trigêmeo/efeitos dos fármacos
2.
Front Genet ; 12: 788318, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35087569

RESUMO

Cancer risk by environmental exposure is modulated by an individual's genetics and age at exposure. This age-specific period of susceptibility is referred to as the "Window of Susceptibility" (WOS). Rats have a similar WOS for developing breast cancer. A previous study in rat identified an age-specific long-range regulatory interaction for the cancer gene, Pappa, that is associated with breast cancer susceptibility. However, the global role of three-dimensional genome organization and downstream gene expression programs in the WOS is not known. Therefore, we generated Hi-C and RNA-seq data in rat mammary epithelial cells within and outside the WOS. To systematically identify higher-order changes in 3D genome organization, we developed NE-MVNMF that combines network enhancement followed by multitask non-negative matrix factorization. We examined three-dimensional genome organization dynamics at the level of individual loops as well as higher-order domains. Differential chromatin interactions tend to be associated with differentially up-regulated genes with the WOS and recapitulate several human SNP-gene interactions associated with breast cancer susceptibility. Our approach identified genomic blocks of regions with greater overall differences in contact count between the two time points when the cluster assignments change and identified genes and pathways implicated in early carcinogenesis and cancer treatment. Our results suggest that WOS-specific changes in 3D genome organization are linked to transcriptional changes that may influence susceptibility to breast cancer.

3.
J Vis Exp ; (157)2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32202519

RESUMO

As early as the 1970s, researchers have successfully transplanted mammary epithelial cells into the interscapular white fat pad of rats. Grafting mammary epithelium using transplantation techniques takes advantage of the hormonal environment provided by the adolescent rodent host. These studies are ideally suited to explore the impact of various biological manipulations on mammary gland development and dissect many aspects of mammary gland biology. A common, but limiting, feature is that transplanted epithelial cells are strongly influenced by the surrounding stroma and outcompeted by endogenous epithelium; to utilize native mammary tissue, the abdominal-inguinal white fat pad must be cleared to remove host mammary epithelium prior to the transplantation. A major obstacle when using the rat model organism is that clearing the developing mammary tree in post-weaned rats is not efficient. When transplanted into gland-free fat pads, donor epithelial cells can repopulate the cleared host fat pad and form a functional mammary gland. The interscapular fat pad is an alternative location for these grafts. A major advantage is that it lacks ductal structures yet provides the normal stroma that is necessary to promote epithelial outgrowth and is easily accessible in the rat. Another major advantage of this technique is that it is minimally invasive, because it eliminates the need to cauterize and remove the growing endogenous mammary tree. Additionally, the interscapular fat pad contains a medial blood vessel that can be used to separate sites for grafting. Because the endogenous glands remain intact, this technique can also be used for studies comparing the endogenous mammary gland to the transplanted gland. This paper describes the method of mammary epithelial cell transplantation into the interscapular white fat pad of rats.


Assuntos
Tecido Adiposo/citologia , Transplante de Células/métodos , Células Epiteliais/transplante , Glândulas Mamárias Animais/citologia , Tecido Adiposo/cirurgia , Tecido Adiposo Branco/citologia , Animais , Feminino , Ratos
4.
BMC Cancer ; 18(1): 1233, 2018 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-30526553

RESUMO

BACKGROUND: The gene desert on human chromosomal band 8q24 harbors multiple genetic variants associated with common cancers, including breast cancer. The locus, including the gene desert and its flanking genes, MYC, PVT1 and FAM84B, is also frequently amplified in human breast cancer. We generated a megadeletion (MD) mouse model lacking 430-Kb of sequence orthologous to the breast cancer-associated region in the gene desert. The goals were to examine the effect of the deletion on mammary cancer development and on transcript level regulation of the candidate genes within the locus. METHODS: The MD allele was engineered using the MICER system in embryonic stem cells and bred onto 3 well-characterized transgenic models for breast cancer, namely MMTV-PyVT, MMTV-neu and C3(1)-TAg. Mammary tumor growth, latency, multiplicity and metastasis were compared between homozygous MD and wild type mice carrying the transgenes. A reciprocal mammary gland transplantation assay was conducted to distinguish mammary cell-autonomous from non-mammary cell-autonomous anti-cancer effects. Gene expression analysis was done using quantitative real-time PCR. Chromatin interactions were evaluated by 3C. Gene-specific patient outcome data were analysed using the METABRIC and TCGA data sets through the cBioPortal website. RESULTS: Mice homozygous for the MD allele are viable, fertile, lactate sufficiently to nourish their pups, but maintain a 10% lower body weight mainly due to decreased adiposity. The deletion interferes with mammary tumorigenesis in mouse models for luminal and basal breast cancer. In the MMTV-PyVT model the mammary cancer-reducing effects of the allele are mammary cell-autonomous. We found organ-specific effects on transcript level regulation, with Myc and Fam84b being downregulated in mammary gland, prostate and mammary tumor samples. Through analysis using the METABRIC and TCGA datasets, we provide evidence that MYC and FAM84B are frequently co-amplified in breast cancer, but in contrast with MYC, FAM84B is frequently overexpressed in the luminal subtype, whereas MYC activity affect basal breast cancer outcomes. CONCLUSION: Deletion of a breast cancer-associated non-protein coding region affects mammary cancer development in 3 transgenic mouse models. We propose Myc as a candidate susceptibility gene, regulated by the gene desert locus, and a potential role for Fam84b in modifying breast cancer development.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Genes myc/genética , Neoplasias Mamárias Experimentais/genética , Proteínas de Neoplasias/genética , Animais , Feminino , Técnicas de Inativação de Genes , Proteínas de Membrana , Camundongos , Camundongos Transgênicos
5.
Breast Cancer Res ; 20(1): 111, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30208917

RESUMO

BACKGROUND: Antimicrobial T cells play key roles in the disease progression of cancers arising in mucosal epithelial tissues, such as the colon. However, little is known about microbe-reactive T cells within human breast ducts and whether these impact breast carcinogenesis. METHODS: Epithelial ducts were isolated from primary human breast tissue samples, and the associated T lymphocytes were characterized using flow cytometric analysis. Functional assays were performed to determine T-cell cytokine secretion in response to bacterially treated human breast carcinoma cells. RESULTS: We show that human breast epithelial ducts contain mucosal associated invariant T (MAIT) cells, an innate T-cell population that recognizes specific bacterial metabolites presented by nonclassical MR1 antigen-presenting molecules. The MAIT cell population from breast ducts resembled that of peripheral blood in its innate lymphocyte phenotype (i.e., CD161, PLZF, and interleukin [IL]-18 receptor coexpression), but the breast duct MAIT cell population had a distinct T-cell receptor Vß use profile and was markedly enriched for IL-17-producing cells compared with blood MAIT cells. Breast carcinoma cells that had been exposed to Escherichia coli activated MAIT cells in an MR1-dependent manner. However, whereas phorbol 12-myristate 13-acetate/ionomycin stimulation induced the production of both interferon-γ and IL-17 by breast duct MAIT cells, bacterially exposed breast carcinoma cells elicited a strongly IL-17-biased response. Breast carcinoma cells also showed upregulated expression of natural killer group 2 member D (NKG2D) ligands compared with primary breast epithelial cells, and the NKG2D receptor contributed to MAIT cell activation by the carcinoma cells. CONCLUSIONS: These results demonstrate that MAIT cells from human breast ducts mediate a selective T-helper 17 cell response to human breast carcinoma cells that were exposed to E. coli. Thus, cues from the breast microbiome and the expression of stress-associated ligands by neoplastic breast duct epithelial cells may shape MAIT cell responses during breast carcinogenesis.


Assuntos
Neoplasias da Mama/imunologia , Células Epiteliais/imunologia , Interleucina-17/imunologia , Células T Invariantes Associadas à Mucosa/imunologia , Mama/citologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/microbiologia , Linhagem Celular Tumoral , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Células Epiteliais/metabolismo , Escherichia coli/imunologia , Escherichia coli/fisiologia , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/metabolismo , Ativação Linfocitária/imunologia , Antígenos de Histocompatibilidade Menor/genética , Antígenos de Histocompatibilidade Menor/imunologia , Células T Invariantes Associadas à Mucosa/metabolismo
6.
PLoS Genet ; 12(8): e1006261, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27537370

RESUMO

In understanding the etiology of breast cancer, the contributions of both genetic and environmental risk factors are further complicated by the impact of breast developmental stage. Specifically, the time period ranging from childhood to young adulthood represents a critical developmental window in a woman's life when she is more susceptible to environmental hazards that may affect future breast cancer risk. Although the effects of environmental exposures during particular developmental Windows of Susceptibility (WOS) are well documented, the genetic mechanisms governing these interactions are largely unknown. Functional characterization of the Mammary Carcinoma Susceptibility 5c, Mcs5c, congenic rat model of breast cancer at various stages of mammary gland development was conducted to gain insight into the interplay between genetic risk factors and WOS. Using quantitative real-time PCR, chromosome conformation capture, and bisulfite pyrosequencing we have found that Mcs5c acts within the mammary gland to regulate expression of the neighboring gene Pappa during a critical mammary developmental time period in the rat, corresponding to the human young adult WOS. Pappa has been shown to positively regulate the IGF signaling pathway, which is required for proper mammary gland/breast development and is of increasing interest in breast cancer pathogenesis. Mcs5c-mediated regulation of Pappa appears to occur through age-dependent and mammary gland-specific chromatin looping, as well as genotype-dependent CpG island shore methylation. This represents, to our knowledge, the first insight into cellular mechanisms underlying the WOS phenomenon and demonstrates the influence developmental stage can have on risk locus functionality. Additionally, this work represents a novel model for further investigation into how environmental factors, together with genetic factors, modulate breast cancer risk in the context of breast developmental stage.


Assuntos
Neoplasias da Mama/genética , Metilação de DNA/genética , Predisposição Genética para Doença , Neoplasias Mamárias Experimentais/genética , Somatomedinas/genética , Animais , Neoplasias da Mama/patologia , Cromatina/genética , Ilhas de CpG/genética , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Glândulas Mamárias Humanas/patologia , Neoplasias Mamárias Experimentais/patologia , Locos de Características Quantitativas/genética , Ratos , Transdução de Sinais
7.
Cancer Prev Res (Phila) ; 9(4): 305-16, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26811335

RESUMO

Developing strategies to enhance cancer prevention is a paramount goal, particularly given recent concerns about surgical treatment of preinvasive states such as ductal carcinoma in situ. Promoting effective immunosurveillance by leukocytes that scan for nascent neoplastic transformations represents a potential means to achieve this goal. Because most breast cancers arise within the ductal epithelium, enhancing protective immunosurveillance will likely necessitate targeting one or more of the distinctive lymphocyte types found in these sites under normal conditions. Here, we have characterized the intraepithelial lymphocyte compartment of non-cancerous human breast tissue and identified a subset of T lymphocytes that can be pharmacologically targeted to enhance their responses to breast cancer cells. Specifically, Vδ2(+) γδ T cells were consistently present in preparations of mammary ductal epithelial organoids and they proliferated in response to zoledronic acid, an aminobisphosphonate drug. Vδ2(+) T cells from breast ductal organoids produced the antitumor cytokine IFNγ and efficiently killed bisphosphonate-pulsed breast carcinoma cells. These findings demonstrate the potential for exploiting the ability of Vδ2(+) γδ T cells to respond to FDA-approved bisphosphonate drugs as a novel immunotherapeutic approach to inhibit the outgrowth of breast cancers.


Assuntos
Carcinoma Intraductal não Infiltrante/imunologia , Difosfonatos/farmacologia , Imidazóis/farmacologia , Interferon gama/metabolismo , Glândulas Mamárias Humanas/citologia , Organoides/citologia , Linfócitos T/imunologia , Carcinoma Intraductal não Infiltrante/prevenção & controle , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Feminino , Humanos , Vigilância Imunológica/efeitos dos fármacos , Interferon gama/imunologia , Glândulas Mamárias Humanas/imunologia , Cultura Primária de Células , Linfócitos T/metabolismo , Ácido Zoledrônico
8.
PLoS Genet ; 9(6): e1003549, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23785296

RESUMO

Genome-wide association studies have revealed that many low-penetrance breast cancer susceptibility loci are located in non-protein coding genomic regions; however, few have been characterized. In a comparative genetics approach to model such loci in a rat breast cancer model, we previously identified the mammary carcinoma susceptibility locus Mcs1a. We now localize Mcs1a to a critical interval (277 Kb) within a gene desert. Mcs1a reduces mammary carcinoma multiplicity by 50% and acts in a mammary cell-autonomous manner. We developed a megadeletion mouse model, which lacks 535 Kb of sequence containing the Mcs1a ortholog. Global gene expression analysis by RNA-seq revealed that in the mouse mammary gland, the orphan nuclear receptor gene Nr2f1/Coup-tf1 is regulated by Mcs1a. In resistant Mcs1a congenic rats, as compared with susceptible congenic control rats, we found Nr2f1 transcript levels to be elevated in mammary gland, epithelial cells, and carcinoma samples. Chromatin looping over ∼820 Kb of sequence from the Nr2f1 promoter to a strongly conserved element within the Mcs1a critical interval was identified. This element contains a 14 bp indel polymorphism that affects a human-rat-mouse conserved COUP-TF binding motif and is a functional Mcs1a candidate. In both the rat and mouse models, higher Nr2f1 transcript levels are associated with higher abundance of luminal mammary epithelial cells. In both the mouse mammary gland and a human breast cancer global gene expression data set, we found Nr2f1 transcript levels to be strongly anti-correlated to a gene cluster enriched in cell cycle-related genes. We queried 12 large publicly available human breast cancer gene expression studies and found that the median NR2F1 transcript level is consistently lower in 'triple-negative' (ER-PR-HER2-) breast cancers as compared with 'receptor-positive' breast cancers. Our data suggest that the non-protein coding locus Mcs1a regulates Nr2f1, which is a candidate modifier of differentiation, proliferation, and mammary cancer risk.


Assuntos
Neoplasias da Mama/genética , Fator I de Transcrição COUP/genética , Diferenciação Celular/genética , Loci Gênicos , Predisposição Genética para Doença , Neoplasias Mamárias Animais/genética , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator I de Transcrição COUP/metabolismo , Proliferação de Células , Mapeamento Cromossômico , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Ratos
9.
Bioinformatics ; 29(8): 1035-43, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23428641

RESUMO

MOTIVATION: Messenger RNA expression is important in normal development and differentiation, as well as in manifestation of disease. RNA-seq experiments allow for the identification of differentially expressed (DE) genes and their corresponding isoforms on a genome-wide scale. However, statistical methods are required to ensure that accurate identifications are made. A number of methods exist for identifying DE genes, but far fewer are available for identifying DE isoforms. When isoform DE is of interest, investigators often apply gene-level (count-based) methods directly to estimates of isoform counts. Doing so is not recommended. In short, estimating isoform expression is relatively straightforward for some groups of isoforms, but more challenging for others. This results in estimation uncertainty that varies across isoform groups. Count-based methods were not designed to accommodate this varying uncertainty, and consequently, application of them for isoform inference results in reduced power for some classes of isoforms and increased false discoveries for others. RESULTS: Taking advantage of the merits of empirical Bayesian methods, we have developed EBSeq for identifying DE isoforms in an RNA-seq experiment comparing two or more biological conditions. Results demonstrate substantially improved power and performance of EBSeq for identifying DE isoforms. EBSeq also proves to be a robust approach for identifying DE genes. AVAILABILITY AND IMPLEMENTATION: An R package containing examples and sample datasets is available at http://www.biostat.wisc.edu/kendzior/EBSEQ/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Perfilação da Expressão Gênica/métodos , Isoformas de RNA/metabolismo , Análise de Sequência de RNA/métodos , Teorema de Bayes , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Genoma , Modelos Estatísticos , RNA Mensageiro/metabolismo , Software
10.
Biotechniques ; 53(1): 57-60, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22780320

RESUMO

Here we introduce a modified antibody staining method that uses up to 80% less antibody for flow cytometry. We demonstrate this method for the detection of antigens expressed at high, moderate, or low levels in mouse and rat lymphocytes as well as mouse mammary epithelial cells. We obtained reproducibly accurate results for the detection of up to seven parameters for activation induced-proliferation, cell cycle analysis, and phenotyping of cell-surface and intracellular antigens.


Assuntos
Antígenos CD/análise , Células Epiteliais/imunologia , Citometria de Fluxo/métodos , Linfócitos/imunologia , Animais , Antígenos CD/imunologia , Células Cultivadas , Células Epiteliais/citologia , Feminino , Citometria de Fluxo/economia , Linfócitos/citologia , Glândulas Mamárias Animais/citologia , Camundongos , Ratos , Tamanho da Amostra , Coloração e Rotulagem/economia , Coloração e Rotulagem/métodos
11.
Nucleic Acids Res ; 40(1): 132-47, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21914726

RESUMO

Many low-penetrance breast cancer susceptibility loci are found to be located in non-protein-coding regions, suggesting their involvement in gene expression regulation. We identified the human/rat-conserved breast cancer susceptibility locus MCS5A/Mcs5a. This locus has been shown to act in a non-mammary cell-autonomous fashion through the immune system. The resistant Mcs5a allele from the Wistar-Kyoto (WKy) rat strain consists of two non-protein-coding genetic elements that must be located on the same chromosome to elicit the phenotype. In this study, we show the presence of a conserved higher order chromatin structure in MCS5A/Mcs5a located in between the synthetically interacting genetic elements. The looped elements are shown to be bound by CTCF and cohesin. We identify the downregulation of Fbxo10 expression in T cells as a strong candidate mechanism through which the interacting genetic elements of the resistant Mcs5a allele modulate mammary carcinoma susceptibility. Finally, we show that the human MCS5A polymorphisms associated with breast cancer risk are located at both sides of the looped structure and functionally interact to downregulate transcriptional activity, similar to rat Mcs5a. We propose a mechanistic model for MCS5a/Mcs5a in which a CTCF-mediated insulator loop encompassing the TOMM5/Tomm5 gene, resides in between and brings into closer physical proximity the synthetically and functionally interacting resistant genetic variants.


Assuntos
Neoplasias da Mama/genética , Regulação para Baixo , Loci Gênicos , Predisposição Genética para Doença , Elementos Isolantes , Animais , Sequência de Bases , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular/metabolismo , Cromatina/química , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas F-Box/genética , Feminino , Regulação da Expressão Gênica , Humanos , Células Jurkat , Neoplasias Mamárias Experimentais/genética , Dados de Sequência Molecular , Polimorfismo Genético , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos WKY , Proteínas Repressoras/metabolismo , Linfócitos T/metabolismo , Sítio de Iniciação de Transcrição , Ubiquitina-Proteína Ligases/genética , Coesinas
12.
PLoS One ; 6(10): e26145, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22022542

RESUMO

Rat mammary carcinogenesis models have been used extensively to study breast cancer initiation, progression, prevention, and intervention. Nevertheless, quantitative molecular data on epithelial cell differentiation in mammary glands of untreated and carcinogen-exposed rats is limited. Here, we describe the characterization of rat mammary epithelial cells (RMECs) by multicolor flow cytometry using antibodies against cell surface proteins CD24, CD29, CD31, CD45, CD49f, CD61, Peanut Lectin, and Thy-1, intracellular proteins CK14, CK19, and FAK, along with phalloidin and Hoechst staining. We identified the luminal and basal/myoepithelial populations and actively dividing RMECs. In inbred rats susceptible to mammary carcinoma development, we quantified the changes in differentiation of the RMEC populations at 1, 2, and 4 weeks after exposure to mammary carcinogens DMBA and MNU. DMBA exposure did not alter the percentage of basal or luminal cells, but upregulated CD49f (Integrin α6) expression and increased cell cycle activity. MNU exposure resulted in a temporary disruption of the luminal/basal ratio and no CD49f upregulation. When comparing DMBA- or MNU-induced mammary carcinomas, the RMEC differentiation profiles are indistinguishable. The carcinomas compared with mammary glands from untreated rats, showed upregulation of CD29 (Integrin ß1) and CD49f expression, increased FAK (focal adhesion kinase) activation especially in the CD29hi population, and decreased CD61 (Integrin ß3) expression. This study provides quantitative insight into the protein expression phenotypes underlying RMEC differentiation. The results highlight distinct RMEC differentiation etiologies of DMBA and MNU exposure, while the resulting carcinomas have similar RMEC differentiation profiles. The methodology and data will enhance rat mammary carcinogenesis models in the study of the role of epithelial cell differentiation in breast cancer.


Assuntos
Carcinoma/patologia , Diferenciação Celular , Células Epiteliais/patologia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/patologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Carcinoma/metabolismo , Divisão Celular , Membrana Celular/metabolismo , Proliferação de Células , Células Epiteliais/metabolismo , Feminino , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Metilnitrosoureia , Modelos Biológicos , Ratos , Regulação para Cima
13.
Breast Cancer Res ; 13(4): R81, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21846333

RESUMO

INTRODUCTION: Mechanisms underlying low-penetrance, common, non-protein coding variants in breast cancer risk loci are largely undefined. We showed previously that the non-protein coding mammary carcinoma susceptibility locus Mcs5a/MCS5A modulates breast cancer risk in rats and women. The Mcs5a allele from the Wistar-Kyoto (WKy) rat strain consists of two genetically interacting elements that have to be present on the same chromosome to confer mammary carcinoma resistance. We also found that the two interacting elements of the resistant allele are required for the downregulation of transcript levels of the Fbxo10 gene specifically in T-cells. Here we describe mechanisms through which Mcs5a may reduce mammary carcinoma susceptibility. METHODS: We performed mammary carcinoma multiplicity studies with three mammary carcinoma-inducing treatments, namely 7,12-dimethylbenz(a)anthracene (DMBA) and N-nitroso-N-methylurea (NMU) carcinogenesis, and mammary ductal infusion of retrovirus expressing the activated HER2/neu oncogene. We used mammary gland and bone marrow transplantation assays to assess the target tissue of Mcs5a activity. We used immunophenotyping assays on well-defined congenic rat lines carrying susceptible and resistant Mcs5a alleles to identify changes in T-cell homeostasis and function associated with resistance. RESULTS: We show that Mcs5a acts beyond the initial step of mammary epithelial cell transformation, during early cancer progression. We show that Mcs5a controls susceptibility in a non-mammary cell-autonomous manner through the immune system. The resistant Mcs5a allele was found to be associated with an overabundance of gd T-cell receptor (TCR)+ T-cells as well as a CD62L (L-selectin)-high population of all T-cell classes. In contrast to in mammary carcinoma, gdTCR+ T-cells are the predominant T-cell type in the mammary gland and were found to be overabundant in the mammary epithelium of Mcs5a resistant congenic rats. Most of them simultaneously expressed the CD4, CD8, and CD161α markers. In cultured T-cells of Mcs5a resistant congenic rats we found increased mitogen-induced proliferation and production of Th1 cytokines IFNg, IL-2, and Tumor Necrosis Factor (TNF), but not Th2 cytokines IL-4 and IL-6, or Th17 cytokine IL-17 when compared with susceptible control rats. CONCLUSIONS: These data support a hypothesis that Mcs5a displays a non-mammary cell-autonomous mechanism of action to modulate breast cancer risk through the immune system. The resistant Mcs5a allele is associated with alterations in T-cell homeostasis and functions, and overabundance of γδTCR+ T-cells in carcinogen-exposed mammary epithelium.


Assuntos
Neoplasias da Mama/genética , Loci Gênicos , Predisposição Genética para Doença , Neoplasias Mamárias Experimentais/genética , Linfócitos T/imunologia , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Alelos , Animais , Neoplasias da Mama/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos CD8/metabolismo , Citocinas/metabolismo , Células Epiteliais/metabolismo , Feminino , Homeostase , Selectina L/metabolismo , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Metilnitrosoureia/efeitos adversos , Subfamília B de Receptores Semelhantes a Lectina de Células NK/metabolismo , Ratos , Ratos Endogâmicos WKY , Receptores de Antígenos de Linfócitos T/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
14.
PLoS One ; 6(5): e19891, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21625632

RESUMO

Only a portion of the estimated heritability of breast cancer susceptibility has been explained by individual loci. Comparative genetic approaches that first use an experimental organism to map susceptibility QTLs are unbiased methods to identify human orthologs to target in human population-based genetic association studies. Here, overlapping rat mammary carcinoma susceptibility (Mcs) predicted QTLs, Mcs6 and Mcs2, were physically confirmed and mapped to identify the human orthologous region. To physically confirm Mcs6 and Mcs2, congenic lines were established using the Wistar-Furth (WF) rat strain, which is susceptible to developing mammary carcinomas, as the recipient (genetic background) and either Wistar-Kyoto (WKy, Mcs6) or Copenhagen (COP, Mcs2), which are resistant, as donor strains. By comparing Mcs phenotypes of WF.WKy congenic lines with distinct segments of WKy chromosome 7 we physically confirmed and mapped Mcs6 to ~33 Mb between markers D7Rat171 and gUwm64-3. The predicted Mcs2 QTL was also physically confirmed using segments of COP chromosome 7 introgressed into a susceptible WF background. The Mcs6 and Mcs2 overlapping genomic regions contain multiple annotated genes, but none have a clear or well established link to breast cancer susceptibility. Igf1 and Socs2 are two of multiple potential candidate genes in Mcs6. The human genomic region orthologous to rat Mcs6 is on chromosome 12 from base positions 71,270,266 to 105,502,699. This region has not shown a genome-wide significant association to breast cancer risk in pun studies of breast cancer susceptibility.


Assuntos
Mapeamento Cromossômico , Suscetibilidade a Doenças , Fator de Crescimento Insulin-Like I/genética , Neoplasias Mamárias Experimentais/etiologia , Locos de Características Quantitativas , Proteínas Supressoras da Sinalização de Citocina/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Animais Congênicos , Carcinógenos/toxicidade , Feminino , Masculino , Neoplasias Mamárias Experimentais/patologia , Anotação de Sequência Molecular , Ratos , Ratos Endogâmicos WF , Ratos Endogâmicos WKY
15.
Cancer Prev Res (Phila) ; 4(1): 97-106, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21205740

RESUMO

Genetic factors have been estimated to account for at least 30% of a woman's risk to develop breast cancer. We have developed a rat model using Wistar Furth (WF) and Wistar Kyoto (WKy) strains to genetically identify mammary cancer susceptibility loci. The WKy allele of the mammary carcinogenesis susceptibility locus Mcs5c, was previously shown to reduce carcinoma multiplicity after 7,12-dimethylbenz-[a]anthracene (DMBA) exposure. In this study, Mcs5c was fine-mapped using WF.WKy congenic lines. Mcs5c was located to a region of approximately 176 kb on rat chromosome 5. One of the Mcs5c congenic lines containing a narrow Mcs5c WKy interval displayed a 40% decrease in average carcinoma number compared with WF-homozygous congenic controls after mammary carcinogenesis induction using two different models. As genetically mapped, the Mcs5c locus is located in a gene desert and thus is devoid of genes and annotated RNAs; thus, a genetic element in Mcs5c was hypothesized to regulate the expression of genes outside the locus. Tenascin c (Tnc) was identified as a candidate gene due to its reduced expression in thymus and ovarian tissues of Mcs5c WKy-homozygous congenic females compared with WF-homozygous congenic controls. This allele-specific differential expression is environmentally controlled.


Assuntos
Predisposição Genética para Doença/genética , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Locos de Características Quantitativas , Tenascina/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Animais Congênicos , Western Blotting , Carcinógenos/toxicidade , Hibridização Genômica Comparativa , Suscetibilidade a Doenças , Feminino , Neoplasias Mamárias Experimentais/induzido quimicamente , Fenótipo , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos WF , Ratos Endogâmicos WKY , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Cancer Prev Res (Phila) ; 2(2): 153-60, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19196722

RESUMO

Preclinical models that accurately reproduce specific aspects of human disease etiology are invaluable for the initial development and evaluation of chemopreventive agents. We developed a novel, short-term prevention model, which is particularly useful for assessing the efficacy of a compound to prevent hormonally responsive and nonresponsive in situ carcinomas. In this model, carcinogenesis is induced by a high titer of neu-containing, replication-defective retrovirus. The multiplicity and size of the resulting in situ carcinomas are scored in whole-mounted, aluminum carmine-stained mammary glands at 15 days postinfusion. These in situ carcinomas represent a distinct biological time point in the development of neu-induced mammary cancer in the rat. They are characterized by high rates of proliferation (40.0%; P < 0.0001) and apoptosis (2.8%; P < 0.005) compared with mammary carcinomas. The majority of in situ carcinomas regress spontaneously after 20 days postinfusion. The in situ carcinomas at 15 days postinfusion exhibit hormonal responsiveness. The effects of the chemoprevention agents tamoxifen, celecoxib, and targretin on hormonally responsive and nonresponsive in situ carcinomas recapitulate those observed on mammary carcinomas at 12 and 18 weeks postinfusion for intact and ovariectomized rats, respectively. Neu-induced in situ carcinomas in the rat represent etiologically relevant intermediate time points of mammary carcinogenesis. Our prevention model represents a cost-efficient in vivo system to determine whether the preventive effects of a compound extend to hormonally nonresponsive mammary lesions, for which new chemoprevention approaches are needed.


Assuntos
Carcinoma in Situ/prevenção & controle , Modelos Animais de Doenças , Neoplasias Mamárias Experimentais/prevenção & controle , Neoplasias Hormônio-Dependentes/prevenção & controle , Animais , Anticarcinógenos/farmacologia , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Bexaroteno , Carcinoma in Situ/genética , Carcinoma in Situ/patologia , Celecoxib , Proliferação de Células/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Feminino , Genes erbB-2 , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/patologia , Pirazóis/farmacologia , Ratos , Ratos Endogâmicos WF , Retroviridae/genética , Sulfonamidas/farmacologia , Tamoxifeno/farmacologia , Tetra-Hidronaftalenos/farmacologia , Fatores de Tempo
17.
BMC Cancer ; 9: 19, 2009 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-19144184

RESUMO

BACKGROUND: Inflammation has been linked to the etiology of many organ-specific cancers. Indirect evidence suggests a possible role for inflammation in breast cancer. We investigated whether the systemic inflammation induced by Freund's adjuvant (FA) promotes mammary carcinogenesis in a rat model in which cancer is induced by the neu oncogene. METHODS: The effects of FA on hyperplastic mammary lesions and mammary carcinomas were determined in a neu-induced rat model. The inflammatory response to FA treatment was gauged by measuring acute phase serum haptoglobin. In addition, changes in cell proliferation and apoptosis following FA treatment were assessed. RESULTS: Rats receiving FA developed twice the number of mammary carcinomas as controls. Systemic inflammation following FA treatment is chronic, as shown by a doubling of the levels of the serum biomarker, haptoglobin, 15 days following initial treatment. We also show that this systemic inflammation is associated with the increased growth of hyperplastic mammary lesions. This increased growth results from a higher rate of cellular proliferation in the absence of changes in apoptosis. CONCLUSION: Our data suggests that systemic inflammation induced by Freund's adjuvant (FA) promotes mammary carcinogenesis. It will be important to determine whether adjuvants currently used in human vaccines also promote breast cancer.


Assuntos
Adjuvantes Imunológicos/efeitos adversos , Adjuvante de Freund/efeitos adversos , Genes erbB-2/efeitos dos fármacos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Sepse/induzido quimicamente , Sepse/complicações , Animais , Proliferação de Células/efeitos dos fármacos , Feminino , Ratos , Ratos Wistar
18.
Breast Cancer Res ; 10(1): R18, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18279516

RESUMO

INTRODUCTION: While current breast cancer chemoprevention strategies using selective estrogen response modulators and aromatase inhibitors are quite successful, their effects are limited to hormonally responsive breast cancer. Hormonally nonresponsive breast cancer (including estrogen receptor-negative cancer) is associated with poor prognosis for patients, and few chemoprevention agents exist for this type of cancer. The cyclooxygenase-2 inhibitor celecoxib (Celebrex) is a nonsteroidal anti-inflammatory drug and as such is a potential candidate for the prevention of hormonally nonresponsive breast cancer. METHODS: The chemopreventive effects of celecoxib were evaluated in the neu-induced retroviral rat mammary carcinogenesis model, to assess the efficacy of celecoxib on hormonally responsive and hormonally nonresponsive mammary carcinomas. RESULTS: Dietary celecoxib at 1,200 mg/kg diet was highly efficacious in the prevention of hormonally responsive mammary carcinomas in intact rats, decreasing tumor multiplicity by 56% (P < 0.0001) and by 74% (P = 0.0002) in two independent experiments. No significant effect was found, however, on hormonally nonresponsive mammary carcinomas of ovariectomized rats. Treatment with a combination diet, consisting of tamoxifen at 2 mg/kg diet and celecoxib at 1,200 mg/kg diet, reduced tumor multiplicity by 72% (P = 0.0002) in intact rats. This reduction was not statistically different from that observed with celecoxib alone. Furthermore, long-term treatment with celecoxib was not associated with reductions in tumor volume in either intact rats or ovariectomized rats. In contrast, tamoxifen treatment and the combination regimen caused significant reductions in tumor volumes in intact rats (P = 0.01 and P = 0.004, respectively). Consistent with these data, decreases in proliferation and increases in apoptosis were detected in tamoxifen-treated and combination diet-treated tumors. No such modulations were observed in celecoxib-treated tumors. CONCLUSION: The chemopreventive effects of celecoxib appear to be limited to modulations in multiplicity of hormonally responsive mammary carcinomas. The fact that no synergistic or additive effects were observed in combination diet-treated rats raises the question of whether celecoxib is suitable for the prevention of hormonally nonresponsive breast cancer or for use in combination therapy with selective estrogen response modulators or aromatase inhibitors.


Assuntos
Anticarcinógenos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Mama/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Tamoxifeno/farmacologia , Animais , Celecoxib , Modelos Animais de Doenças , Feminino , Ratos , Ratos Endogâmicos WF
19.
Breast Dis ; 28: 53-61, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18057543

RESUMO

Rat mammary cancers generally resemble many features of human breast cancer. With the recent developments in rat genetic engineering, the rat has become an excellent model system to study aspects of the molecular etiology of breast cancer. In this review, we describe the efforts to generate genetically engineered rat models for breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias Mamárias Experimentais/genética , Animais , Animais Geneticamente Modificados , Neoplasias da Mama/metabolismo , Feminino , Genes BRCA1 , Genes BRCA2 , Genes ras , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Ratos , Receptor ErbB-2/metabolismo , Transfecção
20.
Proc Natl Acad Sci U S A ; 104(15): 6299-304, 2007 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-17404222

RESUMO

Breast cancer risk is a polygenic trait. To identify breast cancer modifier alleles that have a high population frequency and low penetrance we used a comparative genomics approach. Quantitative trait loci (QTL) were initially identified by linkage analysis in a rat mammary carcinogenesis model followed by verification in congenic rats carrying the specific QTL allele under study. The Mcs5a locus was identified by fine-mapping Mcs5 in a congenic model. Here we characterize the Mcs5a locus, which when homozygous for the Wky allele, reduces mammary cancer risk by 50%. The Mcs5a locus is a compound QTL with at least two noncoding interacting elements: Mcs5a1 and Mcs5a2. The resistance phenotype is only observed in rats carrying at least one copy of the Wky allele of each element on the same chromosome. Mcs5a1 is located within the ubiquitin ligase Fbxo10, whereas Mcs5a2 includes the 5' portion of Frmpd1. Resistant congenic rats show a down-regulation of Fbxo10 in the thymus and an up-regulation of Frmpd1 in the spleen. The association of the Mcs5a1 and Mcs5a2 human orthologs with breast cancer was tested in two population-based breast cancer case-control studies (approximately 12,000 women). The minor alleles of rs6476643 (MCS5A1) and rs2182317 (MCS5A2) were independently associated with breast cancer risk. The minor allele of rs6476643 increases risk, whereas the rs2182317 minor allele decreases risk. Both alleles have a high population frequency and a low penetrance toward breast cancer risk.


Assuntos
Neoplasias da Mama/genética , Cromossomos Humanos Par 9/genética , Predisposição Genética para Doença , Locos de Características Quantitativas , Animais , Sequência de Bases , Mapeamento Cromossômico , Biologia Computacional , Cruzamentos Genéticos , Feminino , Frequência do Gene , Humanos , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Ratos , Ratos Endogâmicos WF , Ratos Endogâmicos WKY , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Reino Unido , Regiões não Traduzidas/genética , Wisconsin
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...