1
|
Linscott MP, Ren JR, Gestl SA, Gunther EJ. Different Oncogenes and Reproductive Histories Shape the Progression of Distinct Premalignant Clones in Multistage Mouse Breast Cancer Models. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1329-1345. [PMID: 38537934 PMCID: PMC11220927 DOI: 10.1016/j.ajpath.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/06/2024] [Accepted: 02/16/2024] [Indexed: 04/10/2024]
Abstract
A remote carcinogen exposure can predispose to breast cancer onset decades later, suggesting that carcinogen-induced mutations generate long-lived premalignant clones. How subsequent events influence the progression of specific premalignant clones remains poorly understood. Herein, multistage mouse models of mammary carcinogenesis were generated by combining chemical carcinogen exposure [using 7,12-dimethylbenzanthracene (DMBA)] with transgenes that enable inducible expression of one of two clinically relevant mammary oncogenes: c-MYC (MYC) or PIK3CAH1047R (PIK). In prior work, DMBA exposure generated mammary clones bearing signature HrasQ61L mutations, which only progressed to mammary cancer after inducible Wnt1 oncogene expression. Here, after an identical DMBA exposure, MYC versus PIK drove cancer progression from mammary clones bearing mutations in distinct Ras family paralogs. For example, MYC drove cancer progression from either Kras- or Nras-mutant clones, whereas PIK transformed Kras-mutant clones only. These Ras mutation patterns were maintained whether oncogenic transgenes were induced within days of DMBA exposure or months later. Completing a full-term pregnancy (parity) failed to protect against either MYC- or PIK-driven tumor progression. Instead, a postpartum increase in mammary tumor predisposition was observed in the context of PIK-driven progression. However, parity decreased the overall prevalence of tumors bearing Krasmut, and the magnitude of this decrease depended on both the number and timing of pregnancies. These multistage models may be useful for elucidating biological features of premalignant mammary neoplasia.
Collapse
Affiliation(s)
- Maryknoll P Linscott
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jerry R Ren
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Shelley A Gestl
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Edward J Gunther
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
| |
Collapse
|
2
|
Nicotra R, Lutz C, Messal HA, Jonkers J. Rat Models of Hormone Receptor-Positive Breast Cancer. J Mammary Gland Biol Neoplasia 2024; 29:12. [PMID: 38913216 PMCID: PMC11196369 DOI: 10.1007/s10911-024-09566-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024] Open
Abstract
Hormone receptor-positive (HR+) breast cancer (BC) is the most common type of breast cancer among women worldwide, accounting for 70-80% of all invasive cases. Patients with HR+ BC are commonly treated with endocrine therapy, but intrinsic or acquired resistance is a frequent problem, making HR+ BC a focal point of intense research. Despite this, the malignancy still lacks adequate in vitro and in vivo models for the study of its initiation and progression as well as response and resistance to endocrine therapy. No mouse models that fully mimic the human disease are available, however rat mammary tumor models pose a promising alternative to overcome this limitation. Compared to mice, rats are more similar to humans in terms of mammary gland architecture, ductal origin of neoplastic lesions and hormone dependency status. Moreover, rats can develop spontaneous or induced mammary tumors that resemble human HR+ BC. To date, six different types of rat models of HR+ BC have been established. These include the spontaneous, carcinogen-induced, transplantation, hormone-induced, radiation-induced and genetically engineered rat mammary tumor models. Each model has distinct advantages, disadvantages and utility for studying HR+ BC. This review provides a comprehensive overview of all published models to date.
Collapse
Affiliation(s)
- Raquel Nicotra
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Amsterdam, Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| | - Hendrik A Messal
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| |
Collapse
|
3
|
Yang Y, Leonard M, Luo Z, Yeo S, Bick G, Hao M, Cai C, Charif M, Wang J, Guan JL, Lower EE, Zhang X. Functional cooperation between co-amplified genes promotes aggressive phenotypes of HER2-positive breast cancer. Cell Rep 2021; 34:108822. [PMID: 33691110 PMCID: PMC8050805 DOI: 10.1016/j.celrep.2021.108822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 12/21/2020] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
MED1 (mediator subunit 1)co-amplifies with HER2, but its role in HER2-driven mammary tumorigenesis is still unknown. Here, we generate MED1 mammary-specific overexpression mice and cross them with mouse mammary tumor virus (MMTV)-HER2 mice. We observe significantly promoted onset, growth, metastasis, and multiplicity of HER2 tumors by MED1 overexpression. Further studies reveal critical roles for MED1 in epithelial-mesenchymal transition, cancer stem cell formation, and response to anti-HER2 therapy. Mechanistically, RNA sequencing (RNA-seq) transcriptome analyses and clinical sample correlation studies identify Jab1, a component of the COP9 signalosome complex, as the key direct target gene of MED1 contributing to these processes. Further studies reveal that Jab1 can also reciprocally regulate the stability and transcriptional activity of MED1. Together, our findings support a functional cooperation between these co-amplified genes in HER2+ mammary tumorigenesis and their potential usage as therapeutic targets for the treatment of HER2+ breast cancers. In this study, Yang et al. generate a more clinically relevant MMTV-HER2/MMTV-MED1 mammary tumor mouse model and discover the critical roles and molecular mechanisms of MED1 overexpression in mediating the aggressive phenotypes of HER2+ tumor progression, metastasis, cancer stem cell formation, and therapy resistance.
Collapse
Affiliation(s)
- Yongguang Yang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Marissa Leonard
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Graduate Program in Cancer and Cell Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhenhua Luo
- The Liver Care Center and Divisions of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Syn Yeo
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Gregory Bick
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mingang Hao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Chunmiao Cai
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mahmoud Charif
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Elyse E Lower
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xiaoting Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Graduate Program in Cancer and Cell Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; University of Cincinnati Cancer Center, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
4
|
Bu W, Li Y. Intraductal Injection of Lentivirus Vectors for Stably Introducing Genes into Rat Mammary Epithelial Cells in Vivo. J Mammary Gland Biol Neoplasia 2020; 25:389-396. [PMID: 33165800 PMCID: PMC7965254 DOI: 10.1007/s10911-020-09469-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/18/2020] [Accepted: 11/04/2020] [Indexed: 12/12/2022] Open
Abstract
Various retroviral and lentiviral vectors have been used for up-the-teat intraductal injection to deliver markers, oncogenes, and other genes into mammary epithelial cells in mice. These methods along with the large number of genetically engineered mouse lines have greatly helped us learn normal breast development and tumorigenesis. Rats are also valuable models for studying human breast development and cancer. However, genetically engineered rats are still uncommon, and previous reports of intraductal injection of retroviral vectors into rats appear to be inefficient in generating mammary tumors. Here, we report, and describe the method for, stably introducing marker genes and oncogenes into mammary glands in rats using intraductal injection of commonly used lentiviral vectors. This method can infect mammary epithelial cells efficiently, and the infected cells can initiate tumorigenesis, including estrogen receptor-positive and hormone-dependent tumors, which are the most common subtype of human breast cancer but are yet still difficult to model in mice. This technique provides another tool for studying formation, prevention, and treatment of breast cancer, especially estrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Wen Bu
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yi Li
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Cytoplasmic ERα and NFκB Promote Cell Survival in Mouse Mammary Cancer Cell Lines. Discov Oncol 2020; 11:76-86. [PMID: 32008217 DOI: 10.1007/s12672-020-00378-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/16/2020] [Indexed: 12/15/2022] Open
Abstract
There is a desperate need in the field for mouse mammary tumors and cell lines that faithfully mimic estrogen receptor (ER) expression and activity found in human breast cancers. We found that several mouse mammary cancer cell lines express ER but fail to demonstrate classical estrogen-driven proliferation or transcriptional activity. We investigated whether these cell lines may be used to model tamoxifen resistance by using small molecule inhibitors to signaling pathways known to contribute to resistance. We found that the combination of NFκB inhibition and ER antagonists significantly reduced cell proliferation in vitro, as well as growth of syngeneic tumors. Surprisingly, we found that ER was localized to the cytoplasm, regardless of any type of treatment. Based on this, we probed extra-nuclear functions of ER and found that co-inhibition of ER and NFκB led to an increase in oxidative stress and apoptosis. Together, these findings suggest that cytoplasmic ER and NFκB may play redundant roles in protecting mammary cancer cells from oxidative stress and cell death. Although this study has not identified a mouse model with classical ER activity, cytoplasmic ER has been described in a small subset of human breast tumors, suggesting that these findings may be relevant for some breast cancer patients.
Collapse
|
6
|
CDK2-mediated site-specific phosphorylation of EZH2 drives and maintains triple-negative breast cancer. Nat Commun 2019; 10:5114. [PMID: 31704972 PMCID: PMC6841924 DOI: 10.1038/s41467-019-13105-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/15/2019] [Indexed: 12/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC), which lacks estrogen receptor α (ERα), progesterone receptor, and human epidermal growth factor receptor 2 (HER2) expression, is closely related to basal-like breast cancer. Previously, we and others report that cyclin E/cyclin-dependent kinase 2 (CDK2) phosphorylates enhancer of zeste homolog 2 (EZH2) at T416 (pT416-EZH2). Here, we show that transgenic expression of phospho-mimicking EZH2 mutant EZH2T416D in mammary glands leads to tumors with TNBC phenotype. Coexpression of EZH2T416D in mammary epithelia of HER2/Neu transgenic mice reprograms HER2-driven luminal tumors into basal-like tumors. Pharmacological inhibition of CDK2 or EZH2 allows re-expression of ERα and converts TNBC to luminal ERα-positive, rendering TNBC cells targetable by tamoxifen. Furthermore, the combination of either CDK2 or EZH2 inhibitor with tamoxifen effectively suppresses tumor growth and markedly improves the survival of the mice bearing TNBC tumors, suggesting that the mechanism-based combination therapy may be an alternative approach to treat TNBC. EZH2 phosphorylation by CDK2 promotes progression of triple-negative breast cancer (TNBC). Here, the authors show that this signaling axis downregulates ERα, and thus combinatorial blockade of CDK2 and EZH2 sensitizes TNBC cells to tamoxifen.
Collapse
|
7
|
Li S, Gestl SA, Gunther EJ. A Multistage Murine Breast Cancer Model Reveals Long-Lived Premalignant Clones Refractory to Parity-Induced Protection. Cancer Prev Res (Phila) 2019; 13:173-184. [PMID: 31699706 DOI: 10.1158/1940-6207.capr-19-0322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/23/2019] [Accepted: 10/29/2019] [Indexed: 11/16/2022]
Abstract
Breast cancers evolve in a multistage process that can span decades after a carcinogenic exposure. It follows that long-lived precursor breast lesions persist in a subclinical state prior to completing malignant transformation, yet widely used breast cancer models lack an experimental framework for targeting premalignant disease. Inspired by classic multistage skin carcinogenesis protocols, we combined chemical carcinogenesis with transgenic mouse modeling to resolve mouse mammary carcinogenesis into discrete initiation and progression stages. At the initiation stage, exposure to the carcinogen 7,12-dimethylbenzanthracene (DMBA) generated "initiated mammary epithelial cells" (iMEC) by introducing a stereotyped HrasQ61L driver mutation. Whether DMBA exposure occurred during puberty or adulthood, mice efficiently acquired iMEC clones that eluded detection by conventional histology, yet were long lived, persisting in a clinically silent state for months in the absence of a cooperating event. At the progression stage, inducible activation of oncogenic Wnt signaling drove rapid and synchronous transformation of latent iMECs into overt mammary carcinomas, while Wnt activation in neighboring normal mammary epithelium yielded only benign hyperplasia over this same time period. Although early parity (completion of a full-term pregnancy) reduces breast cancer risk in some contexts, standard parity-induced protection schemes failed to eliminate iMECs in our multistage model, suggesting Wnt-responsive iMECs are maintained by hormone-independent mechanisms. Variations on our multistage modeling strategy may help to identify and validate cellular and molecular targets for breast cancer chemoprevention.
Collapse
Affiliation(s)
- Shuo Li
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania.,Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Shelley A Gestl
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania.,Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Edward J Gunther
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania. .,Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania.,Department of Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
8
|
Kreuzaler P, Clarke MA, Brown EJ, Wilson CH, Kortlever RM, Piterman N, Littlewood T, Evan GI, Fisher J. Heterogeneity of Myc expression in breast cancer exposes pharmacological vulnerabilities revealed through executable mechanistic modeling. Proc Natl Acad Sci U S A 2019; 116:22399-22408. [PMID: 31611367 PMCID: PMC6825310 DOI: 10.1073/pnas.1903485116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cells with higher levels of Myc proliferate more rapidly and supercompetitively eliminate neighboring cells. Nonetheless, tumor cells in aggressive breast cancers typically exhibit significant and stable heterogeneity in their Myc levels, which correlates with refractoriness to therapy and poor prognosis. This suggests that Myc heterogeneity confers some selective advantage on breast tumor growth and progression. To investigate this, we created a traceable MMTV-Wnt1-driven in vivo chimeric mammary tumor model comprising an admixture of low-Myc- and reversibly switchable high-Myc-expressing clones. We show that such tumors exhibit interclonal mutualism wherein cells with high-Myc expression facilitate tumor growth by promoting protumorigenic stroma yet concomitantly suppress Wnt expression, which renders them dependent for survival on paracrine Wnt provided by low-Myc-expressing clones. To identify any therapeutic vulnerabilities arising from such interdependency, we modeled Myc/Ras/p53/Wnt signaling cross talk as an executable network for low-Myc, for high-Myc clones, and for the 2 together. This executable mechanistic model replicated the observed interdependence of high-Myc and low-Myc clones and predicted a pharmacological vulnerability to coinhibition of COX2 and MEK. This was confirmed experimentally. Our study illustrates the power of executable models in elucidating mechanisms driving tumor heterogeneity and offers an innovative strategy for identifying combination therapies tailored to the oligoclonal landscape of heterogenous tumors.
Collapse
Affiliation(s)
- Peter Kreuzaler
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
- Oncogenes and Tumour Metabolism Lab, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Matthew A Clarke
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Elizabeth J Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Catherine H Wilson
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Roderik M Kortlever
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Nir Piterman
- Department of Computer Science and Engineering, University of Gothenburg, SE-41296 Gothenburg, Sweden
| | - Trevor Littlewood
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Gerard I Evan
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom;
| | - Jasmin Fisher
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom;
- UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| |
Collapse
|
9
|
Chou CK, Huang HW, Yang CF, Dahms HU, Liang SS, Wang TN, Kuo PL, Hsi E, Tsai EM, Chiu CC. Reduced camptothecin sensitivity of estrogen receptor-positive human breast cancer cells following exposure to di(2-ethylhexyl)phthalate (DEHP) is associated with DNA methylation changes. ENVIRONMENTAL TOXICOLOGY 2019; 34:401-414. [PMID: 30720231 DOI: 10.1002/tox.22694] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 06/09/2023]
Abstract
Di(2-ethylhexyl)phthalate (DEHP) has been considered as an estrogen receptor alpha (ERα) agonist due to its ability to interact with ERα and promote the cell proliferation of ERα-positive breast cancer cells. The impact of DEHP on the chemical therapy in breast cancer is little known. Two breast cancer cell lines, MCF-7 (ERα-dependent) and MDA-MB-231 (ERα-independent) were examined. We found that DEHP impaired the effectiveness of camptothecin (CPT) and alleviated the CPT-induced formation of reactive oxygen species in ERα-positive MCF-7 cells, but not in ERα-negative MDA-MB-231 cells. DEHP also significantly protected MCF-7 cells against the genotoxicity of CPT. Genome-wide DNA methylation profiling revealed that after 48 hours of exposure to 100 μM DEHP, MCF-7 cells exhibited a significant change in their DNA methylation pattern, including hypermethylation of 700 genes and hypomethylation of 221 genes. The impaired therapeutic response to CPT in DEHP-exposed MCF-7 cells is probably mediated by epigenetic changes, especially through Wnt/β-catenin signaling. A zebrafish xenograft model confirmed the disruptive effect of DEHP on CPT-induced anti-growth of MCF-7 cells. In summary, DEHP exposure induces acquired CPT-resistance in breast cancer cells and epigenetic changes associated with Wnt/β-catenin signaling activation are probably depending on an ER-positive status.
Collapse
Affiliation(s)
- Chon-Kit Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hurng-Wern Huang
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chun-Feng Yang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hans-Uwe Dahms
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Shih-Shin Liang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsu-Nai Wang
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Lin Kuo
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Edward Hsi
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Translational Research Center, Cancer Center and Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
10
|
El Ayachi I, Fatima I, Wend P, Alva-Ornelas JA, Runke S, Kuenzinger WL, Silva J, Silva W, Gray JK, Lehr S, Barch HC, Krutilina RI, White AC, Cardiff R, Yee LD, Yang L, O'Regan RM, Lowry WE, Seagroves TN, Seewaldt V, Krum SA, Miranda-Carboni GA. The WNT10B Network Is Associated with Survival and Metastases in Chemoresistant Triple-Negative Breast Cancer. Cancer Res 2019; 79:982-993. [PMID: 30563890 PMCID: PMC6855600 DOI: 10.1158/0008-5472.can-18-1069] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 09/11/2018] [Accepted: 12/14/2018] [Indexed: 01/05/2023]
Abstract
Triple-negative breast cancer (TNBC) commonly develops resistance to chemotherapy, yet markers predictive of chemoresistance in this disease are lacking. Here, we define WNT10B-dependent biomarkers for β-CATENIN/HMGA2/EZH2 signaling predictive of reduced relapse-free survival. Concordant expression of HMGA2 and EZH2 proteins is observed in MMTV-Wnt10bLacZ transgenic mice during metastasis, and Hmga2 haploinsufficiency decreased EZH2 protein expression, repressing lung metastasis. A novel autoregulatory loop interdependent on HMGA2 and EZH2 expression is essential for β-CATENIN/TCF-4/LEF-1 transcription. Mechanistically, both HMGA2 and EZH2 displaced Groucho/TLE1 from TCF-4 and served as gatekeepers for K49 acetylation on β-CATENIN, which is essential for transcription. In addition, we discovered that HMGA2-EZH2 interacts with the PRC2 complex. Absence of HMGA2 or EZH2 expression or chemical inhibition of Wnt signaling in a chemoresistant patient-derived xenograft (PDX) model of TNBC abolished visceral metastasis, repressing AXIN2, MYC, EZH2, and HMGA2 expression in vivo. Combinatorial therapy of a WNT inhibitor with doxorubicin synergistically activated apoptosis in vitro, resensitized PDX-derived cells to doxorubicin, and repressed lung metastasis in vivo. We propose that targeting the WNT10B biomarker network will provide improved outcomes for TNBC. SIGNIFICANCE: These findings reveal targeting the WNT signaling pathway as a potential therapeutic strategy in triple-negative breast cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/5/982/F1.large.jpg.
Collapse
Affiliation(s)
- Ikbale El Ayachi
- Department of Medicine, College of Medicine at UTHSC, UTHSC Center for Cancer Research, Memphis, Tennessee
| | - Iram Fatima
- Department of Medicine, College of Medicine at UTHSC, UTHSC Center for Cancer Research, Memphis, Tennessee
| | - Peter Wend
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Jackelyn A Alva-Ornelas
- Department of Population Science, City of Hope Comprehensive Cancer Center and Beckman Institute, Duarte, California
| | - Stephanie Runke
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - William L Kuenzinger
- Department of Medicine, College of Medicine at UTHSC, UTHSC Center for Cancer Research, Memphis, Tennessee
| | - Julio Silva
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Wendy Silva
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Joseph K Gray
- Department of Medicine, College of Medicine at UTHSC, UTHSC Center for Cancer Research, Memphis, Tennessee
| | | | - Hilaire C Barch
- Department of Pathology and Laboratory Medicine, College of Medicine at UTHSC, UTHSC Center for Cancer Research, Memphis, Tennessee
| | - Raisa I Krutilina
- Department of Pathology and Laboratory Medicine, College of Medicine at UTHSC, UTHSC Center for Cancer Research, Memphis, Tennessee
| | - Andrew C White
- Department of Biomedical Sciences, Cornell University, Ithaca, New York
| | - Robert Cardiff
- Department of Medical Pathology, School of Medicine, University of California, Davis, California
| | - Lisa D Yee
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Lily Yang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Ruth M O'Regan
- Division of Hematology and Oncology, Departments of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - William E Lowry
- Molecular, Cell and Developmental Biology, University of California, Los Angeles, California
| | - Tiffany N Seagroves
- Department of Pathology and Laboratory Medicine, College of Medicine at UTHSC, UTHSC Center for Cancer Research, Memphis, Tennessee
| | - Victoria Seewaldt
- Department of Population Science, City of Hope Comprehensive Cancer Center and Beckman Institute, Duarte, California
| | - Susan A Krum
- Department of Orthopaedic Surgery and Biomedical Engineering, UTHSC Center for Cancer Research, UTHSC, Memphis, Tennessee
| | - Gustavo A Miranda-Carboni
- Department of Medicine, College of Medicine at UTHSC, UTHSC Center for Cancer Research, Memphis, Tennessee.
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Jonsson Comprehensive Cancer Center, Los Angeles, California
| |
Collapse
|
11
|
Wu J, Crowe DL. Molecular and cellular basis of mammary gland fibrosis and cancer risk. Int J Cancer 2018; 144:2239-2253. [PMID: 30450584 DOI: 10.1002/ijc.32000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/16/2018] [Accepted: 11/07/2018] [Indexed: 11/06/2022]
Abstract
Mammary gland luminal cells are maintained by the proliferation of ER- luminal progenitor (LP) cells. Human breast LP cells exhibit telomere DNA damage, which is associated with mammographic density and increased cancer risk. Telomeric repeat factor 2 (TRF2) protects telomeres from DNA damage response. TRF2 expression is reduced in human breast cancers. We deleted TRF2 expression in mammary gland epithelium. Mammary glands lacking TRF2 expression exhibited increased telomere DNA damage response, histopathological and functional degeneration, and prominent ductal fibrosis. TRF2-deficient mammary tumors exhibited rapid onset and increased proliferation. Tumor derived LP cells failed to form tumors after transplantation. The MSC population was highly tumorigenic and maintained telomeres via the ALT mechanism. Telomere DNA damage response in mammary tumors resulted in p53 dependent ER+ cellular differentiation and sensitivity to anti-estrogen therapy. Our results provide a new in vivo model of mammographic density, stem cell differentiation, cancer risk, and therapeutic sensitivity.
Collapse
Affiliation(s)
- Jianchun Wu
- Cancer Biology Program, University of Illinois Cancer Center, Chicago, IL
| | - David L Crowe
- Cancer Biology Program, University of Illinois Cancer Center, Chicago, IL
| |
Collapse
|
12
|
Lamb CA, Fabris VT, Jacobsen B, Molinolo AA, Lanari C. Biological and clinical impact of imbalanced progesterone receptor isoform ratios in breast cancer. Endocr Relat Cancer 2018; 25:ERC-18-0179. [PMID: 29991638 DOI: 10.1530/erc-18-0179] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/17/2022]
Abstract
There is a consensus that progestins and thus their cognate receptor molecules, the progesterone receptors (PR), are essential in the development of the adult mammary gland and regulators of proliferation and lactation. However, a role for natural progestins in breast carcinogenesis remains poorly understood. A hint to that possible role came from studies in which the synthetic progestin medroxyprogesterone acetate was associated with an increased breast cancer risk in women under hormone replacement therapy. However, progestins have been also used for breast cancer treatment and to inhibit the growth of several experimental breast cancer models. More recently, PR have been shown to be regulators of estrogen receptor signaling. With all this information, the question is how can we target PR, and if so, which patients may benefit from such an approach? PR are not single unique molecules. Two main PR isoforms have been characterized, PRA and PRB, that exert different functions and the relative abundance of one isoform respect to the other determines the response of PR agonists and antagonists. Immunohistochemistry with standard antibodies against PR do not discriminate between isoforms. In this review, we summarize the current knowledge on the expression of both PR isoforms in mammary glands, in experimental models of breast cancer and in breast cancer patients, to better understand how the PRA/PRB ratio can be exploited therapeutically to design personalized therapeutic strategies.
Collapse
Affiliation(s)
- Caroline A Lamb
- C Lamb, Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - Victoria T Fabris
- V Fabris, Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - Britta Jacobsen
- B Jacobsen, Department of Pathology, University of Colorado at Denver - Anschutz Medical Campus, Aurora, United States
| | - Alfredo A Molinolo
- A Molinolo, Biorepository and Tissue Technology Shared Resource, University of California San Diego Moores Cancer Center, La Jolla, United States
| | - Claudia Lanari
- C Lanari, Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| |
Collapse
|
13
|
Hyperprolactinemia-inducing antipsychotics increase breast cancer risk by activating JAK-STAT5 in precancerous lesions. Breast Cancer Res 2018; 20:42. [PMID: 29778097 PMCID: PMC5960176 DOI: 10.1186/s13058-018-0969-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/11/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Psychiatric medications are widely prescribed in the USA. Many antipsychotics cause serum hyperprolactinemia as an adverse side effect; prolactin-Janus kinase 2 (JAK2)-signal transducer and activator of transcription 5 (STAT5) signaling both induces cell differentiation and suppresses apoptosis. It is controversial whether these antipsychotics increase breast cancer risk. METHODS We investigated the impact of several antipsychotics on mammary tumorigenesis initiated by retrovirus-mediated delivery of either ErbB2 or HRas or by transgenic expression of Wnt-1. RESULTS We found that the two hyperprolactinemia-inducing antipsychotics, risperidone and pimozide, prompted precancerous lesions to progress to cancer while aripiprazole, which did not cause hyperprolactinemia, did not. We observed that risperidone and pimozide (but not aripiprazole) caused precancerous cells to activate STAT5 and suppress apoptosis while exerting no impact on proliferation. Importantly, we demonstrated that these effects of antipsychotics on early lesions required the STAT5 gene function. Furthermore, we showed that only two-week treatment of mice with ruxolitinib, a JAK1/2 inhibitor, blocked STAT5 activation, restored apoptosis, and prevented early lesion progression. CONCLUSIONS Hyperprolactinemia-inducing antipsychotics instigate precancerous cells to progress to cancer via JAK/STAT5 to suppress the apoptosis anticancer barrier, and these cancer-promoting effects can be prevented by prophylactic anti-JAK/STAT5 treatment. This preclinical work exposes a potential breast cancer risk from hyperprolactinemia-inducing antipsychotics in certain patients and suggests a chemoprevention regime that is relatively easy to implement compared to the standard 5-year anti-estrogenic treatment in women who have or likely have already developed precancerous lesions while also requiring hyperprolactinemia-inducing antipsychotics.
Collapse
|
14
|
Özdemir BC, Sflomos G, Brisken C. The challenges of modeling hormone receptor-positive breast cancer in mice. Endocr Relat Cancer 2018; 25:R319-R330. [PMID: 29563191 DOI: 10.1530/erc-18-0063] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 03/21/2018] [Indexed: 12/26/2022]
Abstract
Estrogen receptor-positive (ER+) tumors account for 70-80% of all breast cancer (BC) cases and are characterized by estrogen dependency for their growth. Endocrine therapies using estrogen receptor antagonists or aromatase inhibitors represent a key component of the standard of care for these tumors. The occurrence of de novo or acquired resistance to estrogen withdrawal represents an important clinical problem, impacting on patient survival. In addition, despite an initially favorable outcome, a part of ER+ BC patients present with disease recurrence locally or at distant sites years or even decades after apparent remission. In vivo models that closely mimic human disease are urgently needed to study the biology of these tumors, investigate the molecular mechanisms underlying endocrine resistance and identify patients at risk of recurrence. Despite the similarities in the overall hormonal regulation of mammary gland development between mice and humans, the majority of the mammary carcinomas occurring in genetically engineered mouse models (GEMMs) are ER negative and most xenograft models are based on few ER+ cancer cell lines. We recently showed that the microenvironment is critical for ER+ cancer cells and discuss in this review the potential of intraductal xenograft model for basic and preclinical research.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- International Cancer Prevention Institute, Epalinges, Switzerland
| | - George Sflomos
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Cathrin Brisken
- International Cancer Prevention Institute, Epalinges, Switzerland
- ISREC - Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
15
|
Montales MTE, Melnyk SB, Liu SJ, Simmen FA, Liu YL, Simmen RCM. Metabolic history impacts mammary tumor epithelial hierarchy and early drug response in mice. Endocr Relat Cancer 2016; 23:677-90. [PMID: 27402613 PMCID: PMC4997088 DOI: 10.1530/erc-16-0136] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/08/2016] [Indexed: 12/22/2022]
Abstract
The emerging links between breast cancer and metabolic dysfunctions brought forth by the obesity pandemic predict a disproportionate early disease onset in successive generations. Moreover, sensitivity to chemotherapeutic agents may be influenced by the patient's metabolic status that affects the disease outcome. Maternal metabolic stress as a determinant of drug response in progeny is not well defined. Here, we evaluated mammary tumor response to doxorubicin in female mouse mammary tumor virus-Wnt1 transgenic offspring exposed to a metabolically compromised environment imposed by maternal high-fat diet. Control progeny were from dams consuming diets with regular fat content. Maternal high-fat diet exposure increased tumor incidence and reduced tumor latency but did not affect tumor volume response to doxorubicin, compared with control diet exposure. However, doxorubicin-treated tumors from high-fat-diet-exposed offspring demonstrated higher proliferation status (Ki-67), mammary stem cell-associated gene expression (Notch1, Aldh1) and basal stem cell-like (CD29(hi)CD24(+)) epithelial subpopulation frequencies, than tumors from control diet progeny. Notably, all epithelial subpopulations (CD29(hi)CD24(+), CD29(lo)CD24(+), CD29(hi)CD24(+)Thy1(+)) in tumors from high-fat-diet-exposed offspring were refractory to doxorubicin. Further, sera from high-fat-diet-exposed offspring promoted sphere formation of mouse mammary tumor epithelial cells and of human MCF7 cells. Untargeted metabolomics analyses identified higher levels of kynurenine and 2-hydroxyglutarate in plasma of high-fat diet than control diet offspring. Kynurenine/doxorubicin co-treatment of MCF7 cells enhanced the ability to form mammosphere and decreased apoptosis, relative to doxorubicin-only-treated cells. Maternal metabolic dysfunctions during pregnancy and lactation may be targeted to reduce breast cancer risk and improve early drug response in progeny, and may inform clinical management of disease.
Collapse
Affiliation(s)
- Maria Theresa E Montales
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Stepan B Melnyk
- Department of PediatricsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA Arkansas Children's Hospital Research InstituteUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Shi J Liu
- Department of Pharmaceutical SciencesUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Frank A Simmen
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA The Winthrop P Rockefeller Cancer InstituteUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Y Lucy Liu
- The Winthrop P Rockefeller Cancer InstituteUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA Department of Internal MedicineUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Rosalia C M Simmen
- Department of Physiology and BiophysicsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA The Winthrop P Rockefeller Cancer InstituteUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
16
|
Abstract
The majority of human breast cancers are estrogen receptor-positive (ER+), but this has proven challenging to model in genetically engineered mice. This review summarizes information on 21 mouse models that develop ER+ mammary cancer. Where available, information on cancer pathology and gene expression profiles is referenced to assist in understanding which histological subtype of ER+ human cancer each model might represent. ESR1, CCDN1, prolactin, TGFα, AIB1, ESPL1, and WNT1 overexpression, PIK3CA gain of function, as well as loss of P53 (Trp53) or STAT1 are associated with ER+ mammary cancer. Treatment with the PPARγ agonist efatutazone in a mouse with Brca1 and p53 deficiency and 7,12-dimethylbenz(a)anthracene exposure in combination with an activated myristoylated form of AKT1 also induce ER+ mammary cancer. A spontaneous mutant in nude mice that develops metastatic ER+ mammary cancer is included. Age of cancer development ranges from 3 to 26 months and the percentage of cancers that are ER+ vary from 21 to 100%. Not all models are characterized as to their estrogen dependency and/or response to anti-hormonal therapy. Strain backgrounds include C57Bl/6, FVB, BALB/c, 129S6/SvEv, CB6F1, and NIH nude. Most models have only been studied on one strain background. In summary, while a range of models are available for studies of pathogenesis and therapy of ER+ breast cancers, many could benefit from further characterization, and opportunity for development of new models remains.
Collapse
Affiliation(s)
- Sarah A. Dabydeen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA 20057
| | - Priscilla A. Furth
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA 20057
- Department of Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA 20057
| |
Collapse
|
17
|
MicroRNA-128 regulates the differentiation of rat bone mesenchymal stem cells into neuron-like cells by Wnt signaling. Mol Cell Biochem 2013; 387:151-8. [DOI: 10.1007/s11010-013-1880-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 10/18/2013] [Indexed: 01/15/2023]
|
18
|
Ford NA, Devlin KL, Lashinger LM, Hursting SD. Deconvoluting the obesity and breast cancer link: secretome, soil and seed interactions. J Mammary Gland Biol Neoplasia 2013; 18:267-75. [PMID: 24091864 PMCID: PMC3874287 DOI: 10.1007/s10911-013-9301-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/24/2013] [Indexed: 12/20/2022] Open
Abstract
Obesity is associated with increased risk of breast cancer in postmenopausal women and is linked with poor prognosis in pre- and postmenopausal breast cancer patients. The mechanisms underlying the obesity-breast cancer connection are becoming increasingly clear and provide multiple opportunities for primary to tertiary prevention. Several obesity-related host factors can influence breast tumor initiation, progression and/or response to therapy, and these have been implicated as key contributors to the complex effects of obesity on cancer incidence and outcomes. These host factors include components of the secretome, including insulin, insulin-like growth factor-1, leptin, adiponectin, steroid hormones, cytokines, vascular regulators, and inflammation-related molecules, as well as the cellular and structural components of the tumor microenvironment. These secreted and structural host factors are extrinsic to, and interact with, the intrinsic molecular characteristics of breast cancer cells (including breast cancer stem cells), and each will be considered in the context of energy balance and as potential targets for cancer prevention.
Collapse
Affiliation(s)
- Nikki A. Ford
- Department of Nutritional Sciences, University of Texas at Austin, Austin, Texas 78722, USA
| | - Kaylyn L. Devlin
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas 78722, USA
| | - Laura M. Lashinger
- Department of Nutritional Sciences, University of Texas at Austin, Austin, Texas 78722, USA
| | - Stephen D. Hursting
- Department of Nutritional Sciences, University of Texas at Austin, Austin, Texas 78722, USA
- Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| |
Collapse
|
19
|
Ford NA, Dunlap SM, Wheatley KE, Hursting SD. Obesity, independent of p53 gene dosage, promotes mammary tumor progression and upregulates the p53 regulator microRNA-504. PLoS One 2013; 8:e68089. [PMID: 23840816 PMCID: PMC3696069 DOI: 10.1371/journal.pone.0068089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/24/2013] [Indexed: 12/21/2022] Open
Abstract
Obesity, prevalent in >35% of US women, is an established risk and progression factor for postmenopausal breast cancer, and strategies to break the obesity-breast cancer link are urgently needed. Approximately 30% of breast cancers carry p53 tumor suppressor gene alterations; however, the effects of obesity on breast cancer progression in relation to p53 gene dosage are unclear. Using murine models of postmenopausal breast cancer, we characterized the interactive effects of diet-induced obesity (DIO) and p53 gene dosage on mammary tumor growth and associated p53-related regulatory mechanisms. Ovariectomized C57BL/6 mice were randomly assigned to receive a DIO or control diet, and (at 10 weeks) orthotopic injection of MMTV-Wnt-1 p53+/− or MMTV-Wnt-1 p53+/+ mammary tumor cells (n = 20 mice per diet and genotype group). DIO and control diets produced distinct phenotypes (mean percent body fat at 10 weeks: 57% and 39%, respectively, P<0.001). Regardless of phenotype, time to first palpable tumor was 57% less for Wnt-1 p53+/− than Wnt-1 p53+/+ tumors. Regardless of tumoral p53 genotype, DIO (relative to control) increased tumor burden, tumor cell proliferation (Ki-67), severity of tumor pathology, local tissue invasion, epithelial-to-mesenchymal transition (EMT) programming, and tumoral microRNA-504 (a negative regulator of p53) expression; and suppressed p53, p21, and estrogen receptor-alpha protein expression. These findings in murine models of postmenopausal breast cancer suggest that obesity may augment procancer effects related to p53 gene alterations. Furthermore, microRNA-504, an obesity-responsive negative regulator of p53 and putative EMT regulator, may represent a novel molecular target for breaking the obesity-breast cancer link.
Collapse
Affiliation(s)
- Nikki A. Ford
- Department of Nutritional Sciences University of Texas at Austin, Austin, Texas, United States of America
| | - Sarah M. Dunlap
- Department of Nutritional Sciences University of Texas at Austin, Austin, Texas, United States of America
| | - Karrie E. Wheatley
- Department of Nutritional Sciences University of Texas at Austin, Austin, Texas, United States of America
| | - Stephen D. Hursting
- Department of Nutritional Sciences University of Texas at Austin, Austin, Texas, United States of America
- Department of Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, United States of America
- * E-mail:
| |
Collapse
|
20
|
Mohibi S, Mirza S, Band H, Band V. Mouse models of estrogen receptor-positive breast cancer. J Carcinog 2011; 10:35. [PMID: 22279420 PMCID: PMC3263010 DOI: 10.4103/1477-3163.91116] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 10/20/2011] [Indexed: 12/22/2022] Open
Abstract
Breast cancer is the most frequent malignancy and second leading cause of cancer-related deaths among women. Despite advances in genetic and biochemical analyses, the incidence of breast cancer and its associated mortality remain very high. About 60 – 70% of breast cancers are Estrogen Receptor alpha (ER-α) positive and are dependent on estrogen for growth. Selective estrogen receptor modulators (SERMs) have therefore provided an effective targeted therapy to treat ER-α positive breast cancer patients. Unfortunately, development of resistance to endocrine therapy is frequent and leads to cancer recurrence. Our understanding of molecular mechanisms involved in the development of ER-α positive tumors and their resistance to ER antagonists is currently limited due to lack of experimental models of ER-α positive breast cancer. In most mouse models of breast cancer, the tumors that form are typically ER-negative and independent of estrogen for their growth. However, in recent years more attention has been given to develop mouse models that develop different subtypes of breast cancers, including ER-positive tumors. In this review, we discuss the currently available mouse models that develop ER-α positive mammary tumors and their potential use to elucidate the molecular mechanisms of ER-α positive breast cancer development and endocrine resistance.
Collapse
Affiliation(s)
- Shakur Mohibi
- Department of Genetics, Cell Biology, and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | | | | | | |
Collapse
|
21
|
Zhang C, Mori M, Gao S, Li A, Hoshino I, Aupperlee MD, Haslam SZ, Xiao H. Tip30 deletion in MMTV-Neu mice leads to enhanced EGFR signaling and development of estrogen receptor-positive and progesterone receptor-negative mammary tumors. Cancer Res 2011; 70:10224-33. [PMID: 21159643 DOI: 10.1158/0008-5472.can-10-3057] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estrogen receptor-positive and progesterone receptor-negative (ER+/PR-) breast cancers account for 15% to 25% of all human breast cancers and display more aggressive malignant characteristics than ER+/PR+ cancers. However, the molecular mechanism underlying development of ER+/PR- breast cancers still remains elusive. We show here that Tip30 deletion dramatically accelerated the onset of mammary tumors in the MMTV-Neu mouse model of breast cancer. The mammary tumors arising in Tip30(-/-)/MMTV-Neu mice were exclusively ER+/PR-. The growth of these ER+/PR- tumors depends not only on estrogen but also on progesterone despite the absence of detectable PR. Tip30 is predominantly expressed in ER+ mammary epithelial cells, and its deletion leads to an increase in the number of phospho-ERα-positive cells in mammary glands and accelerated activation of Akt in MMTV-Neu mice. Moreover, we found that Tip30 regulates the EGFR pathway through controlling endocytic downregulation of EGFR protein level and signaling. Together, these findings suggest a novel mechanism in which loss of Tip30 cooperates with Neu activation to enhance the activation of Akt signaling, leading to the development of ER+/PR- mammary tumors.
Collapse
Affiliation(s)
- Chengliang Zhang
- Department of Biomedical and Integrative Physiology, College of Human Medicine, Michigan State University, 3193 Biomedical and Physical Sciences Building, East Lansing, MI 48824-3320, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Somatic expression of PyMT or activated ErbB2 induces estrogen-independent mammary tumorigenesis. Neoplasia 2011; 12:718-26. [PMID: 20824048 DOI: 10.1593/neo.10516] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 06/04/2010] [Accepted: 06/07/2010] [Indexed: 11/18/2022] Open
Abstract
Estrogen signaling is required for the proliferation of normal breast epithelial cells. However, prophylactic inhibition of estrogen signaling fails to prevent 56% of human breast cancer cases. The underlying mechanism is not well understood. Aberrant activation of growth factor signaling is known to provide alternative proliferation pathways in breast cells that are fully transformed, but it is not known whether activation of growth factor signaling can substitute for estrogen signaling in causing aberrant proliferation in the normal breast epithelium. Here, we report that in a retrovirus-based somatic mouse model (replication-competent ALV-LTR splice acceptor/tumor virus A) that closely mimics the evolution of sporadic human breast cancers, mammary epithelial cells harboring PyMT or activated ErbB2 evolve into tumors independent of estrogen or other ovarian functions in contrast to previous observations of estrogen-dependent cancer formation in germ line mouse models of ErbB2 activation. Importantly, ErbB2 activation in normal mammary cells causes estrogen-independent proliferation in both estrogen receptor (ER)-negative cells as well as in normally quiescent ER-positive cells. Therefore, aberrant activation of growth factor signaling contributes to estrogen-independent proliferation of both preneoplastic and cancerous mammary cells, and prophylactic therapy against both growth factor signaling and estrogen signaling may need to be considered in women with increased risk of breast cancer.
Collapse
|
23
|
Fuchs-Young R, Shirley SH, Lambertz I, Colby JKL, Tian J, Johnston D, Gimenez-Conti IB, Donehower LA, Conti CJ, Hursting SD. P53 genotype as a determinant of ER expression and tamoxifen response in the MMTV-Wnt-1 model of mammary carcinogenesis. Breast Cancer Res Treat 2010; 130:399-408. [PMID: 21191649 DOI: 10.1007/s10549-010-1308-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 12/10/2010] [Indexed: 10/18/2022]
Abstract
Clinical studies show that estrogen receptor-α (ER) expressing tumors tend to have better prognosis, respond to antiestrogen therapy and have wild-type p53. Conversely, tumors with inactivating mutations in p53 tend to have worse outcomes and to be ER-negative and unresponsive to antihormone treatment. Previous studies from our laboratory have shown that p53 regulates ER expression transcriptionally, by binding the ER promoter and forming a complex with CARM1, CBP, c-Jun, RNA polymerase II and Sp1. In this study, the MMTV-Wnt-1 transgenic mouse model was used to demonstrate that p53 regulation of ER expression and function is not solely an in vitro phenomenon, but it is also operational in mammary tumorigenesis in vivo. The expression of ER and the ability to respond to tamoxifen were determined in mammary tumors arising in p53 wild type (WT) or p53 heterozygous (HT) animals carrying the Wnt-1 transgene. In p53 WT mice, development of ER-positive tumors was delayed by tamoxifen treatment, while tumors arising in p53 HT mice had significantly reduced levels of ER and were not affected by tamoxifen. P53 null tumors were also found in the p53 HT mice and these tumors were ER-negative. ER expression was upregulated in mouse mammary tumor cell lines following transfection with WT p53 or treatment with doxorubicin. These data demonstrate that p53 regulates ER expression in vivo, and affects response to tamoxifen. Results also provide an explanation for the concordant relationship between these prognostic proteins in human breast tumors.
Collapse
Affiliation(s)
- Robin Fuchs-Young
- Department of Molecular Carcinogenesis, The Virginia Harris Cockrell Cancer Research Center at The University of Texas MD Anderson Cancer Center, Science Park Research Division, 1808 Park Road 1C, Smithville, TX 78957, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Mastroianni M, Kim S, Kim YC, Esch A, Wagner C, Alexander CM. Wnt signaling can substitute for estrogen to induce division of ERalpha-positive cells in a mouse mammary tumor model. Cancer Lett 2009; 289:23-31. [PMID: 19665836 DOI: 10.1016/j.canlet.2009.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 04/30/2009] [Accepted: 07/15/2009] [Indexed: 01/03/2023]
Abstract
The interaction of estrogen with the estrogen receptor (ER, principally ERalpha) induces growth of human breast tumor cells. In contrast, ERalpha-positive cells have been described as non-dividing cells in normal breast (though estrogen stimulation of ERalpha cells directs the division of neighboring cells). However, there is a small sub-population of cells in normal mammary tissue that are ERalpha-positive, that can divide, and therefore share this property with human breast tumor cells. In order to investigate their pattern of growth regulation, we measured the fraction of dividing ERalpha(+) cells during normal growth and compared that to glands stimulated by oncogenic Wnt effectors. First, we found there was no difference between the rate of division of ERalpha(+) cells and ERalpha(-) cells, whether the population was responding to estrogen or Wnt mitogens. The proportion of dividing ERalpha(+) mammary epithelial cells was increased (10x) in response to pregnancy, and similar increases were observed in response to ectopic Wnt signaling. We propose that Wnt signaling can substitute for estrogen to drive total population growth (that includes ERalpha(+) cells). Although the E-ERalpha-derived mitogenic response is situated in a minority of the luminal cells, and the Wnt-LRP5/6-derived mitogenic response is situated in a minority of basal cells, overall, the growth response of the mammary epithelial population is remarkably similar.
Collapse
Affiliation(s)
- Melissa Mastroianni
- McArdle Lab for Cancer Research, University of Wisconsin, 1400 University Avenue, Madison 53706, United States
| | | | | | | | | | | |
Collapse
|
25
|
Met induces diverse mammary carcinomas in mice and is associated with human basal breast cancer. Proc Natl Acad Sci U S A 2009; 106:12909-14. [PMID: 19567831 DOI: 10.1073/pnas.0810403106] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Understanding the signaling pathways that drive aggressive breast cancers is critical to the development of effective therapeutics. The oncogene MET is associated with decreased survival in breast cancer, yet the role that MET plays in the various breast cancer subtypes is unclear. We describe a knockin mouse with mutationally activated Met (Met(mut)) that develops a high incidence of diverse mammary tumors with basal characteristics, including metaplasia, absence of progesterone receptor and ERBB2 expression, and expression of cytokeratin 5. With gene expression and tissue microarray analysis, we show that high MET expression in human breast cancers significantly correlated with estrogen receptor negative/ERBB2 negative tumors and with basal breast cancers. Few treatment options exist for breast cancers of the basal or trastuzumab-resistant ERBB2 subtypes. We conclude from these studies that MET may play a critical role in the development of the most aggressive breast cancers and may be a rational therapeutic target.
Collapse
|
26
|
Liu S, Umezu-Goto M, Murph M, Lu Y, Liu W, Zhang F, Yu S, Stephens LC, Cui X, Murrow G, Coombes K, Muller W, Hung MC, Perou CM, Lee AV, Fang X, Mills GB. Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell 2009; 15:539-50. [PMID: 19477432 PMCID: PMC4157573 DOI: 10.1016/j.ccr.2009.03.027] [Citation(s) in RCA: 306] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 01/05/2009] [Accepted: 03/26/2009] [Indexed: 02/06/2023]
Abstract
Lysophosphatidic acid (LPA) acts through high-affinity G protein-coupled receptors to mediate a plethora of physiological and pathological activities associated with tumorigenesis. LPA receptors and autotaxin (ATX/LysoPLD), the primary enzyme producing LPA, are aberrantly expressed in multiple cancer lineages. However, the role of ATX and LPA receptors in the initiation and progression of breast cancer has not been evaluated. We demonstrate that expression of ATX or each edg family LPA receptor in mammary epithelium of transgenic mice is sufficient to induce a high frequency of late-onset, estrogen receptor (ER)-positive, invasive, and metastatic mammary cancer. Thus, ATX and LPA receptors can contribute to the initiation and progression of breast cancer.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/secondary
- Animals
- Carcinoma, Adenosquamous/metabolism
- Carcinoma, Adenosquamous/pathology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cloning, Molecular
- Female
- Humans
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lymphatic Metastasis
- Male
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Transgenic
- Multienzyme Complexes/genetics
- Multienzyme Complexes/metabolism
- Neoplasm Invasiveness
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Phosphodiesterase I/genetics
- Phosphodiesterase I/metabolism
- Phosphoric Diester Hydrolases
- Pyrophosphatases/genetics
- Pyrophosphatases/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Lysophosphatidic Acid/genetics
- Receptors, Lysophosphatidic Acid/physiology
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Shuying Liu
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Makiko Umezu-Goto
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Mandi Murph
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Yiling Lu
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Wenbin Liu
- Department of Bioinformatics and Computational Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Fan Zhang
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Shuangxing Yu
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - L. Clifton Stephens
- Department of Veterinary Medicine & Surgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaojiang Cui
- Lester and Sue Smith Breast Center, Baylor College of Medicine; Houston, TX 77030, USA
- Department of Molecular Oncology, John Wayne Cancer Institute Saint John's Health Center, Santa Monica, CA 90404, USA
| | - George Murrow
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kevin Coombes
- Department of Bioinformatics and Computational Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Adrian V. Lee
- Lester and Sue Smith Breast Center, Baylor College of Medicine; Houston, TX 77030, USA
| | - Xianjun Fang
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Gordon B. Mills
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: Dr. Gordon B. Mills, Department of Systems Biology, M D Anderson Cancer Center 1515 Holcombe Blvd., Houston, TX 77030, USA, , Tel (713) 563-4200, Fax (713) 563-4235
| |
Collapse
|
27
|
Glover CE, Gurley KE, Kim KH, Storer B, Fero ML, Kemp CJ. Endocrine dysfunction in p27Kip1 deficient mice and susceptibility to Wnt-1 driven breast cancer. Carcinogenesis 2009; 30:1058-63. [PMID: 19380520 DOI: 10.1093/carcin/bgp089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The cyclin-dependent kinase (Cdk) inhibitor p27(Kip1) (p27) is a marker of prognosis in many cancers, including breast cancer. Low p27 expression correlates with poor prognosis, especially in hormone receptor positive breast tumors. This association suggests a role for p27 in hormone-dependent cancer. We used the Wnt-1 transgenic mouse model to further explore the role of p27 in hormone-driven breast cancer. We found that p27 deficiency did not alter breast cancer rate in either male or female Wnt-1 mice. However, we did find p27-/- females had reduced levels of serum progesterone (P) and increased variability in estradiol (E), which could have affected their cancer susceptibility. To equalize hormone levels, an additional cohort of Wnt-1 female mice was ovariectomized and implanted with slow release pellets of E and P. Although this treatment did not alter the breast cancer rate, it did accelerate the development of pituitary and gastric tumors in p27-/- mice. This study shows that while not a significant inhibitor of Wnt-1-driven breast cancer, p27 inhibits gastric tumors, whose latency is modulated by sex steroids.
Collapse
|
28
|
Brown PH, Subbaramaiah K, Salmon AP, Baker R, Newman RA, Yang P, Zhou XK, Bissonnette RP, Dannenberg AJ, Howe LR. Combination chemoprevention of HER2/neu-induced breast cancer using a cyclooxygenase-2 inhibitor and a retinoid X receptor-selective retinoid. Cancer Prev Res (Phila) 2009; 1:208-14. [PMID: 19138958 DOI: 10.1158/1940-6207.capr-08-0021] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The inducible prostaglandin synthase isoform cyclooxygenase-2 (COX-2) is overexpressed in approximately 40% of human breast carcinomas and in precancerous breast lesions, particularly in association with overexpression of human epidermal growth factor receptor 2 (HER2/neu). Experimental breast cancer can be suppressed by pharmacologic inhibition or genetic ablation of Cox-2, suggesting potential clinical utility of COX-2 inhibitors with respect to breast cancer. Importantly, several clinical trials have found reduced colorectal adenoma formation in individuals administered selective COX-2 inhibitors. However, such trials also identified increased cardiovascular risk associated with COX-2 inhibitor use. The goal of this research was to test whether improved chemopreventive efficacy could be achieved by combining submaximal doses of a selective COX-2 inhibitor and a retinoid X receptor-selective retinoid (rexinoid). The rate of HER2/neu-induced mammary tumor formation was substantially delayed by coadministration of the COX-2 inhibitor celecoxib (500 ppm in diet) and the rexinoid LGD1069 (10 mg/kg body weight; oral gavage) to MMTV/neu mice. Median time to tumor formation was increased from 304 to >600 days (P < 0.0001). The combination was substantially more effective than either drug individually. Similarly, potent suppression of aromatase activity was observed in mammary tissues from the combination cohort (44% of control; P < 0.001). Regulation of aromatase expression and activity by COX-derived prostaglandins is well established. Interestingly however, single agent LGD1069 significantly reduced mammary aromatase activity (71% of control; P < 0.001) without modulating eicosanoid levels. Our data show that simultaneous blockade of COX/prostaglandin signaling and retinoid X receptor-dependent transcription confers potent anticancer efficacy, suggesting a novel avenue for clinical evaluation.
Collapse
Affiliation(s)
- Powel H Brown
- Breast Center, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Nunez NP, Perkins SN, Smith NCP, Berrigan D, Berendes DM, Varticovski L, Barrett JC, Hursting SD. Obesity accelerates mouse mammary tumor growth in the absence of ovarian hormones. Nutr Cancer 2008; 60:534-41. [PMID: 18584488 DOI: 10.1080/01635580801966195] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Obesity increases incidence and mortality of breast cancer in postmenopausal women. Mechanisms underlying this association are poorly understood. Suitable animal models are needed to elucidate potential mechanisms for this association. To determine the effects of obesity on mammary tumor growth, nonovariectomized and ovariectomized C57BL/6 mice of various body weights (lean, overweight, and obese) were implanted subcutaneously with mammary tumor cells from syngeneic Wnt-1 transgenic mice. In mice, the lean phenotype was associated with reduced Wnt-1 tumor growth regardless of ovarian hormone status. Ovariectomy delayed Wnt-1 tumor growth consistent with the known hormone responsiveness of these tumors. However, obesity accelerated tumor growth in ovariectomized but not in nonovariectomized animals. Diet-induced obesity in a syngeneic mouse model of breast cancer enhanced tumor growth, specifically in the absence of ovarian hormones. These results support epidemiological evidence that obesity is associated with increased breast cancer incidence and mortality in postmenopausal but not premenopausal women. In contrast, maintaining a lean body weight phenotype was associated with reduced Wnt-1 tumor growth regardless of ovarian hormone status.
Collapse
Affiliation(s)
- Nomeli P Nunez
- Laboratory of Biosystems and Cancer, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1758, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Bennett CN, Green JE. Unlocking the power of cross-species genomic analyses: identification of evolutionarily conserved breast cancer networks and validation of preclinical models. Breast Cancer Res 2008; 10:213. [PMID: 18828875 PMCID: PMC2614501 DOI: 10.1186/bcr2125] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The application of high-throughput genomic technologies has revealed that individual breast tumors display a variety of molecular features that require more personalized approaches to treatment. Several recent studies have demonstrated that a cross-species analytic approach provides a powerful means to filter through genetic complexity by identifying evolutionarily conserved genetic networks that are fundamental to the oncogenic process. Mouse-human tumor comparisons will provide insights into cellular origins of tumor subtypes, define interactive oncogenetic networks, identify potential novel therapeutic targets, and further validate as well as guide the selection of genetically engineered mouse models for preclinical testing.
Collapse
Affiliation(s)
- Christina N Bennett
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | | |
Collapse
|
31
|
Comparison of expression profiles of metastatic versus primary mammary tumors in MMTV-Wnt-1 and MMTV-Neu transgenic mice. Neoplasia 2008; 10:118-24. [PMID: 18283333 DOI: 10.1593/neo.07637] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 11/15/2007] [Accepted: 11/16/2007] [Indexed: 01/09/2023] Open
Abstract
Distant metastases of human breast cancers have been suggested to be more different from each other than from their respective primary tumors, based on expression profiling. The mechanism behind this lack of similarity between individual metastases is not known. We used cDNA microarrays to determine the expression profiles of pulmonary metastases and primary mammary tumors in two distinct transgenic models expressing either the Neu or the Wnt-1 oncogene from the mouse mammary tumor virus long terminal repeat (MMTV LTR). We found that pulmonary metastases are similar to each other and to their primary tumors within the same line. However, metastases arising in one transgenic mouse line are very different from either metastases or primary tumors arising in the other line. In addition, we found that, like their primary tumors, lung metastases in Wnt-1 transgenic mice harbor both epithelial and myoepithelial tumor cells and cells that express the putative progenitor cell marker keratin 6. Our data suggest that both gene expression profiles and cellular heterogeneity are preserved after breast cancer has spread to distant sites, and that metastases are similar to each other when their primary tumors were induced by the same oncogene and from the same subset of mammary cells.
Collapse
|
32
|
Cho RW, Wang X, Diehn M, Shedden K, Chen GY, Sherlock G, Gurney A, Lewicki J, Clarke MF. Isolation and molecular characterization of cancer stem cells in MMTV-Wnt-1 murine breast tumors. Stem Cells 2007; 26:364-71. [PMID: 17975224 DOI: 10.1634/stemcells.2007-0440] [Citation(s) in RCA: 233] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In human breast cancers, a phenotypically distinct minority population of tumorigenic (TG) cancer cells (sometimes referred to as cancer stem cells) drives tumor growth when transplanted into immunodeficient mice. Our objective was to identify a mouse model of breast cancer stem cells that could have relevance to the study of human breast cancer. To do so, we used breast tumors of the mouse mammary tumor virus (MMTV)-Wnt-1 mice. MMTV-Wnt-1 breast tumors were harvested, dissociated into single-cell suspensions, and sorted by flow cytometry on Thy1, CD24, and CD45. Sorted cells were then injected into recipient background FVB/NJ female syngeneic mice. In six of seven tumors examined, Thy1+CD24+ cancer cells, which constituted approximately 1%-4% of tumor cells, were highly enriched for cells capable of regenerating new tumors compared with cells of the tumor that did not fit this profile ("not-Thy1+CD24+"). Resultant tumors had a phenotypic diversity similar to that of the original tumor and behaved in a similar manner when passaged. Microarray analysis comparing Thy1+CD24+ tumor cells to not-Thy1+CD24+ cells identified a list of differentially expressed genes. Orthologs of these differentially expressed genes predicted survival of human breast cancer patients from two different study groups. These studies suggest that there is a cancer stem cell compartment in the MMTV-Wnt-1 murine breast tumor and that there is a clinical utility of this model for the study of cancer stem cells.
Collapse
Affiliation(s)
- Robert W Cho
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kumar MJM, Ponvijay KS, Nandhini R, Nagarajan RS, Jose J, Srinivas G, Nagarajan P, Venkatesan R, Kumar K, Singh S. A mouse model for luminal epithelial like ER positive subtype of human breast cancer. BMC Cancer 2007; 7:180. [PMID: 17880731 PMCID: PMC2094712 DOI: 10.1186/1471-2407-7-180] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Accepted: 09/20/2007] [Indexed: 11/10/2022] Open
Abstract
Background Generation of novel spontaneous ER positive mammary tumor animal model from heterozygous NIH nude mice. Methods Using brother-sister mating with pedigree expansion system, we derived a colony of heterozygous breeding females showing ER-Positive tumors around the age of 6 months. Complete blood picture, differential leukocyte count, and serum levels of Estrogen, Alanine amino transferase (SGPT), Aspartate amino transferase (SGOT), total protein and albumin were estimated. Aspiration biopsies and microbiology were carried out. Gross pathology of the tumors and their metastatic potential were assessed. The tumors were excised and further characterized using histopathology, cytology, electron microscopy (EM), molecular markers and Mouse mammary Tumor Virus – Long Terminal Repeats (MMTV LTR) specific RT-PCR. Results The tumors originated from 2ndor 5thor both the mammary glands and were multi-nodulated with variable central necrosis accompanied with an accumulation of inflammatory exudate. Significant increases in estrogen, SGPT, SGOT and neutrophils levels were noticed. Histopathologically, invasive nodular masses of pleomorphic tubular neoplastic epithelial cells invaded fibro-vascular stroma, adjacent dermis and subcutaneous tissue. Metastatic spread through hematogenous and regional lymph nodes, into liver, lungs, spleen, heart and dermal lymphatics was observed. EM picture revealed no viral particles and MMTV-negativity was confirmed through MMTV LTR-specific RT-PCR. High expression of ER α, moderate to high expression of proliferating cell nuclear antigen (PCNA), moderate expression of vimentin and Cytokeratin 19 (K19) and low expression of p53 were observed in tumor sections, when compared with that of the normal mammary gland. Conclusion Since 75% of human breast cancer were classified ER-positive and as our model mimics (in most of the characteristics, such as histopathology, metastasis, high estrogen levels) the ER-positive luminal epithelial-like human breast cancer, this model will be an attractive tool to understand the biology of estrogen-dependant breast cancer in women. To our knowledge, this is the first report of a spontaneous mammary model displaying regional lymph node involvement with both hematogenous and lymphatic spread to liver, lung, heart, spleen and lymph nodes.
Collapse
MESH Headings
- Animals
- BRCA1 Protein/metabolism
- Biomarkers, Tumor/metabolism
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/secondary
- Disease Models, Animal
- Estrogens/metabolism
- Female
- Heart Neoplasms/metabolism
- Heart Neoplasms/secondary
- Heterozygote
- Humans
- Immunohistochemistry
- Inbreeding
- Keratin-19/metabolism
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lymphatic Metastasis
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Nude
- Proliferating Cell Nuclear Antigen/metabolism
- Receptors, Estrogen/metabolism
- Splenic Neoplasms/metabolism
- Splenic Neoplasms/secondary
- Tumor Suppressor Protein p53/metabolism
- Vimentin/metabolism
Collapse
Affiliation(s)
- MJ Mahesh Kumar
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - KS Ponvijay
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - R Nandhini
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - RS Nagarajan
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - J Jose
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - G Srinivas
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - P Nagarajan
- National Institute of Immunology, New Delhi 110067, India
| | - R Venkatesan
- National Institute of Immunology, New Delhi 110067, India
| | - Kishor Kumar
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - S Singh
- Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| |
Collapse
|
34
|
Kuorelahti A, Rulli S, Huhtaniemi I, Poutanen M. Human chorionic gonadotropin (hCG) up-regulates wnt5b and wnt7b in the mammary gland, and hCGbeta transgenic female mice present with mammary Gland tumors exhibiting characteristics of the Wnt/beta-catenin pathway activation. Endocrinology 2007; 148:3694-703. [PMID: 17510243 DOI: 10.1210/en.2007-0249] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transgenic (TG) mice expressing human chorionic gonadotropin (hCG) beta-subunit under the ubiquitin C promoter, presenting with a moderately elevated level of LH/hCG bioactivity develop multiple neoplasms secondary to the endocrine abnormalities, including mammary gland tumors after the age of 9 months. The increased levels of circulating estradiol, progesterone, and prolactin of the TG females after puberty boost the lobuloalveolar development in the mammary gland resulting ultimately in the formation of estrogen and progesterone receptor-negative, malignant tumors. These tumors have a similar histopathology with those observed in TG mice with activated wnt/beta-catenin pathway, showing increased expression of beta-catenin, also a common finding in human breast tumors. Transdifferentiation is observed in mammary tumors of the hCGbeta TG mice, accompanied by abnormal expression of the Wnt genes in the tumorous and nontumorous mammary gland tissue. Specifically we found increased expression of Wnt5b in the TG mammary glands at the age of 3 months and up-regulation of Wnt7b and -5b in the subsequently appearing tumors. Importantly, hCG was found to up-regulate these wnt ligands in mouse mammary gland, independent of the changes in ovarian steroidogenesis. Thus, the hCGbeta-overexpressing TG mice represent a novel model that links enhanced hCG action to dysregulated wnt signaling in the mammary gland, resulting in beta-catenin-stabilizing mammary tumorigenesis. The novel finding of hCG up-regulating wnt7b and wnt5b could contribute to pregnancy-induced breast cancer in humans.
Collapse
MESH Headings
- Age Factors
- Animals
- Chorionic Gonadotropin, beta Subunit, Human/genetics
- Chorionic Gonadotropin, beta Subunit, Human/metabolism
- Estrogen Receptor alpha/metabolism
- Female
- Glycoproteins/metabolism
- Humans
- Mammary Glands, Animal/pathology
- Mammary Glands, Animal/physiology
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mammary Neoplasms, Animal/physiopathology
- Mice
- Mice, Inbred Strains
- Mice, Transgenic
- Myoepithelioma/metabolism
- Myoepithelioma/pathology
- Myoepithelioma/physiopathology
- Ovariectomy
- Proto-Oncogene Proteins/metabolism
- Receptors, Progesterone/metabolism
- Up-Regulation/physiology
- Wnt Proteins/metabolism
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Aino Kuorelahti
- Department of Physiology, Institute of Biomedicine, and Turku Graduate School of Biomedical Sciences, University of Turku, 20520 Turku, Finland
| | | | | | | |
Collapse
|
35
|
Rose-Hellekant TA, Schroeder MD, Brockman JL, Zhdankin O, Bolstad R, Chen KS, Gould MN, Schuler LA, Sandgren EP. Estrogen receptor-positive mammary tumorigenesis in TGFalpha transgenic mice progresses with progesterone receptor loss. Oncogene 2007; 26:5238-46. [PMID: 17334393 PMCID: PMC2587149 DOI: 10.1038/sj.onc.1210340] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We characterized the novel NRL-transforming growth factor alpha (NRL-TGFalpha) transgenic mouse model in which growth factor - steroid receptor interactions were explored. The NRL promoter directs transgene expression to mammary ductal and alveolar cells and is nonresponsive to estrogen manipulations in vitro and in vivo. NRL-TGFalpha mice acquire proliferative hyperplasias as well as cystic and solid tumors. Quantitative transcript analysis revealed a progressive decrease in estrogen receptor alpha (ER) and progesterone receptor (PR) mRNA levels with tumorigenesis. However, ER protein was evident in all lesion types and in surrounding stromal cells using immunohistochemistry. PR protein was identified in normal epithelial cells and in very few cells of small epithelial hyperplasias, but never in stromal or tumor cells. Prophylactic ovariectomy significantly delayed tumor development and decreased incidence. Finally, while heterozygous (+/-) p53 mice did not acquire mammary lesions, p53+/- mice carrying the NRL-TGFalpha transgene developed ER negative/PR negative undifferentiated carcinomas. These data demonstrate that unregulated TGFalpha expression in the mammary gland leads to oncogenesis that is dependent on ovarian steroids early in tumorigenesis. Resulting tumors resemble a clinical phenotype of ER+/PR-, and when combined with a heterozygous p53 genotype, ER-/PR-.
Collapse
Affiliation(s)
- T A Rose-Hellekant
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Huang S, Podsypanina K, Chen Y, Cai W, Tsimelzon A, Hilsenbeck S, Li Y. Wnt-1 is dominant over neu in specifying mammary tumor expression profiles. Technol Cancer Res Treat 2007; 5:565-71. [PMID: 17121432 DOI: 10.1177/153303460600500603] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Wnt-1 and Neu collaborate to induce mammary tumors in bitransgenic mice carrying both MMTV-Wnt-1 and MMTV-Neu. In this report, gene expression profiles were determined for tumors from these bitransgenic mice, and compared with expression profiles of tumors from mice singly transgenic for MMTV-Wnt-1 or MMTV-Neu. While very different from tumors arising in MMTV-Neu transgenic mice, tumors from these bitransgenic mice were found not to have identifiable differences from tumors from MMTV-Wnt-1 transgenic mice, using clustering and multidimensional scaling analyses (unsupervised and supervised), One-way Analysis of Variance (ANOVA), and two sample t test (the later two of which were combined with false discovery rate computation). These observations suggest that Wnt-1 is dominant over Neu in specifying mammary tumor expression profiles.
Collapse
Affiliation(s)
- Shixia Huang
- Breast Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Singh M, Johnson L. Using genetically engineered mouse models of cancer to aid drug development: an industry perspective. Clin Cancer Res 2006; 12:5312-28. [PMID: 17000664 DOI: 10.1158/1078-0432.ccr-06-0437] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Recent developments in the generation and characterization of genetically engineered mouse models of human cancer have resulted in notable improvements in these models as platforms for preclinical target validation and experimental therapeutics. In this review, we enumerate the criteria used to assess the accuracy of various models with respect to human disease and provide some examples of their prognostic and therapeutic utility, focusing on models for cancers that affect the largest populations. Technological advancements that allow greater exploitation of genetically engineered mouse models, such as RNA interference in vivo, are described in the context of target and drug validation. Finally, this review discusses stratagems for, and obstacles to, the application of these models in the drug development process.
Collapse
Affiliation(s)
- Mallika Singh
- Genentech, Inc., South San Francisco, California 94080, USA
| | | |
Collapse
|
38
|
Namba R, Young LJT, Maglione JE, McGoldrick ET, Liu S, Wurz GT, DeGregorio MW, Borowsky AD, MacLeod CL, Cardiff RD, Gregg JP. Selective estrogen receptor modulators inhibit growth and progression of premalignant lesions in a mouse model of ductal carcinoma in situ. Breast Cancer Res 2005; 7:R881-9. [PMID: 16280035 PMCID: PMC1410776 DOI: 10.1186/bcr1317] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Revised: 08/10/2005] [Accepted: 08/17/2005] [Indexed: 12/04/2022] Open
Abstract
Introduction Ductal carcinoma in situ (DCIS) is a noninvasive premalignant lesion and is considered a precursor to invasive carcinoma. DCIS accounts for nearly 20% of newly diagnosed breast cancer, but the lack of experimentally amenable in vivo DCIS models hinders the development of treatment strategies. Here, we demonstrate the utility of a mouse transplantation model of DCIS for chemoprevention studies using selective estrogen receptor modulators (SERMs). This model consists of a set of serially transplanted lines of genetically engineered mouse mammary intraepithelial neoplasia (MIN) outgrowth (MIN-O) tissue that have stable characteristics. We studied the ovarian-hormone-responsiveness of one of the lines with a particular focus on the effects of two related SERMs, tamoxifen and ospemifene. Methods The estrogen receptor (ER) status and ovarian-hormone-dependence of the mouse MIN outgrowth tissue were determined by immunohistochemistry and ovarian ablation. The effects of tamoxifen and ospemifene on the growth and tumorigenesis of MIN outgrowth were assessed at 3 and 10 weeks after transplantation. The effects on ER status, cell proliferation, and apoptosis were studied with immunohistochemistry. Results The MIN-O was ER-positive and ovarian ablation resulted in reduced MIN-O growth and tumor development. Likewise, tamoxifen and ospemifene treatments decreased the MIN growth and tumor incidence in comparison with the control (P < 0.01). Both SERMs significantly decreased cell proliferation. Between the two SERM treatment groups, there were no statistically significant differences in MIN-O size, tumor latency, or proliferation rate. In contrast, the ospemifene treatment significantly increased ER levels while tamoxifen significantly decreased them. Conclusion Tamoxifen and ospemifene inhibit the growth of premalignant mammary lesions and the progression to invasive carcinoma in a transplantable mouse model of DCIS. The inhibitory effects of these two SERMs are similar except for their effects on ER modulation. These differences in ER modulation may suggest different mechanisms of action between the two related SERMs and may portend different long-term outcomes. These data demonstrate the value of this model system for preclinical testing of antiestrogen or other therapies designed to prevent or delay the malignant transformation of premalignant mammary lesions in chemoprevention.
Collapse
Affiliation(s)
- Ruria Namba
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Lawrence JT Young
- Center for Comparative Medicine, University of California, Davis, CA, USA
| | - Jeannie E Maglione
- Center for Comparative Medicine, University of California, Davis, CA, USA
| | - Erik T McGoldrick
- Center for Comparative Medicine, University of California, Davis, CA, USA
| | - Stephenie Liu
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Gregory T Wurz
- Department of Internal Medicine, Division of Hematology and Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Michael W DeGregorio
- Department of Internal Medicine, Division of Hematology and Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Center for Comparative Medicine, University of California, Davis, CA, USA
| | - Carol L MacLeod
- Department of Medicine, UCSD Cancer Center, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Robert D Cardiff
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Center for Comparative Medicine, University of California, Davis, CA, USA
| | - Jeffrey P Gregg
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
39
|
Abstract
This review summarizes the current evidence for the existence of human breast stem cells and the pathways involved in their regulation, and discusses how the disruption of these pathways may result in the generation of a population of cells with the capacity for unlimited self-renewal. Relevant data from mouse model systems are also discussed where appropriate. By understanding the molecular pathways that regulate self-renewal of normal mammary stem cells, it may be possible to target the activation of these pathways in breast tumours.
Collapse
Affiliation(s)
- Helen Kalirai
- Breast Biology Group, Division of Cancer Studies, University of Manchester, Christie Hospital, Wilmslow Road, Manchester, M20 4BX, UK
| | | |
Collapse
|