51
|
Toivanen R, Shen MM. Prostate organogenesis: tissue induction, hormonal regulation and cell type specification. Development 2017; 144:1382-1398. [PMID: 28400434 DOI: 10.1242/dev.148270] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Prostate organogenesis is a complex process that is primarily mediated by the presence of androgens and subsequent mesenchyme-epithelial interactions. The investigation of prostate development is partly driven by its potential relevance to prostate cancer, in particular the apparent re-awakening of key developmental programs that occur during tumorigenesis. However, our current knowledge of the mechanisms that drive prostate organogenesis is far from complete. Here, we provide a comprehensive overview of prostate development, focusing on recent findings regarding sexual dimorphism, bud induction, branching morphogenesis and cellular differentiation.
Collapse
Affiliation(s)
- Roxanne Toivanen
- Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Michael M Shen
- Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| |
Collapse
|
52
|
MicroRNAs as regulators and mediators of forkhead box transcription factors function in human cancers. Oncotarget 2017; 8:12433-12450. [PMID: 27999212 PMCID: PMC5355356 DOI: 10.18632/oncotarget.14015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/07/2016] [Indexed: 02/07/2023] Open
Abstract
Evidence has shown that microRNAs are widely implicated as indispensable components of tumor suppressive and oncogenic pathways in human cancers. Thus, identification of microRNA targets and their relevant pathways will contribute to the development of microRNA-based therapeutics. The forkhead box transcription factors regulate numerous processes including cell cycle progression, metabolism, metastasis and angiogenesis, thereby facilitating tumor initiation and progression. A complex network of protein and non-coding RNAs mediates the expression and activity of forkhead box transcription factors. In this review, we summarize the current knowledge and concepts concerning the involvement of microRNAs and forkhead box transcription factors and describe the roles of microRNAs-forkhead box axis in various disease states including tumor initiation and progression. Additionally, we describe some of the technical challenges in the use of the microRNA-forkhead box signaling pathway in cancer treatment.
Collapse
|
53
|
TMPRSS2-ERG fusion co-opts master transcription factors and activates NOTCH signaling in primary prostate cancer. Nat Genet 2017; 49:1336-1345. [PMID: 28783165 DOI: 10.1038/ng.3930] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 07/12/2017] [Indexed: 12/19/2022]
Abstract
TMPRSS2-ERG (T2E) structural rearrangements typify ∼50% of prostate tumors and result in overexpression of the ERG transcription factor. Using chromatin, genomic and expression data, we show distinct cis-regulatory landscapes between T2E-positive and non-T2E primary prostate tumors, which include clusters of regulatory elements (COREs). This difference is mediated by ERG co-option of HOXB13 and FOXA1, implementing a T2E-specific transcriptional profile. We also report a T2E-specific CORE on the structurally rearranged ERG locus arising from spreading of the TMPRSS2 locus pre-existing CORE, assisting in its overexpression. Finally, we show that the T2E-specific cis-regulatory landscape underlies a vulnerability against the NOTCH pathway. Indeed, NOTCH pathway inhibition antagonizes the growth and invasion of T2E-positive prostate cancer cells. Taken together, our work shows that overexpressed ERG co-opts master transcription factors to deploy a unique cis-regulatory landscape, inducing a druggable dependency on NOTCH signaling in T2E-positive prostate tumors.
Collapse
|
54
|
Li L, Gong M, Zhao Y, Zhao X, Li Q. FOXK1 facilitates cell proliferation through regulating the expression of p21, and promotes metastasis in ovarian cancer. Oncotarget 2017; 8:70441-70451. [PMID: 29050292 PMCID: PMC5642567 DOI: 10.18632/oncotarget.19713] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/28/2017] [Indexed: 12/03/2022] Open
Abstract
Ovarian cancer is one of the most common cancer in the world. FOX family plays essential function in multiple cancers. In our work, FOXK1 was found to up-regulate in ovarian cancer tissue samples and cell lines; moreover, the expression of FOXK1 was correlated with tumor size, metastasis and poorly prognosis. To evaluate the function of FOXK1 in ovarian cancer, we performed colony formation analysis, CCK-8 assay and cell cycle analysis to determine the effect of FOXK1 on cell proliferation and cell cycle. We found that FOXK1 obviously improved the ability of cell proliferation through promoting cell cycle. Furthermore, ChIP assay and luciferase reporter assay indicated that FOXK1 facilitated cell cycle through regulating the expression of p21, but FOXK1 had no effect on cell apoptosis. In addition, wound healing assay and transwell invasion analysis demonstrated that FOXK1 promoted migration and invasion in ovarian cancer. In conclusion, our work indicate FOXK1 plays a key function in the ovarian cancer, it promotes cell proliferation and metastasis. FOXK1 serves as a novel molecular therapy target in ovarian cancer.
Collapse
Affiliation(s)
- Li Li
- Department of Histology and Embryology, Hebei Medical University, Hebei, China
| | - Miao Gong
- Department of Histology and Embryology, Hebei Medical University, Hebei, China
| | - Yu Zhao
- Department of Histology and Embryology, Hebei Medical University, Hebei, China
| | - Xiujun Zhao
- Department of Histology and Embryology, Hebei Medical University, Hebei, China
| | - Quanhai Li
- Department of Immunology, Hebei Medical University, Hebei, China
| |
Collapse
|
55
|
Interaction of prostate carcinoma-associated fibroblasts with human epithelial cell lines in vivo. Differentiation 2017; 96:40-48. [PMID: 28779656 DOI: 10.1016/j.diff.2017.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
Stromal-epithelial interactions play a crucial and poorly understood role in carcinogenesis and tumor progression. Mesenchymal-epithelial interactions have a long history of research in relation to the development of organs. Models designed to study development are often also applicable to studies of benign and malignant disease. Tumor stroma is a complex mixture of cells that includes a fibroblastic component often referred to as cancer-associated fibroblasts (CAF), desmoplasia or "reactive" stroma. Here we discuss the history of, and approaches to, understanding these interactions with particular reference to prostate cancer and to in vivo modeling using human cells and tissues. A series of studies have revealed a complex mixture of signaling molecules acting both within the stromal tissue and between the stromal and epithelial tissues. We are starting to understand the interactions of some of these pathways, however the work is still ongoing. This area of research provide a basis for new medical approaches aimed at stabilizing early stage cancers rendering them chronic rather than acute problems. Such work is especially relevant to slow growing tumors found in older patients, a class that would include many prostate cancers.
Collapse
|
56
|
Talos F, Mitrofanova A, Bergren SK, Califano A, Shen MM. A computational systems approach identifies synergistic specification genes that facilitate lineage conversion to prostate tissue. Nat Commun 2017; 8:14662. [PMID: 28429718 PMCID: PMC5413950 DOI: 10.1038/ncomms14662] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 01/21/2017] [Indexed: 12/19/2022] Open
Abstract
To date, reprogramming strategies for generating cell types of interest have been facilitated by detailed understanding of relevant developmental regulatory factors. However, identification of such regulatory drivers often represents a major challenge, as specific gene combinations may be required for reprogramming. Here we show that a computational systems approach can identify cell type specification genes (master regulators) that act synergistically, and demonstrate its application for reprogramming of fibroblasts to prostate tissue. We use three such master regulators (FOXA1, NKX3.1 and androgen receptor, AR) in a primed conversion strategy starting from mouse fibroblasts, resulting in prostate tissue grafts with appropriate histological and molecular properties that respond to androgen-deprivation. Moreover, generation of reprogrammed prostate does not require traversal of a pluripotent state. Thus, we describe a general strategy by which cell types and tissues can be generated even with limited knowledge of the developmental pathways required for their specification in vivo.
Collapse
Affiliation(s)
- Flaminia Talos
- Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | - Antonina Mitrofanova
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | - Sarah K Bergren
- Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | - Andrea Califano
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| | - Michael M Shen
- Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.,Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA
| |
Collapse
|
57
|
Groner AC, Brown M. Role of steroid receptor and coregulator mutations in hormone-dependent cancers. J Clin Invest 2017; 127:1126-1135. [PMID: 28368289 PMCID: PMC5373886 DOI: 10.1172/jci88885] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Steroid hormones mediate critical lineage-specific developmental and physiologic responses. They function by binding their cognate receptors, which are transcription factors that drive specific gene expression programs. The requirement of most prostate cancers for androgen and most breast cancers for estrogen has led to the development of endocrine therapies that block the action of these hormones in these tumors. While initial endocrine interventions are successful, resistance to therapy often arises. We will review how steroid receptor-dependent genomic signaling is affected by genetic alterations in endocrine therapy resistance. The detailed understanding of these interactions will not only provide improved treatment options to overcome resistance, but, in the future, will also be the basis for implementing precision cancer medicine approaches.
Collapse
Affiliation(s)
- Anna C. Groner
- Department of Medical Oncology and
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Myles Brown
- Department of Medical Oncology and
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
58
|
Abstract
Forkhead box (Fox) transcription factors are evolutionarily conserved in organisms ranging from yeast to humans. They regulate diverse biological processes both during development and throughout adult life. Mutations in many Fox genes are associated with human disease and, as such, various animal models have been generated to study the function of these transcription factors in mechanistic detail. In many cases, the absence of even a single Fox transcription factor is lethal. In this Primer, we provide an overview of the Fox family, highlighting several key Fox transcription factor families that are important for mammalian development.
Collapse
Affiliation(s)
- Maria L Golson
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
59
|
Hedgehog Signaling in Prostate Development, Regeneration and Cancer. J Dev Biol 2016; 4:jdb4040030. [PMID: 29615593 PMCID: PMC5831806 DOI: 10.3390/jdb4040030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 12/17/2022] Open
Abstract
The prostate is a developmental model system study of prostate growth regulation. Historically the research focus was on androgen regulation of development and growth and instructive interactions between the mesenchyme and epithelium. The study of Hh signaling in prostate development revealed important roles in ductal morphogenesis and in epithelial growth regulation that appear to be recapitulated in prostate cancer. This overview of Hh signaling in the prostate will address the well-described role of paracrine signaling prostate development as well as new evidence suggesting a role for autocrine signaling, the role of Hh signaling in prostate regeneration and reiterative activities in prostate cancer.
Collapse
|
60
|
Yang YA, Zhao JC, Fong KW, Kim J, Li S, Song C, Song B, Zheng B, He C, Yu J. FOXA1 potentiates lineage-specific enhancer activation through modulating TET1 expression and function. Nucleic Acids Res 2016; 44:8153-64. [PMID: 27257062 PMCID: PMC5041455 DOI: 10.1093/nar/gkw498] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 05/24/2016] [Indexed: 12/22/2022] Open
Abstract
Forkhead box A1 (FOXA1) is an FKHD family protein that plays pioneering roles in lineage-specific enhancer activation and gene transcription. Through genome-wide location analyses, here we show that FOXA1 expression and occupancy are, in turn, required for the maintenance of these epigenetic signatures, namely DNA hypomethylation and histone 3 lysine 4 methylation. Mechanistically, this involves TET1, a 5-methylcytosine dioxygenase. We found that FOXA1 induces TET1 expression via direct binding to its cis-regulatory elements. Further, FOXA1 physically interacts with the TET1 protein through its CXXC domain. TET1 thus co-occupies FOXA1-dependent enhancers and mediates local DNA demethylation and concomitant histone 3 lysine 4 methylation, further potentiating FOXA1 recruitment. Consequently, FOXA1 binding events are markedly reduced following TET1 depletion. Together, our results suggest that FOXA1 is not only able to recognize but also remodel the epigenetic signatures at lineage-specific enhancers, which is mediated, at least in part, by a feed-forward regulatory loop between FOXA1 and TET1.
Collapse
Affiliation(s)
- Yeqing A Yang
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jonathan C Zhao
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ka-Wing Fong
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jung Kim
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shangze Li
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chunxiao Song
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, the University of Chicago, Chicago, IL 60637, USA
| | - Bing Song
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Bin Zheng
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, the University of Chicago, Chicago, IL 60637, USA Howard Hughes Medical Institute, the University of Chicago, Chicago, IL 60637, USA
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
61
|
Grabowska MM, Kelly SM, Reese AL, Cates JM, Case TC, Zhang J, DeGraff DJ, Strand DW, Miller NL, Clark PE, Hayward SW, Gronostajski RM, Anderson PD, Matusik RJ. Nfib Regulates Transcriptional Networks That Control the Development of Prostatic Hyperplasia. Endocrinology 2016; 157:1094-109. [PMID: 26677878 PMCID: PMC4769366 DOI: 10.1210/en.2015-1312] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A functional complex consisting of androgen receptor (AR) and forkhead box A1 (FOXA1) proteins supports prostatic development, differentiation, and disease. In addition, the interaction of FOXA1 with cofactors such as nuclear factor I (NFI) family members modulates AR target gene expression. However, the global role of specific NFI family members has yet to be described in the prostate. In these studies, chromatin immunoprecipitation followed by DNA sequencing in androgen-dependent LNCaP prostate cancer cells demonstrated that 64.3% of NFIB binding sites are associated with AR and FOXA1 binding sites. Interrogation of published data revealed that genes associated with NFIB binding sites are predominantly induced after dihydrotestosterone treatment of LNCaP cells, whereas NFIB knockdown studies demonstrated that loss of NFIB drives increased AR expression and superinduction of a subset of AR target genes. Notably, genes bound by NFIB only are associated with cell division and cell cycle. To define the role of NFIB in vivo, mouse Nfib knockout prostatic tissue was rescued via renal capsule engraftment. Loss of Nfib expression resulted in prostatic hyperplasia, which did not resolve in response to castration, and an expansion of an intermediate cell population in a small subset of grafts. In human benign prostatic hyperplasia, luminal NFIB loss correlated with more severe disease. Finally, some areas of intermediate cell expansion were also associated with NFIB loss. Taken together, these results show a fundamental role for NFIB as a coregulator of AR action in the prostate and in controlling prostatic hyperplasia.
Collapse
Affiliation(s)
- Magdalena M Grabowska
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Stephen M Kelly
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Amy L Reese
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Justin M Cates
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Tom C Case
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Jianghong Zhang
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - David J DeGraff
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Douglas W Strand
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Nicole L Miller
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Peter E Clark
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Simon W Hayward
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Richard M Gronostajski
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Philip D Anderson
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| | - Robert J Matusik
- Department of Urologic Surgery (M.M.G., T.C.C., J.Z., N.L.M., P.E.C., S.W.H., R.J.M.), Department of Pathology, Microbiology, and Immunology (J.M.C.), and Vanderbilt-Ingram Cancer Center (P.E.C., R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Biological Sciences (S.M.K., A.L.R., P.D.A.), Salisbury University, Salisbury, Maryland 21801; Department of Pathology (D.J.G.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Urology (D.W.S.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Cancer Biology (S.W.H.), NorthShore HealthSystem Research Institute, Evanston, Illinois 60201; Department of Biochemistry, Genetics, Genomics and Bioinformatics Program (R.M.G.), University at Buffalo, Buffalo, New York 14203; and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37235
| |
Collapse
|
62
|
Gamat M, Chew KY, Shaw G, Renfree MB. FOXA1 and SOX9 Expression in the Developing Urogenital Sinus of the Tammar Wallaby (Macropus eugenii). Sex Dev 2015; 9:216-28. [PMID: 26406875 DOI: 10.1159/000439499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2015] [Indexed: 11/19/2022] Open
Abstract
The mammalian prostate is a compact structure in humans but multi-lobed in mice. In humans and mice, FOXA1 and SOX9 play pivotal roles in prostate morphogenesis, but few other species have been examined. We examined FOXA1 and SOX9 in the marsupial tammar wallaby, Macropus eugenii, which has a segmented prostate more similar to human than to mouse. In males, prostatic budding in the urogenital epithelium (UGE) was initiated by day 24 postpartum (pp), but in the female the UGE remained smooth and had begun forming the marsupial vaginal structures. FOXA1 was upregulated in the male urogenital sinus (UGS) by day 51 pp, whilst in the female UGS FOXA1 remained basal. FOXA1 was localised in the UGE in both sexes between day 20 and 80 pp. SOX9 was upregulated in the male UGS at day 21-30 pp and remained high until day 51-60 pp. SOX9 protein was localised in the distal tips of prostatic buds which were highly proliferative. The persistent upregulation of the transcription factors SOX9 and FOXA1 after the initial peak and fall of androgen levels suggest that in the tammar, as in other mammals, these factors are required to sustain prostate differentiation, development and proliferation as androgen levels return to basal levels.
Collapse
Affiliation(s)
- Melissa Gamat
- ARC Centre of Excellence in Kangaroo Genomics, Department of Zoology, The University of Melbourne, Melbourne, Vic., Australia
| | | | | | | |
Collapse
|
63
|
Abstract
FOXA1 (also known as hepatocyte nuclear factor 3α, or HNF-3α) is a protein of the FKHD family transcription factors. FOXA1 has been termed as a pioneer transcription factor due to its unique ability of chromatin remodeling in which the chromatin can be de-compacted to allow genomic access by nuclear hormone receptors, including androgen receptor (AR) and estrogen receptor (ER). In this review, we discuss our current understanding of FOXA1 regulation of prostatic and non-prostatic AR-chromatin targeting. We present an updated model wherein FOXA1:AR equilibrium in the nuclei defines prostatic AR binding profile, which is perturbed in prostate cancer with FOXA1 and/or AR de-regulation. Finally, we discuss recent efforts in exploring new horizons of AR-independent functions of FOXA1 in prostate cancer and interesting directions to pursue in future studies.
Collapse
Affiliation(s)
- Yeqing Angela Yang
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA ; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
64
|
Boberg J, Johansson HKL, Hadrup N, Dreisig K, Berthelsen L, Almstrup K, Vinggaard AM, Hass U. Perinatal exposure to mixtures of anti-androgenic chemicals causes proliferative lesions in rat prostate. Prostate 2015; 75:126-40. [PMID: 25327291 DOI: 10.1002/pros.22897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 08/21/2014] [Indexed: 11/07/2022]
Abstract
BACKGROUND Elevated levels of endogenous or exogenous estrogens during fetal life can induce permanent disturbances in prostate growth and predispose to precancerous lesions. Recent studies have indicated that also early anti-androgen exposure may affect prostate cancer risk. METHODS We examined the influence of perinatal exposure to mixtures of anti-androgenic and estrogenic chemicals on prostate development. Wistar rats were exposed from gestation day 7 to postnatal day 22 to a mixture of 8 anti-androgenic compounds (AAMix), a mixture of four estrogenic compounds (EMix), or paracetamol or a mixture of all 13 compounds (TotalMix) in mixture ratios reflecting human exposure levels. RESULTS Ventral prostate weights were reduced by the TotalMix and AAMix in pre-pubertal rats. Histological changes in prostate appeared with increasing age and indicated a shift from the normal age-dependent epithelial atrophy towards hyperplasia. These lesions showed similarities to pre-cancerous lesions in humans. Increased proliferation was observed already in pre-puberty and it was hypothesized that this could be associated with reduced ERβ signaling, but no clear conclusions could be made from gene expression studies on ERβ-related pathways. The influences of the estrogenic chemicals and paracetamol on prostate morphology were minor, but in young adulthood the estrogen mixture reduced ventral prostate mRNA levels of Igf1 and paracetamol reduced the mRNA level ofPbpc3. CONCLUSIONS Mixtures of endocrine disrupters relevant for human exposure was found to elicit persistent effects on the rat prostate following perinatal exposure, suggesting that human perinatal exposure to environmental chemicals may increase the risk of prostate cancer later in life.
Collapse
Affiliation(s)
- Julie Boberg
- Division of Toxicology and Risk Assessment, National Food Institute, Technical University of Denmark, Søborg, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
65
|
Hedgehog signaling in prostate epithelial-mesenchymal growth regulation. Dev Biol 2015; 400:94-104. [PMID: 25641695 DOI: 10.1016/j.ydbio.2015.01.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 12/24/2022]
Abstract
The prostate gland plays an important role in male reproduction, and is also an organ prone to diseases such as benign prostatic hyperplasia (BPH) and prostate cancer. The prostate consists of ducts with an inner layer of epithelium surrounded by stroma. Reciprocal signaling between these two cell compartments is instrumental to normal prostatic development, homeostasis, regeneration, as well as tumor formation. Hedgehog (HH) signaling is a master regulator in numerous developmental processes. In many organs, HH plays a key role in epithelial-mesenchymal signaling that regulates organ growth and tissue differentiation, and abnormal HH signaling has been implicated in the progression of various epithelial carcinomas. In this review, we focus on recent studies exploring the multipotency of endogenous postnatal and adult epithelial and stromal stem cells and studies addressing the role of HH in prostate development and cancer. We discuss the implications of the results for a new understanding of prostate development and disease. Insight into the cellular and molecular mechanisms underlying epithelial-mesenchymal growth regulation should provide a basis for devising innovative therapies to combat diseases of the prostate.
Collapse
|
66
|
Single luminal epithelial progenitors can generate prostate organoids in culture. Nat Cell Biol 2014; 16:951-61, 1-4. [PMID: 25241035 PMCID: PMC4183706 DOI: 10.1038/ncb3047] [Citation(s) in RCA: 262] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 09/01/2014] [Indexed: 12/22/2022]
Abstract
The intrinsic ability to display self-organizing morphogenetic properties in ex vivo culture may represent a general property of tissue stem cells. Here we show that single luminal stem/progenitor cells can generate prostate organoids in a three-dimensional culture system in the absence of stroma. Organoids generated from CARNs (castration-resistant Nkx3.1-expressing cells) or normal prostate epithelium exhibit tissue architecture containing luminal and basal cells, undergo long-term expansion in culture, and display functional androgen receptor signaling. Lineage-tracing demonstrates that luminal cells are favored for organoid formation, and generate basal cells in culture. Furthermore, tumor organoids can initiate from CARNs after oncogenic transformation, and from mouse models of prostate cancer, and can facilitate analyses of drug response. Finally, we provide evidence supporting the feasibility of organoid studies of human prostate tissue. Our studies underscore the progenitor properties of luminal cells, and identify in vitro approaches for studying prostate biology.
Collapse
|
67
|
Shtivelman E, Beer TM, Evans CP. Molecular pathways and targets in prostate cancer. Oncotarget 2014; 5:7217-59. [PMID: 25277175 PMCID: PMC4202120 DOI: 10.18632/oncotarget.2406] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/28/2014] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer co-opts a unique set of cellular pathways in its initiation and progression. The heterogeneity of prostate cancers is evident at earlier stages, and has led to rigorous efforts to stratify the localized prostate cancers, so that progression to advanced stages could be predicted based upon salient features of the early disease. The deregulated androgen receptor signaling is undeniably most important in the progression of the majority of prostate tumors. It is perhaps because of the primacy of the androgen receptor governed transcriptional program in prostate epithelium cells that once this program is corrupted, the consequences of the ensuing changes in activity are pleotropic and could contribute to malignancy in multiple ways. Following localized surgical and radiation therapies, 20-40% of patients will relapse and progress, and will be treated with androgen deprivation therapies. The successful development of the new agents that inhibit androgen signaling has changed the progression free survival in hormone resistant disease, but this has not changed the almost ubiquitous development of truly resistant phenotypes in advanced prostate cancer. This review summarizes the current understanding of the molecular pathways involved in localized and metastatic prostate cancer, with an emphasis on the clinical implications of the new knowledge.
Collapse
Affiliation(s)
| | - Tomasz M. Beer
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Christopher P. Evans
- Department of Urology and Comprehensive Cancer Center, University of California Davis, Davis, CA
| |
Collapse
|
68
|
Wang Y, Zhang Y, Wang H, Wang J, Zhang Y, Wang Y, Pan Z, Luo S. Aberrantly up-regulated miR-20a in pre-eclampsic placenta compromised the proliferative and invasive behaviors of trophoblast cells by targeting forkhead box protein A1. Int J Biol Sci 2014; 10:973-82. [PMID: 25210495 PMCID: PMC4159688 DOI: 10.7150/ijbs.9088] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 08/01/2014] [Indexed: 11/05/2022] Open
Abstract
Preeclampsia is a serious complication in pregnancy. Dysregulation of trophoblast cell proliferation and invasion is a major pathological alteration observed in preeclampsia. Recently, microRNAs were shown to participate in the pathogenesis of preeclampsia. In this study we explored the effect of miR-20a on the proliferation and invasion of trophoblast cells and the underlying mechanism. We verified the distribution of miR-20a in human placenta by in situ hybridization. Real time PCR data showed that the level of miR-20a increased by 2.6 folds in human preeclampsia than normal tissues. We then cultured trophoblast-like JEG-3 cells and evaluated the effect of miR-20a on JEG-3 cell proliferation, migration and invasion. Overexpression of miR-20a significantly inhibited the proliferation, migration and invasion of cultured JEG-3 cells, which were abolished by co-transfection of AMO-20a. Transfection of miR-20a also inhibited JEG-3 cell xenograft tumor growth in nude mice. Luciferase assay technique was used to identify the direct regulation of miR-20a on Forkhead Box Protein A1(FOXA1). Transfection of miR-20a markedly reduced the luciferase activity of the chimeric plasmid containing the 3'UTR of FOXA1, indicating FOXA1 is the target of miR-20a. Furthermore, transfection of miR-20a inhibited both protein and mRNA expression of FOXA1 in JEG-3 cells. In summary, the upregulated miR-20a in human preeclampsia tissue can inhibit the proliferative and invasive activities of trophoblast cells by repressing the expression of FOXA1.
Collapse
Affiliation(s)
- Ying Wang
- 2. Department of Pharmacology (Key Laboratory of Cardiovascular Medicine Research, Ministry of Education; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang 150081, P. R. China
| | - Yang Zhang
- 2. Department of Pharmacology (Key Laboratory of Cardiovascular Medicine Research, Ministry of Education; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang 150081, P. R. China
| | - Huimin Wang
- 2. Department of Pharmacology (Key Laboratory of Cardiovascular Medicine Research, Ministry of Education; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang 150081, P. R. China
| | - Junxiao Wang
- 1. Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P. R. China
| | - Yiyuan Zhang
- 2. Department of Pharmacology (Key Laboratory of Cardiovascular Medicine Research, Ministry of Education; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang 150081, P. R. China
| | - Yingzhe Wang
- 2. Department of Pharmacology (Key Laboratory of Cardiovascular Medicine Research, Ministry of Education; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang 150081, P. R. China
| | - Zhenwei Pan
- 2. Department of Pharmacology (Key Laboratory of Cardiovascular Medicine Research, Ministry of Education; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, Heilongjiang 150081, P. R. China
| | - Shanshun Luo
- 1. Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P. R. China
| |
Collapse
|
69
|
Stromal androgen receptor in prostate development and cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2598-607. [PMID: 25088980 DOI: 10.1016/j.ajpath.2014.06.022] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/18/2014] [Accepted: 06/26/2014] [Indexed: 11/22/2022]
Abstract
The androgen receptor (AR) in stromal cells contributes significantly to the development and growth of prostate during fetal stages as well as during prostate carcinogenesis and cancer progression. During prostate development, stromal AR induces and promotes epithelial cell growth, as observed from tissue recombinant and mouse knockout studies. During prostate carcinogenesis and progression, the stromal cells begin to lose AR expression as early as at the stage of high-grade prostatic intraepithelial neoplasia. The extent of loss of stromal AR is directly proportional to the degree of differentiation (Gleason grade) and progression of prostate cancer (PCa). Co-culture studies suggested that stromal AR inhibits the growth of malignant epithelial cells, possibly through expression of certain paracrine factors in the presence of androgens. This functional reversal of stromal AR, from growth promotion during fetal prostate development to mediating certain growth-inhibiting effects in cancer, explains to some extent the reason that loss of AR expression in stromal cells may be crucial for development of resistance to androgen ablation therapy for PCa. From a translational perspective, it generates the need to re-examine the current therapeutic options and opens a fundamental new direction for therapeutic interventions, especially in advanced PCa.
Collapse
|
70
|
Androgen receptor and its splice variant, AR-V7, differentially regulate FOXA1 sensitive genes in LNCaP prostate cancer cells. Int J Biochem Cell Biol 2014; 54:49-59. [PMID: 25008967 DOI: 10.1016/j.biocel.2014.06.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 05/25/2014] [Accepted: 06/18/2014] [Indexed: 11/23/2022]
Abstract
Prostate cancer (PCa) is an androgen-dependent disease, and tumors that are resistant to androgen ablation therapy often remain androgen receptor (AR) dependent. Among the contributors to castration-resistant PCa are AR splice variants that lack the ligand-binding domain (LBD). Instead, they have small amounts of unique sequence derived from cryptic exons or from out of frame translation. The AR-V7 (or AR3) variant is constitutively active and is expressed under conditions consistent with CRPC. AR-V7 is reported to regulate a transcriptional program that is similar but not identical to that of AR. However, it is unknown whether these differences are due to the unique sequence in AR-V7, or simply to loss of the LBD. To examine transcriptional regulation by AR-V7, we have used lentiviruses encoding AR-V7 (amino acids 1-627 of AR with the 16 amino acids unique to the variant) to prepare a derivative of the androgen-dependent LNCaP cells with inducible expression of AR-V7. An additional cell line was generated with regulated expression of AR-NTD (amino acids 1-660 of AR); this mutant lacks the LBD but does not have the AR-V7 specific sequence. We find that AR and AR-V7 have distinct activities on target genes that are co-regulated by FOXA1. Transcripts regulated by AR-V7 were similarly regulated by AR-NTD, indicating that loss of the LBD is sufficient for the observed differences. Differential regulation of target genes correlates with preferential recruitment of AR or AR-V7 to specific cis-regulatory DNA sequences providing an explanation for some of the observed differences in target gene regulation.
Collapse
|
71
|
Omori A, Miyagawa S, Ogino Y, Harada M, Ishii K, Sugimura Y, Ogino H, Nakagata N, Yamada G. Essential roles of epithelial bone morphogenetic protein signaling during prostatic development. Endocrinology 2014; 155:2534-44. [PMID: 24731097 PMCID: PMC4060178 DOI: 10.1210/en.2013-2054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Prostate is a male sex-accessory organ. The prostatic epithelia consist primarily of basal and luminal cells that differentiate from embryonic urogenital sinus epithelia. Prostate tumors are believed to originate in the basal and luminal cells. However, factors that promote normal epithelial differentiation have not been well elucidated, particularly for bone morphogenetic protein (Bmp) signaling. This study shows that Bmp signaling prominently increases during prostatic differentiation in the luminal epithelia, which is monitored by the expression of phosphorylated Smad1/5/8. To elucidate the mechanism of epithelial differentiation and the function of Bmp signaling during prostatic development, conditional male mutant mouse analysis for the epithelial-specific Bmp receptor 1a (Bmpr1a) was performed. We demonstrate that Bmp signaling is indispensable for luminal cell maturation, which regulates basal cell proliferation. Expression of the prostatic epithelial regulatory gene Nkx3.1 was significantly reduced in the Bmpr1a mutants. These results indicate that Bmp signaling is a key factor for prostatic epithelial differentiation, possibly by controlling the prostatic regulatory gene Nkx3.1.
Collapse
MESH Headings
- Animals
- Bone Morphogenetic Protein Receptors, Type I/genetics
- Bone Morphogenetic Protein Receptors, Type I/metabolism
- Cell Differentiation/genetics
- Cell Line, Tumor
- Cell Proliferation
- Epithelium/metabolism
- Epithelium/pathology
- Female
- Fluorescent Antibody Technique
- Gene Expression Regulation, Developmental
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Hyperplasia
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred ICR
- Mice, Knockout
- Mice, Transgenic
- Mutation
- Phosphorylation
- Prostate/metabolism
- Prostate/pathology
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/genetics
- Smad Proteins/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Akiko Omori
- Department of Developmental Genetics (A.O., G.Y.), Institute of Advanced Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan; Okazaki Institute for Integrative Bioscience (S.M., Y.O.), National Institute for Basic Biology, National Institutes of Natural Science, Okazaki, 444-8787, Japan; Department of Clinical Anatomy (M.H.), Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8591, Japan; Department of Oncologic Pathology (K.I.), and Nephro-Urologic Surgery and Andrology (Y.S.), Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan; Department of Animal Bioscience (H.O.), Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, 526-0829, Japan; and Division of Reproductive Engineering (N.N.), Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
DeGraff DJ, Grabowska MM, Case T, Yu X, Herrick MK, Hayward W, Strand DW, Cates JM, Hayward SW, Gao N, Walter MA, Buttyan R, Yi Y, Kaestner KH, Matusik RJ. FOXA1 deletion in luminal epithelium causes prostatic hyperplasia and alteration of differentiated phenotype. J Transl Med 2014; 94:726-39. [PMID: 24840332 PMCID: PMC4451837 DOI: 10.1038/labinvest.2014.64] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 02/20/2014] [Accepted: 03/26/2014] [Indexed: 01/25/2023] Open
Abstract
The forkhead box (Fox) superfamily of transcription factors has essential roles in organogenesis and tissue differentiation. Foxa1 and Foxa2 are expressed during prostate budding and ductal morphogenesis, whereas Foxa1 expression is retained in adult prostate epithelium. Previous characterization of prostatic tissue rescued from embryonic Foxa1 knockout mice revealed Foxa1 to be essential for ductal morphogenesis and epithelial maturation. However, it is unknown whether Foxa1 is required to maintain the differentiated status in adult prostate epithelium. Here, we employed the PBCre4 transgenic system and determined the impact of prostate-specific Foxa1 deletion in adult murine epithelium. PBCre4/Foxa1(loxp/loxp) mouse prostates showed progressive florid hyperplasia with extensive cribriform patterning, with the anterior prostate being most affected. Immunohistochemistry studies show mosaic Foxa1 KO consistent with PBCre4 activity, with Foxa1 KO epithelial cells specifically exhibiting altered cell morphology, increased proliferation, and elevated expression of basal cell markers. Castration studies showed that, while PBCre4/Foxa1(loxp/loxp) prostates did not exhibit altered sensitivity in response to hormone ablation compared with control prostates, the number of Foxa1-positive cells in mosaic Foxa1 KO prostates was significantly reduced compared with Foxa1-negative cells following castration. Unexpectedly, gene expression profile analyses revealed that Foxa1 deletion caused abnormal expression of seminal vesicle-associated genes in KO prostates. In summary, these results indicate Foxa1 expression is required for the maintenance of prostatic cellular differentiation.
Collapse
Affiliation(s)
- David J. DeGraff
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | | | - Tom Case
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Xiuping Yu
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Mary K. Herrick
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - William Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Douglas W. Strand
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Justin M. Cates
- Department of Pathology, Vanderbilt University Medical Center, Nashville TN
| | - Simon W. Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark NJ
| | | | | | - Yajun Yi
- Institute for Integrative Genomics and Department of Medicine, Vanderbilt University, Nashville TN
| | | | - Robert J. Matusik
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN,Department of Cell and Developmental Biology, Vanderbilt University, Nashville TN,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville TN,Correspondence and reprint requests should be made to: Robert J. Matusik, Ph.D., William L. Bray Chair of Urologic Surgery, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232,
| |
Collapse
|
73
|
Jin HJ, Zhao JC, Wu L, Kim J, Yu J. Cooperativity and equilibrium with FOXA1 define the androgen receptor transcriptional program. Nat Commun 2014; 5:3972. [PMID: 24875621 PMCID: PMC4088269 DOI: 10.1038/ncomms4972] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 04/25/2014] [Indexed: 12/31/2022] Open
Abstract
The pioneering factor FOXA1 opens chromatin to facilitate androgen receptor (AR) binding to prostate-specific genes. How FOXA1 controls the AR cistrome, however, is incompletely understood. Here we show that AR directly binds chromatin through the androgen response elements (AREs). FOXA1 is not required for AR-chromatin interaction, but instrumental in recruiting AR to low-affinity half-AREs by opening local chromatin around adjacent FKHD sites. Too much FOXA1 creates excessive open chromatin regions, which serve as reservoirs that retain AR via abundant half-AREs, thereby reducing its availability for specific sites. FOXA1 downregulation, by contrast, relinquishes AR to permissively bind AREs across the genome, resulting in substantial AR-binding events and AR target gene expression even in the absence of androgen. Taken together, our data illustrate the mechanistic details by which cooperativity and equilibrium with FOXA1 define AR cistrome and reveal a previously unknown function of FOXA1 in inhibiting AR signalling and castration-resistant prostate cancer growth.
Collapse
Affiliation(s)
- Hong-Jian Jin
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Jonathan C Zhao
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Longtao Wu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Jung Kim
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Jindan Yu
- 1] Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA [2] Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
74
|
Grabowska MM, Elliott AD, DeGraff DJ, Anderson PD, Anumanthan G, Yamashita H, Sun Q, Friedman DB, Hachey DL, Yu X, Sheehan JH, Ahn JM, Raj GV, Piston DW, Gronostajski RM, Matusik RJ. NFI transcription factors interact with FOXA1 to regulate prostate-specific gene expression. Mol Endocrinol 2014; 28:949-64. [PMID: 24801505 DOI: 10.1210/me.2013-1213] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Androgen receptor (AR) action throughout prostate development and in maintenance of the prostatic epithelium is partly controlled by interactions between AR and forkhead box (FOX) transcription factors, particularly FOXA1. We sought to identity additional FOXA1 binding partners that may mediate prostate-specific gene expression. Here we identify the nuclear factor I (NFI) family of transcription factors as novel FOXA1 binding proteins. All four family members (NFIA, NFIB, NFIC, and NFIX) can interact with FOXA1, and knockdown studies in androgen-dependent LNCaP cells determined that modulating expression of NFI family members results in changes in AR target gene expression. This effect is probably mediated by binding of NFI family members to AR target gene promoters, because chromatin immunoprecipitation (ChIP) studies found that NFIB bound to the prostate-specific antigen enhancer. Förster resonance energy transfer studies revealed that FOXA1 is capable of bringing AR and NFIX into proximity, indicating that FOXA1 facilitates the AR and NFI interaction by bridging the complex. To determine the extent to which NFI family members regulate AR/FOXA1 target genes, motif analysis of publicly available data for ChIP followed by sequencing was undertaken. This analysis revealed that 34.4% of peaks bound by AR and FOXA1 contain NFI binding sites. Validation of 8 of these peaks by ChIP revealed that NFI family members can bind 6 of these predicted genomic elements, and 4 of the 8 associated genes undergo gene expression changes as a result of individual NFI knockdown. These observations suggest that NFI regulation of FOXA1/AR action is a frequent event, with individual family members playing distinct roles in AR target gene expression.
Collapse
Affiliation(s)
- Magdalena M Grabowska
- Department of Urologic Surgery (M.M.G., G.A. H.Y., Q.S., X.Y., R.J.M.), Department of Molecular Physiology and Biophysics (A.D.E., D.W.P.), and Vanderbilt-Ingram Cancer Center (R.J.M.), Vanderbilt University Medical Center, Nashville, Tennessee 37232; Department of Pathology (D.J.D.), Penn State University College of Medicine, Hershey, Pennsylvania 17033; Department of Biological Sciences (P.D.A.), Salisbury University, Salisbury, Maryland 21801; Mass Spectrometry Research Center (D.B.F., D.L.H.), Department of Biochemistry, Department of Biochemistry and Center for Structural Biology (J.H.S.), and Department of Cell and Developmental Biology (R.J.M.), Vanderbilt University, Nashville, Tennessee 37232; Department of Chemistry (J.-M.A.), University of Texas Dallas, Dallas, Texas 75080; Department of Urology (G.V.R.), University of Texas Southwestern, Dallas, Texas 75390; and Department of Biochemistry (R.M.G.), Developmental Genomics Group, NY State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, New York 14203
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Pihlajamaa P, Sahu B, Lyly L, Aittomäki V, Hautaniemi S, Jänne OA. Tissue-specific pioneer factors associate with androgen receptor cistromes and transcription programs. EMBO J 2014; 33:312-26. [PMID: 24451200 DOI: 10.1002/embj.201385895] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Androgen receptor (AR) binds male sex steroids and mediates physiological androgen actions in target tissues. ChIP-seq analyses of AR-binding events in murine prostate, kidney and epididymis show that in vivo AR cistromes and their respective androgen-dependent transcription programs are highly tissue specific mediating distinct biological pathways. This high order of tissue specificity is achieved by the use of exclusive collaborating factors in the three androgen-responsive tissues. We find two novel collaborating factors for AR signaling in vivo--Hnf4α (hepatocyte nuclear factor 4α) in mouse kidney and AP-2α (activating enhancer binding protein 2α) in mouse epididymis--that define tissue-specific AR recruitment. In mouse prostate, FoxA1 serves for the same purpose. FoxA1, Hnf4α and AP-2α motifs are over-represented within unique AR-binding loci, and the cistromes of these factors show substantial overlap with AR-binding events distinct to each tissue type. These licensing or pioneering factors are constitutively bound to chromatin and guide AR to specific genomic loci upon hormone exposure. Collectively, liganded receptor and its DNA-response elements are required but not sufficient for establishment of tissue-specific transcription programs.
Collapse
Affiliation(s)
- Päivi Pihlajamaa
- Institute of Biomedicine University of Helsinki, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
76
|
Wu D, Sunkel B, Chen Z, Liu X, Ye Z, Li Q, Grenade C, Ke J, Zhang C, Chen H, Nephew KP, Huang THM, Liu Z, Jin VX, Wang Q. Three-tiered role of the pioneer factor GATA2 in promoting androgen-dependent gene expression in prostate cancer. Nucleic Acids Res 2014; 42:3607-22. [PMID: 24423874 PMCID: PMC3973339 DOI: 10.1093/nar/gkt1382] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In prostate cancer, androgen receptor (AR) binding and androgen-responsive gene expression are defined by hormone-independent binding patterns of the pioneer factors FoxA1 and GATA2. Insufficient evidence of the mechanisms by which GATA2 contributes to this process precludes complete understanding of a key determinant of tissue-specific AR activity. Our observations suggest that GATA2 facilitates androgen-responsive gene expression by three distinct modes of action. By occupying novel binding sites within the AR gene locus, GATA2 positively regulates AR expression before and after androgen stimulation. Additionally, GATA2 engages AR target gene enhancers prior to hormone stimulation, producing an active and accessible chromatin environment via recruitment of the histone acetyltransferase p300. Finally, GATA2 functions in establishing and/or sustaining basal locus looping by recruiting the Mediator subunit MED1 in the absence of androgen. These mechanisms may contribute to the generally positive role of GATA2 in defining AR genome-wide binding patterns that determine androgen-responsive gene expression profiles. We also find that GATA2 and FoxA1 exhibit both independent and codependent co-occupancy of AR target gene enhancers. Identifying these determinants of AR transcriptional activity may provide a foundation for the development of future prostate cancer therapeutics that target pioneer factor function.
Collapse
Affiliation(s)
- Dayong Wu
- Department of Molecular and Cellular Biochemistry and the Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH 43210, USA, Ohio State Biochemistry Graduate Program, The Ohio State University, Columbus, OH 43210, USA, Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA, State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China and Medical Sciences Program and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Robinson JLL, Hickey TE, Warren AY, Vowler SL, Carroll T, Lamb AD, Papoutsoglou N, Neal DE, Tilley WD, Carroll JS. Elevated levels of FOXA1 facilitate androgen receptor chromatin binding resulting in a CRPC-like phenotype. Oncogene 2013; 33:5666-74. [PMID: 24292680 PMCID: PMC4051595 DOI: 10.1038/onc.2013.508] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/18/2013] [Accepted: 10/23/2013] [Indexed: 12/18/2022]
Abstract
Castration-resistant prostate cancer (CRPC) continues to pose a significant clinical challenge with new generation second line hormonal therapies affording limited improvement in disease outcome. As the androgen receptor (AR) remains a critical driver in CRPC, understanding the determinants of its transcriptional activity is important for developing new AR targeted therapies. FOXA1 is a key component of the AR transcriptional complex yet its role in prostate cancer progression and the relationship between AR and FOXA1 are not completely resolved. It is well established that FOXA1 levels are elevated in advanced prostate cancer and metastases. We mimicked these conditions by over-expressing FOXA1 in the androgen-responsive LNCaP prostate cancer cell line and observed a significant increase in AR genomic binding at novel regions that possess increased chromatin accessibility. High levels of FOXA1 resulted in increased proliferation at both sub-optimal and high 5α-dihydrotestosterone (DHT) concentrations. Immunohistochemical staining for FOXA1 in a clinical prostate cancer cohort revealed that high FOXA1 expression is associated with shorter time to biochemical recurrence after radical prostatectomy (HR 5.0, 95% CI 1.2-21.1, p=0.028), positive surgical margins and higher stage disease at diagnosis. The gene expression program that results from FOXA1 over-expression is enriched for PTEN, Wnt and other pathways typically represented in CRPC gene signatures. Together these results suggest that in an androgen-depleted state, elevated levels of FOXA1 enhance AR binding at genomic regions not normally occupied by AR, which in turn facilitates prostate cancer cell growth.
Collapse
Affiliation(s)
- J L L Robinson
- 1] Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Department of Oncology, University of Cambridge, Cambridge, UK
| | - T E Hickey
- Dame Roma Mitchell Cancer Research Laboratories and the Adelaide Prostate Cancer Research Centre, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - A Y Warren
- Department of Histopathology, Cambridge University Hospitals NHS Foundations Trust, Cambridge, UK
| | - S L Vowler
- 1] Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Department of Oncology, University of Cambridge, Cambridge, UK
| | - T Carroll
- 1] Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Department of Oncology, University of Cambridge, Cambridge, UK
| | - A D Lamb
- 1] Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Department of Oncology, University of Cambridge, Cambridge, UK [3] Department of Urology, Cambridge University Hospitals NHS Foundations Trust, Cambridge, UK
| | - N Papoutsoglou
- Department of Urology, Cambridge University Hospitals NHS Foundations Trust, Cambridge, UK
| | - D E Neal
- 1] Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Department of Oncology, University of Cambridge, Cambridge, UK [3] Dame Roma Mitchell Cancer Research Laboratories and the Adelaide Prostate Cancer Research Centre, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - W D Tilley
- Dame Roma Mitchell Cancer Research Laboratories and the Adelaide Prostate Cancer Research Centre, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - J S Carroll
- 1] Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK [2] Department of Oncology, University of Cambridge, Cambridge, UK
| |
Collapse
|
78
|
Kruithof-de Julio M, Shibata M, Desai N, Reynon M, Halili MV, Hu YP, Price SM, Abate-Shen C, Shen MM. Canonical Wnt signaling regulates Nkx3.1 expression and luminal epithelial differentiation during prostate organogenesis. Dev Dyn 2013; 242:1160-71. [PMID: 23813564 DOI: 10.1002/dvdy.24008] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 06/03/2013] [Accepted: 06/20/2013] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The formation of the prostate gland requires reciprocal interactions between the epithelial and mesenchymal components of the embryonic urogenital sinus. However, the identity of the signaling factors that mediate these interactions is largely unknown. RESULTS Our studies show that expression of the prostate-specific transcription factor Nkx3.1 is regulated by the canonical Wnt signaling pathway. Using mice carrying a targeted lacZ knock-in allele of Nkx3.1, we find that Nkx3.1 is expressed in all epithelial cells of ductal buds during prostate organogenesis. Addition of Wnt inhibitors to urogenital sinus explant culture greatly reduces prostate budding and inhibits Nkx3.1 expression as well as differentiation of luminal epithelial cells. Analyses of a TCF/Lef:H2B-GFP transgene reporter show that canonical Wnt signaling activity is found in urogenital mesenchyme but not urogenital sinus epithelium before prostate formation, and is later observed in the mesenchyme and epithelium of prostate ductal tips. Furthermore, TCF/Lef:H2B-GFP reporter activity is reduced in epithelial cells of Nkx3.1 null neonatal prostates, suggesting that Nkx3.1 functions to maintain canonical Wnt signaling activity in developing prostate bud tips. CONCLUSIONS We propose that activated canonical Wnt signals and Nkx3.1 function in a positive feedback loop to regulate prostate bud growth and luminal epithelial differentiation.
Collapse
Affiliation(s)
- Marianna Kruithof-de Julio
- Departments of Medicine and Genetics and Development, Columbia University Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Schoenborn JR, Nelson P, Fang M. Genomic profiling defines subtypes of prostate cancer with the potential for therapeutic stratification. Clin Cancer Res 2013; 19:4058-66. [PMID: 23704282 DOI: 10.1158/1078-0432.ccr-12-3606] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The remarkable variation in prostate cancer clinical behavior represents an opportunity to identify and understand molecular features that can be used to stratify patients into clinical subgroups for more precise outcome prediction and treatment selection. Significant progress has been made in recent years in establishing the composition of genomic and epigenetic alterations in localized and advanced prostate cancers using array-based technologies and next-generation sequencing approaches. The results of these efforts shed new light on our understanding of this disease and point to subclasses of prostate cancer that exhibit distinct vulnerabilities to therapeutics. The goal of this review is to categorize the genomic data and, where available, corresponding expression, functional, or related therapeutic information, from recent large-scale and in-depth studies that show a new appreciation for the molecular complexity of this disease. We focus on how these results inform our growing understanding of the mechanisms that promote genetic instability, as well as routes by which specific genes and biologic pathways may serve as biomarkers or potential targets for new therapies. We summarize data that indicate the presence of genetic subgroups of prostate cancers and show the high level of intra- and intertumoral heterogeneity, as well as updated information on disseminated and circulating tumor cells. The integrated analysis of all types of genetic alterations that culminate in altering critical biologic pathways may serve as the impetus for developing new therapeutics, repurposing agents used currently for treating other malignancies, and stratifying early and advanced prostate cancers for appropriate interventions.
Collapse
Affiliation(s)
- Jamie R Schoenborn
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | |
Collapse
|
80
|
Gupta A, Yu X, Case T, Paul M, Shen MM, Kaestner KH, Matusik RJ. Mash1 expression is induced in neuroendocrine prostate cancer upon the loss of Foxa2. Prostate 2013; 73:582-9. [PMID: 23060003 PMCID: PMC3714015 DOI: 10.1002/pros.22598] [Citation(s) in RCA: 10] [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: 08/17/2012] [Accepted: 09/10/2012] [Indexed: 01/04/2023]
Abstract
Neuroendocrine (NE) prostate tumors and neuroendocrine differentiation (NED) in prostatic adenocarcinomas have been associated with poor prognosis. In this study, we used the TRAMP mouse model that develops NE prostate tumors to identify key factors that can lead to NED. We have previously reported that NE tumors express the forkhead transcription factor, Foxa2, Mash1 (mouse achaete scute homolog-1), as well as Synaptophysin. In TRAMP, the prostatic intraepithelial neoplasia (PIN) first expresses Foxa2 and Synaptophysin, which then progresses to NE cancer. In order to determine if Foxa2 is dispensable for development or maintenance of NE cancer, a conditional knock-out of Foxa2 in TRAMP mice was generated by breeding mice with two floxed alleles of Foxa2 and one copy of Nkx3.1-Cre. Nkx3.1-Cre/Foxa2(loxP/loxP) mice showed loss of Foxa2 expression in embryonic prostatic buds. No expression of Foxa2 was seen in the adult prostate in either conditional null or control mice. Foxa2 is universally expressed in all wild type TRAMP NE tumors, but Mash1 expression is seen only in a few samples in a few cells. With the loss of Foxa2 in the NE tumors of the TRAMP/Nkx3.1-Cre/Foxa2(loxP/loxP) mice, the expression of the pro-neuronal gene Mash1 is upregulated. NE tumors from both the TRAMP control and Foxa2-deficient TRAMP prostate express Synaptophysin and SV40 Large T-antigen, and both show a loss of androgen receptor expression in NE cells. These studies suggest that the TRAMP NE tumors can form in the absence of Foxa2 by an up regulation of Mash1.
Collapse
Affiliation(s)
- Aparna Gupta
- Division of Gastroenterology & Hepatology, Department of Medicine, Stanford University, Stanford, California
| | - Xiuping Yu
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tom Case
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Manik Paul
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael M. Shen
- Departments of Medicine and Genetics & Development, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Klaus H. Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert J. Matusik
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Correspondence to: Robert J. Matusik, Department of Urologic Surgery, Vanderbilt University School of Medicine, Nashville, TN 37232.
| |
Collapse
|
81
|
Reduced androgen receptor expression accelerates the onset of ERBB2 induced breast tumors in female mice. PLoS One 2013; 8:e60455. [PMID: 23593223 PMCID: PMC3620158 DOI: 10.1371/journal.pone.0060455] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/26/2013] [Indexed: 11/21/2022] Open
Abstract
Androgen receptor (AR) is commonly expressed in both the epithelium of normal mammary glands and in breast cancers. AR expression in breast cancers is independent of estrogen receptor alpha (ERα) status and is frequently associated with overexpression of the ERBB2 oncogene. AR signaling effects on breast cancer progression may depend on ERα and ERBB2 status. Up to 30% of human breast cancers are driven by overactive ERBB2 signaling and it is not clear whether AR expression affects any steps of tumor progression in this cohort of patients. To test this, we generated mammary specific Ar depleted mice (MARKO) by combining the floxed allele of Ar with the MMTV-cre transgene on an MMTV-NeuNT background and compared them to littermate MMTV-NeuNT, Arfl/+ control females. Heterozygous MARKO females displayed reduced levels of AR in mammary glands with mosaic AR expression in ductal epithelium. The loss of AR dramatically accelerated the onset of MMTV-NeuNT tumors in female MARKO mice. In this report we show that accelerated MMTV-NeuNT-dependent tumorigenesis is due specifically to the loss of AR, as hormonal levels, estrogen and progesterone receptors expression, and MMTV-NeuNT expression were similar between MARKO and control groups. MMTV-NeuNT induced tumors in both cohorts displayed distinct loss of AR in addition to ERα, PR, and the pioneer factor FOXA1. Erbb3 mRNA levels were significantly elevated in tumors in comparison to normal mammary glands. Thus the loss of AR in mouse mammary epithelium accelerates malignant transformation rather than the rate of tumorigenesis.
Collapse
|
82
|
Duan Z, Zou JX, Yang P, Wang Y, Borowsky AD, Gao AC, Chen HW. Developmental and androgenic regulation of chromatin regulators EZH2 and ANCCA/ATAD2 in the prostate Via MLL histone methylase complex. Prostate 2013; 73:455-66. [PMID: 23038103 DOI: 10.1002/pros.22587] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 08/16/2012] [Indexed: 02/03/2023]
Abstract
BACKGROUND Chromatin regulators ANCCA and EZH2 are overexpressed in prostate cancer and play crucial roles in androgen-stimulated and castration-refractory prostate tumor growth and survival. However, how their expression is regulated in the tumors and whether they play a role in prostate development remains unclear. METHODS Prostate tissue from different developmental stages of mouse and human were examined by IHC, qRT-PCR and Western for expression of ANCCA, EZH2, and Ki-67. Animals were castrated and T-implanted for the expression response in normal prostate and tumors. siRNA knockdown and ChIP were performed for the mechanism of ANCCA regulation of EZH2. RESULTS In contrast to their very low level expression in adult prostate, ANCCA and EZH2 are strongly expressed in the epithelium and mesenchyme of mouse and human UGS. Their expression becomes more restricted to epithelial cells during later development and displays a second peak during puberty, which correlates with the proliferative status of the epithelium. Importantly, their expression in normal prostate and tumors is strongly suppressed by castration and markedly induced by testosterone replacement. While androgen suppresses EZH2 in CRPC cells, in LNCaP cells, physiological concentrations of androgen stimulate expression of PRC2 genes (EZH2, SUZ12, and EED), which is mediated by androgen-induced ANCCA and involves E2F and histone H3K4me3 methylase MLL1 complex. CONCLUSION EZH2 and ANCCA are androgen regulated and strongly expressed in early prostate morphogenesis and during puberty, suggesting their important role in prostate development. Regulation of EZH2 by ANCCA emphasizes bromodomain protein ANCCA as a potential therapeutic target against prostate cancer.
Collapse
Affiliation(s)
- Zhijian Duan
- Cancer Center/Basic Sciences, University of California at Davis, Sacramento, California 95817, USA
| | | | | | | | | | | | | |
Collapse
|
83
|
Jin HJ, Zhao JC, Ogden I, Bergan RC, Yu J. Androgen receptor-independent function of FoxA1 in prostate cancer metastasis. Cancer Res 2013; 73:3725-36. [PMID: 23539448 DOI: 10.1158/0008-5472.can-12-3468] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
FoxA1 (FOXA1) is a pioneering transcription factor of the androgen receptor (AR) that is indispensible for the lineage-specific gene expression of the prostate. To date, there have been conflicting reports on the role of FoxA1 in prostate cancer progression and prognosis. With recent discoveries of recurrent FoxA1 mutations in human prostate tumors, comprehensive understanding of FoxA1 function has become very important. Here, through genomic analysis, we reveal that FoxA1 regulates two distinct oncogenic processes via disparate mechanisms. FoxA1 induces cell growth requiring the AR pathway. On the other hand, FoxA1 inhibits cell motility and epithelial-to-mesenchymal transition (EMT) through AR-independent mechanism directly opposing the action of AR signaling. Using orthotopic mouse models, we further show that FoxA1 inhibits prostate tumor metastasis in vivo. Concordant with these contradictory effects on tumor progression, FoxA1 expression is slightly upregulated in localized prostate cancer wherein cell proliferation is the main feature, but is remarkably downregulated when the disease progresses to metastatic stage for which cell motility and EMT are essential. Importantly, recently identified FoxA1 mutants have drastically attenuated ability in suppressing cell motility. Taken together, our findings illustrate an AR-independent function of FoxA1 as a metastasis inhibitor and provide a mechanism by which recurrent FoxA1 mutations contribute to prostate cancer progression.
Collapse
Affiliation(s)
- Hong-Jian Jin
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
84
|
Montani M, Hermanns T, Müntener M, Wild P, Sulser T, Kristiansen G. Multidrug resistance protein 4 (MRP4) expression in prostate cancer is associated with androgen signaling and decreases with tumor progression. Virchows Arch 2013; 462:437-43. [PMID: 23503867 DOI: 10.1007/s00428-013-1390-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/12/2013] [Accepted: 02/25/2013] [Indexed: 12/24/2022]
Abstract
Multidrug resistance protein 4 (MRP4) is a transmembrane transport protein found in many cell types and is involved in substrate-specific transport of endogenous and exogenous substrates. Recently, it has shown to be expressed in prostate cancer cell lines and to be among the most commonly upregulated transcripts in prostate cancer, although a comprehensive expression analysis is lacking so far. We aimed to investigate its expression by immunohistochemistry in a larger cohort of neoplastic and nonneoplastic prostate tissues (n = 441) and to correlate its expression with clinicopathological parameters including PSA-free survival times and molecular correlates of androgen signaling (androgen receptor (AR), prostate-specific antigen (PSA), and forkhead box A (FoxA)). MRP4 is widely expressed in benign and neoplastic prostate epithelia, but its expression gradually decreases during tumor progression towards castrate-resistant disease. Concordantly, it correlated with conventional prognosticators of disease progression and-within the group of androgen-dependent tumors-with AR and FoxA expression. Moreover, lower levels of MRP4 expression were associated with shorter PSA relapse-free survival times in the androgen-dependent group. In benign tissues, we found zone-dependent differences of MRP4 expression, with the highest levels in the peripheral and central zones. Although MRP4 is known to be regulated in prostate cancer, this study is the first to demonstrate a gradual downregulation of MRP4 protein during malignant tumor progression and a prognostic value of this loss of expression.
Collapse
Affiliation(s)
- Matteo Montani
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
85
|
Robinson JLL, Holmes KA, Carroll JS. FOXA1 mutations in hormone-dependent cancers. Front Oncol 2013; 3:20. [PMID: 23420418 PMCID: PMC3572741 DOI: 10.3389/fonc.2013.00020] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 01/25/2013] [Indexed: 01/04/2023] Open
Abstract
The forkhead protein, FOXA1, is a critical interacting partner of the nuclear hormone receptors, oestrogen receptor-α (ER) and androgen receptor (AR), which are major drivers of the two most common cancers, namely breast and prostate cancer. Over the past few years, progress has been made in our understanding of how FOXA1 influences nuclear receptor function, with both common and distinct roles in the regulation of ER or AR. Recently, another level of regulation has been described, with the discovery that FOXA1 is mutated in 1.8% of breast and 3–5% prostate cancers. In addition, a subset of both cancer types exhibit amplification of the genomic region encompassing the FOXA1 gene. Furthermore, there is evidence of somatic changes that influence the DNA sequence under FOXA1 binding regions, which may indirectly influence FOXA1-mediated regulation of ER and AR activity. These recent observations provide insight into the heterogeneity observed in ER and AR driven cancers.
Collapse
Affiliation(s)
- Jessica L L Robinson
- Cancer Research UK, Robinson Way Cambridge, UK ; Department of Oncology, University of Cambridge Cambridge, UK
| | | | | |
Collapse
|
86
|
Powers GL, Marker PC. Recent advances in prostate development and links to prostatic diseases. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2013; 5:243-56. [PMID: 23335485 DOI: 10.1002/wsbm.1208] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The prostate is a branched ductal-acinar gland that is part of the male reproductive tract. Prostate development depends upon the integration of steroid hormone signals, paracrine interactions between the stromal and epithelial tissue layers, and the actions of cell autonomous factors. Several genes and signaling pathways are known to be required for one or more steps of prostate development including epithelial budding, duct elongation, branching morphogenesis, and/or cellular differentiation. Recent progress in the field of prostate development has included the application of genome-wide technologies including serial analysis of gene expression, expression profiling microarrays, and other large-scale approaches to identify new genes and pathways that are essential for prostate development. The aggregation of experimental results into online databases by organized multilab projects including the Genitourinary Developmental Molecular Atlas Project has also accelerated the understanding of molecular pathways that function during prostate development and identified links between prostate anatomy and molecular signaling. Rapid progress has also recently been made in understanding the nature and role of candidate stem cells in the developing and adult prostate. This has included the identification of putative prostate stem cell markers, lineage tracing, and organ reconstitution studies. However, several issues regarding their origin, precise nature, and possible role(s) in disease remain unresolved. Nevertheless, several links between prostatic developmental mechanisms and the pathogenesis of prostatic diseases including benign prostatic hyperplasia and prostate cancer have led to recent progress on targeting developmental pathways as therapeutic strategies for these diseases.
Collapse
Affiliation(s)
- Ginny L Powers
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | | |
Collapse
|
87
|
Keil KP, Mehta V, Abler LL, Joshi PS, Schmitz CT, Vezina CM. Visualization and quantification of mouse prostate development by in situ hybridization. Differentiation 2012; 84:232-9. [PMID: 22898663 DOI: 10.1016/j.diff.2012.07.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 05/29/2012] [Accepted: 07/05/2012] [Indexed: 01/07/2023]
Abstract
The purpose of this study was to validate a combined in situ hybridization (ISH)/immunohistochemistry (IHC) staining method for visualizing and quantifying mouse prostatic buds. To refine animal usage in prostate development studies, we also determined whether a comparable number of prostatic buds were formed in male and female mouse urogenital sinus (UGS) explants grown in vitro in the presence of androgen. We used IHC to label UGS epithelium and ISH to label prostatic buds with one of three different prostatic bud marking riboprobes: a previously identified prostatic bud marker, NK-3 transcription factor, locus 1 (Nkx3-1), and two newly identified prostatic bud markers, wingless-related MMTV integration site 10b (Wnt10b) and ectodysplasin-A receptor (Edar). We calculated total buds formed per UGS and the proportion marked by each mRNA after male UGS development in vivo and male and female UGS development in vitro. Nkx3-1 was first to mark the prostate field during UGS development in vivo but all three mRNAs marked prostatic buds during later developmental stages. The mRNAs localized to different domains: Nkx3-1 was present along about half the prostatic bud length while Edar and Wnt10b were restricted to distal bud tips. None of the mRNAs marked all buds formed in vitro and the proportion marked was developmental stage- and gender-dependent. Nkx3-1 marked the highest proportion of prostatic buds during in vitro UGS development. Together, our results reveal that ISH staining of mouse UGS can be used to quantify prostatic bud number, Nkx3-1 is currently the best suited riboprobe for this method, and female UGSs cannot be used interchangeably with male UGSs when conducting prostate development studies in vitro. We also found that Nkx3-1, Edar, and Wnt10b mark different prostatic bud regions and are likely to be useful in future studies of regional differences in prostatic bud gene expression.
Collapse
Affiliation(s)
- Kimberly P Keil
- University of Wisconsin-Madison, Department of Comparative Biosciences, School of Veterinary Medicine, 1656 Linden Dr. Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
88
|
Imamura Y, Sakamoto S, Endo T, Utsumi T, Fuse M, Suyama T, Kawamura K, Imamoto T, Yano K, Uzawa K, Nihei N, Suzuki H, Mizokami A, Ueda T, Seki N, Tanzawa H, Ichikawa T. FOXA1 promotes tumor progression in prostate cancer via the insulin-like growth factor binding protein 3 pathway. PLoS One 2012; 7:e42456. [PMID: 22879989 PMCID: PMC3411739 DOI: 10.1371/journal.pone.0042456] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 07/09/2012] [Indexed: 12/28/2022] Open
Abstract
Fork-head box protein A1 (FOXA1) is a "pioneer factor" that is known to bind to the androgen receptor (AR) and regulate the transcription of AR-specific genes. However, the precise role of FOXA1 in prostate cancer (PC) remains unknown. In this study, we report that FOXA1 plays a critical role in PC cell proliferation. The expression of FOXA1 was higher in PC than in normal prostate tissues (P = 0.0002), and, using immunohistochemical analysis, we found that FOXA1 was localized in the nucleus. FOXA1 expression levels were significantly correlated with both PSA and Gleason scores (P = 0.016 and P = 0.031, respectively). Moreover, FOXA1 up-regulation was a significant factor in PSA failure (P = 0.011). Depletion of FOXA1 in a prostate cancer cell line (LNCaP) using small interfering RNA (siRNA) significantly inhibited AR activity, led to cell-growth suppression, and induced G0/G1 arrest. The anti-proliferative effect of FOXA1 siRNA was mediated through insulin-like growth factor binding protein 3 (IGFBP-3). An increase in IGFBP-3, mediated by depletion of FOXA1, inhibited phosphorylation of MAPK and Akt, and increased expression of the cell cycle regulators p21 and p27. We also found that the anti-proliferative effect of FOXA1 depletion was significantly reversed by simultaneous siRNA depletion of IGFBP-3. These findings provide direct physiological and molecular evidence for a role of FOXA1 in controlling cell proliferation through the regulation of IGFBP-3 expression in PC.
Collapse
Affiliation(s)
- Yusuke Imamura
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
- * E-mail:
| | - Takumi Endo
- Department of Urology, Toho University Medical Center Sakura Hospital, Chiba, Japan
| | - Takanobu Utsumi
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Miki Fuse
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takahito Suyama
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Koji Kawamura
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takashi Imamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kojiro Yano
- Faculty of Information Science and Technology, Osaka Institute of Technology, Osaka, Japan
| | - Katsuhiro Uzawa
- Department of Clinical Molecular Biology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naoki Nihei
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hiroyoshi Suzuki
- Department of Urology, Toho University Medical Center Sakura Hospital, Chiba, Japan
| | - Atsushi Mizokami
- Department of Urology, Kanazawa University Graduate School of Medical Sciences, Ishikawa, Japan
| | - Takeshi Ueda
- Prostate Center and Division of Urology, Chiba Cancer Center, Chiba, Japan
| | - Naohiko Seki
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hideki Tanzawa
- Department of Clinical Molecular Biology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
89
|
Guseva NV, Rokhlin OW, Bair TB, Glover RB, Cohen MB. Inhibition of p53 expression modifies the specificity of chromatin binding by the androgen receptor. Oncotarget 2012; 3:183-94. [PMID: 22383394 PMCID: PMC3326648 DOI: 10.18632/oncotarget.449] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The androgen receptor (AR) is known to play a critical role in prostate cancer (PC). p53 likely also plays a role given that p53 mutations are commonly found in advanced PC, and loss of wild-type protein function contributes to the phenotype of castration-resistant prostate cancer (CRPC). Nevertheless, the extent of the contribution of p53 dysfunction to PC remains unclear. Here we analyze the effects of p53 inhibition in PC cells and show that it has significant consequences for both the interaction between AR, and chromatin and the proliferative capacity of these cells. Inhibition of p53 expression enabled LNCaP cells to proliferate independently of androgens. Moreover, it modified the genome-wide binding pattern of AR. ChIP-sequnce analyis (ChIP-seq) revealed that fewer AR-binding sites were present in the context of p53 inhibition, suggesting that wild-type p53 is required for stable binding of AR to certain chromatin regions. Further analysis revealed that a lower AR occupancy was accompanied by a reduction in FoxA1 binding at regulatory regions of AR-dependent genes. Our study also identifies a pool of genes that may be transcriptionally regulated by AR only in the absence of p53, and that may contribute to the CRPC phenotype. Overall, our results point to p53 playing an important role in regulating AR activity across the genome.
Collapse
Affiliation(s)
- Natalya V Guseva
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA.
| | | | | | | | | |
Collapse
|
90
|
Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet 2012; 44:685-9. [PMID: 22610119 DOI: 10.1038/ng.2279] [Citation(s) in RCA: 1190] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 04/19/2012] [Indexed: 01/01/2023]
Abstract
Prostate cancer is the second most common cancer in men worldwide and causes over 250,000 deaths each year. Overtreatment of indolent disease also results in significant morbidity. Common genetic alterations in prostate cancer include losses of NKX3.1 (8p21) and PTEN (10q23), gains of AR (the androgen receptor gene) and fusion of ETS family transcription factor genes with androgen-responsive promoters. Recurrent somatic base-pair substitutions are believed to be less contributory in prostate tumorigenesis but have not been systematically analyzed in large cohorts. Here, we sequenced the exomes of 112 prostate tumor and normal tissue pairs. New recurrent mutations were identified in multiple genes, including MED12 and FOXA1. SPOP was the most frequently mutated gene, with mutations involving the SPOP substrate-binding cleft in 6-15% of tumors across multiple independent cohorts. Prostate cancers with mutant SPOP lacked ETS family gene rearrangements and showed a distinct pattern of genomic alterations. Thus, SPOP mutations may define a new molecular subtype of prostate cancer.
Collapse
|
91
|
DeGraff DJ, Clark PE, Cates JM, Yamashita H, Robinson VL, Yu X, Smolkin ME, Chang SS, Cookson MS, Herrick MK, Shariat SF, Steinberg GD, Frierson HF, Wu XR, Theodorescu D, Matusik RJ. Loss of the urothelial differentiation marker FOXA1 is associated with high grade, late stage bladder cancer and increased tumor proliferation. PLoS One 2012; 7:e36669. [PMID: 22590586 PMCID: PMC3349679 DOI: 10.1371/journal.pone.0036669] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 04/09/2012] [Indexed: 12/31/2022] Open
Abstract
Approximately 50% of patients with muscle-invasive bladder cancer (MIBC) develop metastatic disease, which is almost invariably lethal. However, our understanding of pathways that drive aggressive behavior of MIBC is incomplete. Members of the FOXA subfamily of transcription factors are implicated in normal urogenital development and urologic malignancies. FOXA proteins are implicated in normal urothelial differentiation, but their role in bladder cancer is unknown. We examined FOXA expression in commonly used in vitro models of bladder cancer and in human bladder cancer specimens, and used a novel in vivo tissue recombination system to determine the functional significance of FOXA1 expression in bladder cancer. Logistic regression analysis showed decreased FOXA1 expression is associated with increasing tumor stage (p<0.001), and loss of FOXA1 is associated with high histologic grade (p<0.001). Also, we found that bladder urothelium that has undergone keratinizing squamous metaplasia, a precursor to the development of squamous cell carcinoma (SCC) exhibited loss of FOXA1 expression. Furthermore, 81% of cases of SCC of the bladder were negative for FOXA1 staining compared to only 40% of urothelial cell carcinomas. In addition, we showed that a subpopulation of FOXA1 negative urothelial tumor cells are highly proliferative. Knockdown of FOXA1 in RT4 bladder cancer cells resulted in increased expression of UPK1B, UPK2, UPK3A, and UPK3B, decreased E-cadherin expression and significantly increased cell proliferation, while overexpression of FOXA1 in T24 cells increased E-cadherin expression and significantly decreased cell growth and invasion. In vivo recombination of bladder cancer cells engineered to exhibit reduced FOXA1 expression with embryonic rat bladder mesenchyme and subsequent renal capsule engraftment resulted in enhanced tumor proliferation. These findings provide the first evidence linking loss of FOXA1 expression with histological subtypes of MIBC and urothelial cell proliferation, and suggest an important role for FOXA1 in the malignant phenotype of MIBC.
Collapse
Affiliation(s)
- David J DeGraff
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Zhu Y, Jiang Q, Lou X, Ji X, Wen Z, Wu J, Tao H, Jiang T, He W, Wang C, Du Q, Zheng S, Mao J, Huang J. MicroRNAs up-regulated by CagA of Helicobacter pylori induce intestinal metaplasia of gastric epithelial cells. PLoS One 2012; 7:e35147. [PMID: 22536353 PMCID: PMC3335061 DOI: 10.1371/journal.pone.0035147] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 03/08/2012] [Indexed: 02/07/2023] Open
Abstract
CagA of Helicobacter pylori is a bacterium-derived oncogenic protein closely associated with the development of gastric cancers. MicroRNAs (miRNAs) are a class of widespread non-coding RNAs, many of which are involved in cell growth, cell differentiation and tumorigenesis. The relationship between CagA protein and miRNAs is unclear. Using mammalian miRNA profile microarrays, we found that miRNA-584 and miRNA-1290 expression was up-regulated in CagA-transformed cells, miRNA-1290 was up-regulated in an Erk1/2-dependent manner, and miRNA-584 was activated by NF-κB. miRNA-584 sustained Erk1/2 activities through inhibition of PPP2a activities, and miRNA-1290 activated NF-κB by knockdown of NKRF. Foxa1 was revealed to be an important target of miRNA-584 and miRNA-1290. Knockdown of Foxa1 promoted the epithelial-mesenchymal transition significantly. Overexpression of miRNA-584 and miRNA-1290 induced intestinal metaplasia of gastric epithelial cells in knock-in mice. These results indicate that miRNA-584 and miRNA-1290 interfere with cell differentiation and remodel the tissues. Thus, the miRNA pathway is a new pathogenic mechanism of CagA.
Collapse
Affiliation(s)
- Yongliang Zhu
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
FOXA1: a transcription factor with parallel functions in development and cancer. Biosci Rep 2012; 32:113-30. [PMID: 22115363 DOI: 10.1042/bsr20110046] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
When aberrant, factors critical for organ morphogenesis are also commonly involved in disease progression. FOXA1 (forkhead box A1), also known as HNF3α (hepatocyte nuclear factor 3α), is required for postnatal survival due to its essential role in controlling pancreatic and renal function. In addition to regulating a variety of tissues during embryogenesis and early life, rescue experiments have revealed a specific role for FOXA1 in the postnatal development of the mammary gland and prostate. Activity of the nuclear hormone receptors ERα (oestrogen receptor α) and AR (androgen receptor) is also required for proper development of the mammary gland and prostate respectively. FOXA1 modulates ER and AR function in breast and prostate cancer cells, supporting the postulate that FOXA1 is involved in ER and AR signalling under normal conditions, and that some carcinogenic processes in these tissues stem from hormonally regulated developmental pathways gone awry. In addition to broadly reviewing the function of FOXA1 in various aspects of development and cancer, this review focuses on the interplay of FOXA1/ER and FOXA1/AR, in normal and cancerous mammary and prostate epithelial cells. Given the hormone dependency of both breast and prostate cancer, a thorough understanding of FOXA1's role in both cancer types is critical for battling hormone receptor-positive disease and acquired anti-hormone resistance.
Collapse
|
94
|
FOXA1 Promotes Tumor Progression in Prostate Cancer and Represents a Novel Hallmark of Castration-Resistant Prostate Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:848-61. [DOI: 10.1016/j.ajpath.2011.10.021] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Revised: 09/23/2011] [Accepted: 10/21/2011] [Indexed: 11/19/2022]
|
95
|
Xu B, Hariharan A, Rakshit S, Dressler GR, Wellik DM. The role of Pax2 in mouse prostate development. Prostate 2012; 72:217-24. [PMID: 21594883 PMCID: PMC3178747 DOI: 10.1002/pros.21424] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 04/28/2011] [Indexed: 11/11/2022]
Abstract
BACKGROUND Loss-of-function of Pax2 results in severe defects of the male reproductive system, and Pax2 expression is detected in mouse prostate lobes and human prostatic cancers. However, the role for Pax2 in prostate development remains poorly understood. METHODS The expression of Pax2 was examined by in situ hybridization at various developmental stages. Urogenital sinuses were dissected out at E18.5 from mouse Pax2 mutants and controls, cultured in vitro or grafted under the renal capsule of CD1 nude mice. The expression of prostate developmental regulatory factors was analyzed by semi-quantitative real-time PCR or immuohistochemistry. RESULTS Pax2 is expressed in the epithelial cells of prostate buds. Loss-of-function of Pax2 does not affect the initiation of prostatic buds, but in vitro culture assays show that the prostates of Pax2 mutants are hypomorphic and branching is severely disrupted compared to controls. RT-PCR data from Pax2 mutant prostates demonstrate increased expression levels of dorsolateral prostate marker MSMB and ventral prostate marker SBP and dramatically reduced expression levels of anterior prostate marker TGM4. CONCLUSIONS Pax2 is essential for mouse prostate development and regulates prostatic ductal growth, branching, and lobe-specific identity. These findings are important for understanding the molecular regulatory mechanisms in prostate development.
Collapse
Affiliation(s)
- Ben Xu
- Department of Internal Medicine, Division of Molecular Medicine and Genetics
| | - Arun Hariharan
- Department of Internal Medicine, Division of Molecular Medicine and Genetics
| | - Sabita Rakshit
- Department of Internal Medicine, Division of Molecular Medicine and Genetics
| | - Gregory R. Dressler
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Deneen M. Wellik
- Department of Internal Medicine, Division of Molecular Medicine and Genetics
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-2200, USA
- Corresponding Author: Deneen M. Wellik, , University of Michigan Medical Center, 109 Zina Pitcher, 2053 BSRB, Ann Arbor, MI 48109-2200, Phone: 734-936-8902, FAX: 734-763-2162
| |
Collapse
|
96
|
Robinson JLL, Carroll JS. FoxA1 is a key mediator of hormonal response in breast and prostate cancer. Front Endocrinol (Lausanne) 2012; 3:68. [PMID: 22649425 PMCID: PMC3355944 DOI: 10.3389/fendo.2012.00068] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 04/30/2012] [Indexed: 12/17/2022] Open
Abstract
Hormonally regulated breast and prostate cancers are the most common cause of cancer in females and males respectively. FoxA1 acts as a pioneer factor for both androgen receptor (AR) and estrogen receptor-α (ER), dictating the binding location, and therefore function of these transcription factors. It is an essential protein for the transcriptional activity of both ER and AR, yet it has distinct roles with the two different nuclear receptors. In both malignancies, FoxA1 plays a pivotal role from early stage cancer through to drug resistant and metastatic disease. Due to this key role in mediating ER and AR function, FoxA1 is not only an attractive therapeutic target but could potentially function as a novel biomarker.
Collapse
Affiliation(s)
- Jessica L. L. Robinson
- Cancer Research UK, Li Ka Shing Centre, Robinson WayCambridge, UK
- Department of Oncology, University of CambridgeCambridge, UK
| | - Jason S. Carroll
- Cancer Research UK, Li Ka Shing Centre, Robinson WayCambridge, UK
- Department of Oncology, University of CambridgeCambridge, UK
- *Correspondence: Jason S. Carroll, Cancer Research UK, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK e-mail:
| |
Collapse
|
97
|
Critical and reciprocal regulation of KLF4 and SLUG in transforming growth factor β-initiated prostate cancer epithelial-mesenchymal transition. Mol Cell Biol 2011; 32:941-53. [PMID: 22203039 DOI: 10.1128/mcb.06306-11] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is implicated in various pathological processes within the prostate, including benign prostate hyperplasia (BPH) and prostate cancer progression. However, an ordered sequence of signaling events initiating carcinoma-associated EMT has not been established. In a model of transforming growth factor β (TGFβ)-induced prostatic EMT, SLUG is the dominant regulator of EMT initiation in vitro and in vivo, as demonstrated by the inhibition of EMT following Slug depletion. In contrast, SNAIL depletion was significantly less rate limiting. TGFβ-stimulated KLF4 degradation is required for SLUG induction. Expression of a degradation-resistant KLF4 mutant inhibited EMT, and furthermore, depletion of Klf4 was sufficient to initiate SLUG-dependent EMT. We show that KLF4 and another epithelial determinant, FOXA1, are direct transcriptional inhibitors of SLUG expression in mouse and human prostate cancer cells. Furthermore, self-reinforcing regulatory loops for SLUG-KLF4 and SLUG-FOXA1 lead to SLUG-dependent binding of polycomb repressive complexes to the Klf4 and Foxa1 promoters, silencing transcription and consolidating mesenchymal commitment. Analysis of tissue arrays demonstrated decreased KLF4 and increased SLUG expression in advanced-stage primary prostate cancer, substantiating the involvement of the EMT signaling events described in model systems.
Collapse
|
98
|
FOXA1: master of steroid receptor function in cancer. EMBO J 2011; 30:3885-94. [PMID: 21934649 DOI: 10.1038/emboj.2011.340] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 08/29/2011] [Indexed: 12/15/2022] Open
Abstract
FOXA transcription factors are potent, context-specific mediators of development that hold specialized functions in hormone-dependent tissues. Over the last several years, FOXA1 has emerged as a critical mediator of nuclear steroid receptor signalling, manifest at least in part through regulation of androgen receptor and oestrogen receptor activity. Recent findings point towards a major role for FOXA1 in modulating nuclear steroid receptor activity in breast and prostate cancer, and suggest that FOXA1 may significantly contribute to pro-tumourigenic phenotypes. The present review article will focus on the mechanisms, consequence, and clinical relevance of FOXA1-mediated steroid nuclear receptor signalling in human malignancy.
Collapse
|
99
|
When urothelial differentiation pathways go wrong: implications for bladder cancer development and progression. Urol Oncol 2011; 31:802-11. [PMID: 21924649 DOI: 10.1016/j.urolonc.2011.07.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 07/27/2011] [Accepted: 07/28/2011] [Indexed: 12/23/2022]
Abstract
Differentiation is defined as the ability of a cell to acquire full functional behavior. For instance, the function of bladder urothelium is to act as a barrier to the diffusion of solutes into or out of the urine after excretion by the kidney. The urothelium also serves to protect the detrusor muscle from toxins present in stored urine. A major event in the initiation and progression of bladder cancer is loss of urothelial differentiation. This is important because less differentiated urothelial tumors (higher histologic tumor grade) are typically associated with increased biologic and clinical aggressiveness. The differentiation status of urothelial carcinomas can be assessed by histopathologic examination and is reflected in the assignment of a histologic grade (low-grade or high-grade). Although typically limited to morphologic evaluation in most routine diagnostic practices, tumor grade can also be assessed using biochemical markers. Indeed, current pathological analysis of tumor specimens is increasingly reliant on molecular phenotyping. Thus, high priorities for bladder cancer research include identification of (1) biomarkers that will enable the identification of high grade T1 tumors that pose the most threat and require the most aggressive treatment; (2) biomarkers that predict the likelihood that a low grade, American Joint Committee on Cancer stage pTa bladder tumor will progress into an invasive carcinoma with metastatic potential; (3) biomarkers that indicate which pTa tumors are most likely to recur, thus enabling clinicians to prospectively identify patients who require aggressive treatment; and (4) how these markers might contribute to biological processes that underlie tumor progression and metastasis, potentially through loss of terminal differentiation. This review will discuss the proteins associated with urothelial cell differentiation, with a focus on those implicated in bladder cancer, and other proteins that may be involved in neoplastic progression. It is hoped that ongoing discoveries associated with the study of these differentiation-promoting proteins can be translated into the clinic to positively impact patient care.
Collapse
|
100
|
Sahu B, Laakso M, Ovaska K, Mirtti T, Lundin J, Rannikko A, Sankila A, Turunen JP, Lundin M, Konsti J, Vesterinen T, Nordling S, Kallioniemi O, Hautaniemi S, Jänne OA. Dual role of FoxA1 in androgen receptor binding to chromatin, androgen signalling and prostate cancer. EMBO J 2011; 30:3962-76. [PMID: 21915096 DOI: 10.1038/emboj.2011.328] [Citation(s) in RCA: 302] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 08/16/2011] [Indexed: 12/23/2022] Open
Abstract
High androgen receptor (AR) level in primary tumour predicts increased prostate cancer-specific mortality. However, the mechanisms that regulate AR function in prostate cancer are poorly known. We report here a new paradigm for the forkhead protein FoxA1 action in androgen signalling. Besides pioneering the AR pathway, FoxA1 depletion elicited extensive redistribution of AR-binding sites (ARBs) on LNCaP-1F5 cell chromatin that was commensurate with changes in androgen-dependent gene expression signature. We identified three distinct classes of ARBs and androgen-responsive genes: (i) independent of FoxA1, (ii) pioneered by FoxA1 and (iii) masked by FoxA1 and functional upon FoxA1 depletion. FoxA1 depletion also reprogrammed AR binding in VCaP cells, and glucocorticoid receptor binding and glucocorticoid-dependent signalling in LNCaP-1F5 cells. Importantly, FoxA1 protein level in primary prostate tumour had significant association to disease outcome; high FoxA1 level was associated with poor prognosis, whereas low FoxA1 level, even in the presence of high AR expression, predicted good prognosis. The role of FoxA1 in androgen signalling and prostate cancer is distinctly different from that in oestrogen signalling and breast cancer.
Collapse
Affiliation(s)
- Biswajyoti Sahu
- Institute of Biomedicine, Physiology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|