1
|
Uno W, Ofuji K, Wymeersch FJ, Takasato M. In vitro induction of prostate buds from murine urogenital epithelium in the absence of mesenchymal cells. Dev Biol 2023; 498:49-60. [PMID: 36963625 DOI: 10.1016/j.ydbio.2023.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/08/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
The prostate is a male reproductive gland which secretes prostatic fluid that enhances male fertility. During development and instigated by fetal testosterone, prostate cells arise caudal to the bladder at the urogenital sinus (UGS), when the urogenital mesenchyme (UGM) secretes signals to the urogenital epithelium (UGE). These initial mesenchymal signals induce prostate-specific gene expression in the UGE, after which epithelial progenitor cells form prostatic buds. Although many important factors for prostate development have been described using UGS organ cultures, those necessary and sufficient for prostate budding have not been clearly identified. This has been in part due to the difficulty to dissect the intricate signaling and feedback between epithelial and mesenchymal UGS cells. In this study, we separated the UGM from the UGE and tested candidate growth factors to show that when FGF10 is present, testosterone is not required for initiating prostate budding from the UGE. Moreover, in the presence of low levels of FGF10, canonical WNT signaling enhances the expression of several prostate progenitor markers in the UGE before budding of the prostate occurs. At the later budding stage, higher levels of FGF10 are required to increase budding and retinoic acid is indispensable for the upregulation of prostate-specific genes. Lastly, we show that under optimized conditions, female UGE can be instructed towards a prostatic fate, and in vitro generated prostate buds from male UGE can differentiate into a mature prostate epithelium after in vivo transplantation. Taken together, our results clarify the signals that can induce fetal prostate buds in the urogenital epithelium in the absence of the surrounding, instructive mesenchyme.
Collapse
Affiliation(s)
- Wataru Uno
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan; Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Kazuhiro Ofuji
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
| | - Filip J Wymeersch
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
| | - Minoru Takasato
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan; Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
2
|
Nybo ML, Kvam JM, Nielsen JE, Frederiksen H, Spiess K, Jensen KHR, Gadgaard S, Walser ALS, Thomsen JS, Cowin P, Juul A, Jensen MB, Rosenkilde M. Loss of Adgra3 causes obstructive azoospermia with high penetrance in male mice. FASEB J 2023; 37:e22781. [PMID: 36688818 PMCID: PMC10107928 DOI: 10.1096/fj.202200762rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023]
Abstract
The adhesion receptor ADGRA3 (GPR125) is a known spermatogonial stem cell marker, but its impact on male reproduction and fertility has not been examined. Using a mouse model lacking Adgra3 (Adgra3-/- ), we show that 55% of the male mice are infertile from puberty despite having normal spermatogenesis and epididymal sperm count. Instead, male mice lacking Adgra3 exhibited decreased estrogen receptor alpha expression and transient dilation of the epididymis. Combined with an increased estradiol production, this indicates a post-pubertal hormonal imbalance and fluid retention. Dye injection revealed a blockage between the ejaculatory duct and the urethra, which is rare in mice suffering from infertility, thereby mimicking the etiologies of obstructive azoospermia found in human male infertility. To summarize, male reproductive tract development is dependent on ADGRA3 function that in concert with estrogen signaling may influence fluid handling during sperm maturation and storage.
Collapse
Affiliation(s)
- Maja L. Nybo
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jone M. Kvam
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - John E. Nielsen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Copenhagen University Hospital – RigshospitaletCopenhagenDenmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Copenhagen University Hospital – RigshospitaletCopenhagenDenmark
| | - Katja Spiess
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Kristian H. R. Jensen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Sarina Gadgaard
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Bainan BiotechCopenhagenDenmark
| | - Anna L. S. Walser
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | | | - Pamela Cowin
- Department of Cell BiologyNew York University School of MedicineNew YorkNew YorkUSA
- Department of DermatologyNew York University School of MedicineNew YorkNew YorkUSA
| | - Anders Juul
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Copenhagen University Hospital – RigshospitaletCopenhagenDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Martin Blomberg Jensen
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and ReproductionCopenhagen University Hospital – RigshospitaletCopenhagenDenmark
- Division of Bone and Mineral Research, HSDM/HMSHarvard UniversityBostonMassachusettsUSA
| | - Mette M. Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
3
|
Joseph DB, Henry GH, Malewska A, Reese JC, Mauck RJ, Gahan JC, Hutchinson RC, Malladi VS, Roehrborn CG, Vezina CM, Strand DW. Single-cell analysis of mouse and human prostate reveals novel fibroblasts with specialized distribution and microenvironment interactions. J Pathol 2021; 255:141-154. [PMID: 34173975 DOI: 10.1002/path.5751] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 11/06/2022]
Abstract
Stromal-epithelial interactions are critical to the morphogenesis, differentiation, and homeostasis of the prostate, but the molecular identity and anatomy of discrete stromal cell types is poorly understood. Using single-cell RNA sequencing, we identified and validated the in situ localization of three smooth muscle subtypes (prostate smooth muscle, pericytes, and vascular smooth muscle) and two novel fibroblast subtypes in human prostate. Peri-epithelial fibroblasts (APOD+) wrap around epithelial structures, whereas interstitial fibroblasts (C7+) are interspersed in extracellular matrix. In contrast, the mouse displayed three fibroblast subtypes with distinct proximal-distal and lobe-specific distribution patterns. Statistical analysis of mouse and human fibroblasts showed transcriptional correlation between mouse prostate (C3+) and urethral (Lgr5+) fibroblasts and the human interstitial fibroblast subtype. Both urethral fibroblasts (Lgr5+) and ductal fibroblasts (Wnt2+) in the mouse contribute to a proximal Wnt/Tgfb signaling niche that is absent in human prostate. Instead, human peri-epithelial fibroblasts express secreted WNT inhibitors SFRPs and DKK1, which could serve as a buffer against stromal WNT ligands by creating a localized signaling niche around individual prostate glands. We also identified proximal-distal fibroblast density differences in human prostate that could amplify stromal signaling around proximal prostate ducts. In human benign prostatic hyperplasia, fibroblast subtypes upregulate critical immunoregulatory pathways and show distinct distributions in stromal and glandular phenotypes. A detailed taxonomy of leukocytes in benign prostatic hyperplasia reveals an influx of myeloid dendritic cells, T cells and B cells, resembling a mucosal inflammatory disorder. A receptor-ligand interaction analysis of all cell types revealed a central role for fibroblasts in growth factor, morphogen, and chemokine signaling to endothelia, epithelia, and leukocytes. These data are foundational to the development of new therapeutic targets in benign prostatic hyperplasia. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Diya B Joseph
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Gervaise H Henry
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Alicia Malewska
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Ryan J Mauck
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey C Gahan
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ryan C Hutchinson
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Venkat S Malladi
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Claus G Roehrborn
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Douglas W Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
4
|
Antao AM, Ramakrishna S, Kim KS. The Role of Nkx3.1 in Cancers and Stemness. Int J Stem Cells 2021; 14:168-179. [PMID: 33632988 PMCID: PMC8138659 DOI: 10.15283/ijsc20121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/17/2020] [Accepted: 12/14/2020] [Indexed: 12/23/2022] Open
Abstract
The well-known androgen-regulated homeobox gene, NKX3.1, is located on the short arm of chromosome 8. It is the first known prostate epithelium-specific marker, and is a transcription factor involved in development of the testes and prostate. In addition to specifying the prostate epithelium and maintaining normal prostate secretory function, Nkx3.1 is an established marker for prostate cancer. Over the years, however, this gene has been implicated in various other cancers, and technological advances have allowed determination of its role in other cellular functions. Nkx3.1 has also been recently identified as a factor capable of replacing Oct4 in cellular reprogramming. This review highlights the role of this tumor suppressor and briefly describes its functions, ranging from prostate development to maintenance of stemness and cellular reprogramming.
Collapse
Affiliation(s)
- Ainsley Mike Antao
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea.,College of Medicine, Hanyang University, Seoul, Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea.,College of Medicine, Hanyang University, Seoul, Korea
| |
Collapse
|
5
|
Wegner KA, Ruetten H, Girardi NM, O’Driscoll CA, Sandhu JK, Turco AE, Abler LL, Wang P, Wang Z, Bjorling DE, Malinowski R, Peterson RE, Strand DW, Marker PC, Vezina CM. Genetic background but not prostatic epithelial beta-catenin influences susceptibility of male mice to testosterone and estradiol-induced urinary dysfunction. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2021; 9:121-131. [PMID: 33816700 PMCID: PMC8012832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
Urinary voiding dysfunction in aging men can cause bothersome symptoms and irreparable tissue damage. Underlying mechanisms are not fully known. We previously demonstrated that subcutaneous, slow-release testosterone and estradiol implants (T+E2) drive a pattern of urinary voiding dysfunction in male mice that resembles that of aging men. The initial goal of this study was to test the hypothesis that prostatic epithelial beta-catenin (Ctnnb1) is required for T+E2-mediated voiding dysfunction. Targeted Ctnnb1 deletion did not significantly change voiding function in control or T+E2 treated mice but led to the surprising discovery that the C57BL/6J × FVB/NJ × 129S1 mixed genetic background onto which Ctnnb1 loss of function alleles were maintained is profoundly susceptible to voiding dysfunction. The mixed background mice develop a more rapid T+E2-mediated increase in spontaneous urine spotting, are more impaired in ability to initiate bladder contraction, and develop larger and heavier bladders than T+E2 treated C57BL/6J pure bred mice. To better understand mechanisms, we separately evaluated contributions of T and E2 and found that E2 mediates voiding dysfunction. Our findings that genetic factors serve as modifiers of responsiveness to T and E2 demonstrate the need to control for genetic background in studies of male voiding dysfunction. We also show that genetic factors could control severity of voiding dysfunction. We demonstrate the importance of E2 as a key mediator of voiding impairment, and show that the concentration of E2 in subcutaneous implants determines the severity of voiding dysfunction in mice, demonstrating that the mouse model is tunable, a factor which is important for future pharmacological intervention studies.
Collapse
Affiliation(s)
- Kyle A Wegner
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Hannah Ruetten
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Nicholas M Girardi
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Chelsea A O’Driscoll
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Jaskiran K Sandhu
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Anne E Turco
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Lisa L Abler
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Peiqing Wang
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Zunyi Wang
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Dale E Bjorling
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| | - Rita Malinowski
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
| | | | - Douglas W Strand
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- Department of Urology, University of Texas Southwestern Medical CenterDallas, Texas, USA
| | - Paul C Marker
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
| | - Chad M Vezina
- University of Wisconsin-Madison/UMASS Boston/UT-Southwestern George M. O’Brien Center for Benign Urologic ResearchMadison, Wisconsin 53706, USA
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-MadisonMadison, Wisconsin 53706, USA
| |
Collapse
|
6
|
Yu Y, Jiang W. Pluripotent stem cell differentiation as an emerging model to study human prostate development. Stem Cell Res Ther 2020; 11:285. [PMID: 32678004 PMCID: PMC7364497 DOI: 10.1186/s13287-020-01801-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
Prostate development is a complex process, and knowledge about this process is increasingly required for both basic developmental biology studies and clinical prostate cancer research, as prostate tumorigenesis can be regarded as the restoration of development in the adult prostate. Using rodent animal models, scientists have revealed that the development of the prostate is mainly mediated by androgen receptor (AR) signaling and that some other signaling pathways also play indispensable roles. However, there are still many unknowns in human prostate biology, mainly due to the limited availability of proper fetal materials. Here, we first briefly review prostate development with a focus on the AR, WNT, and BMP signaling pathways is necessary for prostate budding/BMP signaling pathways. Based on the current progress in in vitro prostatic differentiation and organoid techniques, we propose human pluripotent stem cells as an emerging model to study human prostate development.
Collapse
Affiliation(s)
- Yangyang Yu
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 116 East-Lake Road, District of Wuchang, Wuhan, 430071, Hubei Province, China
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 116 East-Lake Road, District of Wuchang, Wuhan, 430071, Hubei Province, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China. .,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
7
|
Leiblich A, Hellberg JEEU, Sekar A, Gandy C, Mendes CC, Redhai S, Mason J, Wainwright M, Marie P, Goberdhan DCI, Hamdy FC, Wilson C. Mating induces switch from hormone-dependent to hormone-independent steroid receptor-mediated growth in Drosophila secondary cells. PLoS Biol 2019; 17:e3000145. [PMID: 31589603 PMCID: PMC6797231 DOI: 10.1371/journal.pbio.3000145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 10/17/2019] [Accepted: 09/16/2019] [Indexed: 01/19/2023] Open
Abstract
Male reproductive glands like the mammalian prostate and the paired Drosophila melanogaster accessory glands secrete seminal fluid components that enhance fecundity. In humans, the prostate, stimulated by environmentally regulated endocrine and local androgens, grows throughout adult life. We previously showed that in fly accessory glands, secondary cells (SCs) and their nuclei also grow in adults, a process enhanced by mating and controlled by bone morphogenetic protein (BMP) signalling. Here, we demonstrate that BMP-mediated SC growth is dependent on the receptor for the developmental steroid ecdysone, whose concentration is reported to reflect sociosexual experience in adults. BMP signalling appears to regulate ecdysone receptor (EcR) levels via one or more mechanisms involving the EcR's N terminus or the RNA sequence that encodes it. Nuclear growth in virgin males is dependent on ecdysone, some of which is synthesised in SCs. However, mating induces additional BMP-mediated nuclear growth via a cell type-specific form of hormone-independent EcR signalling, which drives genome endoreplication in a subset of adult SCs. Switching to hormone-independent endoreplication after mating allows growth and secretion to be hyperactivated independently of ecdysone levels in SCs, permitting more rapid replenishment of the accessory gland luminal contents. Our data suggest mechanistic parallels between this physiological, behaviour-induced signalling switch and altered pathological signalling associated with prostate cancer progression.
Collapse
Affiliation(s)
- Aaron Leiblich
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | | | - Aashika Sekar
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Carina Gandy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Claudia C. Mendes
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Siamak Redhai
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - John Mason
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Mark Wainwright
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Pauline Marie
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Deborah C. I. Goberdhan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Freddie C. Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Clive Wilson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
8
|
Wegner KA, Mehta V, Johansson JA, Mueller BR, Keil KP, Abler LL, Marker PC, Taketo MM, Headon DJ, Vezina CM. Edar is a downstream target of beta-catenin and drives collagen accumulation in the mouse prostate. Biol Open 2019; 8:bio.037945. [PMID: 30745437 PMCID: PMC6451354 DOI: 10.1242/bio.037945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Beta-catenin (CTNNB1) directs ectodermal appendage spacing by activating ectodysplasin A receptor (EDAR) transcription, but whether CTNNB1 acts by a similar mechanism in the prostate, an endoderm-derived tissue, is unclear. Here we examined the expression, function, and CTNNB1 dependence of the EDAR pathway during prostate development. In situ hybridization studies reveal EDAR pathway components including Wnt10b in the developing prostate and localize these factors to prostatic bud epithelium where CTNNB1 target genes are co-expressed. We used a genetic approach to ectopically activate CTNNB1 in developing mouse prostate and observed focal increases in Edar and Wnt10b mRNAs. We also used a genetic approach to test the prostatic consequences of activating or inhibiting Edar expression. Edar overexpression does not visibly alter prostatic bud formation or branching morphogenesis, and Edar expression is not necessary for either of these events. However, Edar overexpression is associated with an abnormally thick and collagen-rich stroma in adult mouse prostates. These results support CTNNB1 as a transcriptional activator of Edar and Wnt10b in the developing prostate and demonstrate Edar is not only important for ectodermal appendage patterning but also influences collagen organization in adult prostates. This article has an associated First Person interview with the first author of the paper. Summary: This study provides a rare connection between beta catenin and ectodysplasin A receptor in an endoderm derived tissue and presents a potential mechanism for collagen accumulation in the prostate.
Collapse
Affiliation(s)
- Kyle A Wegner
- Molecular and Environmental Toxicology Center University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Vatsal Mehta
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jeanette A Johansson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH25 9RG, United Kingdom.,MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, United Kingdom
| | - Brett R Mueller
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kimberly P Keil
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Lisa L Abler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Paul C Marker
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - M Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University Yoshida-Konoé-cho, Sakyo, Kyoto 606-8501, Japan
| | - Denis J Headon
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH25 9RG, United Kingdom
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
9
|
Joseph DB, Chandrashekar AS, Abler LL, Chu LF, Thomson JA, Vezina CM. Epithelial DNA methyltransferase-1 regulates cell survival, growth and maturation in developing prostatic buds. Dev Biol 2019; 447:157-169. [PMID: 30659795 DOI: 10.1016/j.ydbio.2019.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/17/2018] [Accepted: 01/14/2019] [Indexed: 02/07/2023]
Abstract
DNA methyltransferase 1 (DNMT1) is required for embryogenesis but roles in late forming organ systems including the prostate, which emerges from the urethral epithelium, have not been fully examined. We used a targeted genetic approach involving a Shhcre recombinase to demonstrate requirement of epithelial DNA methyltransferase-1 (Dnmt1) in mouse prostate morphogenesis. Dnmt1 mutant urethral cells exhibit DNA hypomethylation, DNA damage, p53 accumulation and undergo cell cycle arrest and apoptosis. Urethral epithelial cells are disorganized in Dnmt1 mutants, leading to impaired prostate growth and maturation and failed glandular development. We evaluated oriented cell division as a mechanism of bud elongation and widening by demonstrating that mitotic spindle axes typically form parallel or perpendicular to prostatic bud elongation axes. We then deployed a ShhcreERT allele to delete Dnmt1 from a subset of urethral epithelial cells, creating mosaic mutants with which to interrogate the requirement for cell division in specific prostatic bud epithelial populations. DNMT1- cell distribution within prostatic buds is not random as would be expected in a process where DNMT1 was not required. Instead, replication competent DNMT1 + cells primarily accumulate in prostatic bud margins and tips while replication impeded DNMT1- cells accumulate in prostatic bud cores. Together, these results highlight the role of DNMT1 in regulating epithelial bud formation by maintaining cell cycle progression and survival of rapidly dividing urethral epithelial cells, which can be extended to the study of other developing epithelial organs. In addition, our results show that prostatic buds consist of two epithelial cell populations with distinct molecular and functional characteristics that could potentially contribute to specialized lineages in the adult prostate.
Collapse
Affiliation(s)
- Diya B Joseph
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anoop S Chandrashekar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Lisa L Abler
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Li-Fang Chu
- Morgridge Institute for Research, Madison, WI 53715, USA; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53707-7365, USA
| | - James A Thomson
- Morgridge Institute for Research, Madison, WI 53715, USA; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53707-7365, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
10
|
Cunha GR, Vezina CM, Isaacson D, Ricke WA, Timms BG, Cao M, Franco O, Baskin LS. Development of the human prostate. Differentiation 2018; 103:24-45. [PMID: 30224091 PMCID: PMC6234090 DOI: 10.1016/j.diff.2018.08.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/14/2022]
Abstract
This paper provides a detailed compilation of human prostatic development that includes human fetal prostatic gross anatomy, histology, and ontogeny of selected epithelial and mesenchymal differentiation markers and signaling molecules throughout the stages of human prostatic development: (a) pre-bud urogenital sinus (UGS), (b) emergence of solid prostatic epithelial buds from urogenital sinus epithelium (UGE), (c) bud elongation and branching, (d) canalization of the solid epithelial cords, (e) differentiation of luminal and basal epithelial cells, and (f) secretory cytodifferentiation. Additionally, we describe the use of xenografts to assess the actions of androgens and estrogens on human fetal prostatic development. In this regard, we report a new model of de novo DHT-induction of prostatic development from xenografts of human fetal female urethras, which emphasizes the utility of the xenograft approach for investigation of initiation of human prostatic development. These studies raise the possibility of molecular mechanistic studies on human prostatic development through the use of tissue recombinants composed of mutant mouse UGM combined with human fetal prostatic epithelium. Our compilation of human prostatic developmental processes is likely to advance our understanding of the pathogenesis of benign prostatic hyperplasia and prostate cancer as the neoformation of ductal-acinar architecture during normal development is shared during the pathogenesis of benign prostatic hyperplasia and prostate cancer.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States.
| | - Chad M Vezina
- School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, United States
| | - Dylan Isaacson
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - William A Ricke
- Department of Urology, University of Wisconsin, Madison, WI 53705, United States
| | - Barry G Timms
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, United States
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| | - Omar Franco
- Department of Surgery, North Shore University Health System, 1001 University Place, Evanston, IL 60201, United States
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA 94143, United States
| |
Collapse
|
11
|
Abstract
The prostate is a male exocrine gland that secretes components of the seminal fluid. In men, prostate tumors are one of the most prevalent cancers. Studies on the development of the prostate have given a better understanding of the processes and genes that are important in the formation of this organ and have provided insights into the mechanisms of prostate tumorigenesis. These developmental studies have provided evidence that some of the genes and signaling pathways involved in development are reactivated or deregulated during prostate cancer. The prostate goes through a number of different stages during organogenesis, which include organ specification, epithelial budding, branching morphogenesis, canalization, and cytodifferentiation. During development, these processes are tightly regulated, many of which are controlled by the male hormone androgens. The majority of prostate tumors remain hormone regulated, and antiandrogen therapy is a first-line therapy, highlighting the important link between prostate organogenesis and cancer. In this review, we describe some of the data on genes that have important roles during prostate development that also have strong evidence linking them to prostate cancer.
Collapse
Affiliation(s)
- Jeffrey C Francis
- Division of Cancer Biology, Institute of Cancer Research, London SW3 6JB, United Kingdom
| | - Amanda Swain
- Division of Cancer Biology, Institute of Cancer Research, London SW3 6JB, United Kingdom
| |
Collapse
|
12
|
He Y, Hooker E, Yu EJ, Wu H, Cunha GR, Sun Z. An Indispensable Role of Androgen Receptor in Wnt Responsive Cells During Prostate Development, Maturation, and Regeneration. Stem Cells 2018; 36:891-902. [PMID: 29451339 DOI: 10.1002/stem.2806] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/09/2018] [Accepted: 02/05/2018] [Indexed: 02/05/2023]
Abstract
Androgen signaling is essential for prostate development, morphogenesis, and regeneration. Emerging evidence indicates that Wnt/β-catenin signaling also contributes to prostate development specifically through regulation of cell fate determination. Prostatic Axin2-expressing cells are able to respond to Wnt signals and possess the progenitor properties to regenerate prostatic epithelium. Despite critical roles of both signaling pathways, the biological significance of androgen receptor (AR) in Axin2-expressing/Wnt-responsive cells remains largely unexplored. In this study, we investigated this important question using a series of newly generated mouse models. Deletion of Ar in embryonic Axin2-expressing cells impaired early prostate development in both ex vivo and tissue implantation experiments. When Ar expression was deleted in prostatic Axin2-expressing cells at pre-puberty stages, it results in smaller and underdeveloped prostates. A subpopulation of Axin2 expressing cells in prostate epithelium is resistant to castration and, following androgen supplementation, is capable to expand to prostatic luminal cells. Deletion of Ar in these Axin2-expressing cells reduces their regenerative ability. These lines of evidence demonstrate an indispensable role for the Ar in Wnt-responsive cells during the course of prostate development, morphogenesis, and regeneration, which also imply an underlying interaction between the androgen and Wnt signaling pathways in the mouse prostate. Stem Cells 2018;36:891-902.
Collapse
Affiliation(s)
- Yongfeng He
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, California, USA.,Department of Urology, Stanford University School of Medicine, Stanford, California, USA
| | - Erika Hooker
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, California, USA.,Department of Urology, Stanford University School of Medicine, Stanford, California, USA
| | - Eun-Jeong Yu
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, California, USA.,Department of Urology, Stanford University School of Medicine, Stanford, California, USA
| | - Huiqing Wu
- Department of Pathology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, California, USA
| | - Gerald R Cunha
- Department of Urology, School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Zijie Sun
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, California, USA.,Department of Urology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
13
|
Montano M, Bushman W. Morphoregulatory pathways in prostate ductal development. Dev Dyn 2018; 246:89-99. [PMID: 27884054 DOI: 10.1002/dvdy.24478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 01/22/2023] Open
Abstract
The mouse prostate is a male sex-accessory gland comprised of a branched ductal network arranged into three separate bilateral lobes: the anterior, dorsolateral, and ventral lobes. Prostate ductal development is the primary morphogenetic event in prostate development and requires a complex regulation of spatiotemporal factors. This review provides an overview of prostate development and the major genetic regulators and signaling pathways involved. To identify new areas for further study, we briefly highlight the likely important, but relatively understudied, role of the extracellular matrix (ECM). Finally, we point out the potential importance of the ECM in influencing the behavior and prognosis of prostate cancer. Developmental Dynamics 246:89-99, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Monica Montano
- University of Wisconsin Madison, Department of Urology, Madison, Wisconsin.,University of Wisconsin Madison, Cellular and Molecular Pathology, Madison, Wisconsin.,University of Wisconsin Madison, Carbone Cancer Center, Clinical Sciences Center, Madison, Wisconsin
| | - Wade Bushman
- University of Wisconsin Madison, Department of Urology, Madison, Wisconsin.,University of Wisconsin Madison, Carbone Cancer Center, Clinical Sciences Center, Madison, Wisconsin
| |
Collapse
|
14
|
Wegner KA, Cadena MT, Trevena R, Turco AE, Gottschalk A, Halberg RB, Guo J, McMahon JA, McMahon AP, Vezina CM. An immunohistochemical identification key for cell types in adult mouse prostatic and urethral tissue sections. PLoS One 2017; 12:e0188413. [PMID: 29145476 PMCID: PMC5690684 DOI: 10.1371/journal.pone.0188413] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023] Open
Abstract
Though many methods can be used to identify cell types contained in complex tissues, most require cell disaggregation and destroy information about where cells reside in relation to their microenvironment. Here, we describe a polytomous key for cell type identification in intact sections of adult mouse prostate and prostatic urethra. The key is organized as a decision tree and initiates with one round of immunostaining for nerve, epithelial, fibromuscular/hematolymphoid, or vascular associated cells. Cell identities are recursively eliminated by subsequent staining events until the remaining pool of potential cell types can be distinguished by direct comparison to other cells. We validated our identification key using wild type adult mouse prostate and urethra tissue sections and it currently resolves sixteen distinct cell populations which include three nerve fiber types as well as four epithelial, five fibromuscular/hematolymphoid, one nerve-associated, and three vascular-associated cell types. We demonstrate two uses of this novel identification methodology. We first used the identification key to characterize prostate stromal cell type changes in response to constitutive phosphatidylinositide-3-kinase activation in prostate epithelium. We then used the key to map cell lineages in a new reporter mouse strain driven by Wnt10aem1(cre/ERT2)Amc. The identification key facilitates rigorous and reproducible cell identification in prostate tissue sections and can be expanded to resolve additional cell types as new antibodies and other resources become available.
Collapse
Affiliation(s)
- Kyle A. Wegner
- George M. O’Brien Benign Urology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mark T. Cadena
- George M. O’Brien Benign Urology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ryan Trevena
- George M. O’Brien Benign Urology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Anne E. Turco
- George M. O’Brien Benign Urology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Adam Gottschalk
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Richard B. Halberg
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| | - Jill A. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| | - Chad M. Vezina
- George M. O’Brien Benign Urology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
15
|
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: 117] [Impact Index Per Article: 16.7] [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
|
16
|
Thymus neuroendocrine tumors with CTNNB1 gene mutations, disarrayed ß-catenin expression, and dual intra-tumor Ki-67 labeling index compartmentalization challenge the concept of secondary high-grade neuroendocrine tumor: a paradigm shift. Virchows Arch 2017; 471:31-47. [PMID: 28451756 DOI: 10.1007/s00428-017-2130-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/09/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023]
Abstract
We herein report an uncommon association of intimately admixed atypical carcinoid (AC) and large cell neuroendocrine (NE) carcinoma (LCNEC) of the thymus, occurring in two 20- and 39-year-old Caucasian males. Both tumors were treated by maximal thymectomy. The younger patient presented with a synchronous lesion and died of disease after 9 months, while the other patient was associated with a recurrent ectopic adrenocorticotropic hormone Cushing's syndrome and is alive with disease at the 2-year follow-up. MEN1 syndrome was excluded in either case. Immunohistochemically, disarrayed cytoplasmic and nuclear ß-catenin expression was seen alongside an intra-tumor Ki-67 antigen labeling index (LI) ranging from 2 to 80% in the younger patient's tumor and from 3 to 45% in the other. Both exhibited upregulated cyclin D1 and retinoblastoma, while vimentin was overexpressed in the recurrent LCNEC only. Next-generation sequencing revealed CTNNB1, TP53, and JAK3 mutations in the synchronous tumor and CTNNB1 mutation alone in the metachronous tumor (the latter with the same mutation as the first tumor of 17 years prior). None of the 23 T-NET controls exhibited this hallmarking triple alteration (p = 0.003). These findings suggested that LCNEC components developed from pre-existing CTNNB1-mutated AC upon loss-of-function TP53 and gain-of-function JAK3 mutations in one case and an epithelial-mesenchymal transition upon vimentin overexpression in the other case. Both tumors maintained intact cyclin D1-retinoblastoma machinery. Our report challenges the concept of secondary LCNEC as an entity that develops from pre-existing AC as a result of tumor progression, suggesting a paradigm shift to the current pathogenesis of NET.
Collapse
|
17
|
Mulligan WA, Wegner KA, Keil KP, Mehta V, Taketo MM, Vezina CM. Beta-catenin and estrogen signaling collaborate to drive cyclin D1 expression in developing mouse prostate. Differentiation 2016; 93:66-71. [PMID: 27918915 DOI: 10.1016/j.diff.2016.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/31/2016] [Accepted: 11/07/2016] [Indexed: 12/17/2022]
Abstract
Androgen, beta-catenin (CTNNB1), and estrogen pathways stimulate proliferative growth of developing mouse prostate but how these pathways interact is not fully understood. We previously found that androgens induce CTNNB1 signaling in mouse urogenital sinus (UGS) epithelium from which prostatic ductal epithelium derives. Others have shown that low estradiol concentrations induce UGS epithelial proliferative growth. Here, we found that CTNNB1 signaling overlaps cyclin D1 (CCND1) expression in prostatic buds and we used a genetic approach to test whether CTNNB1 signaling induces CCND1 expression. We observed an unexpected sexually dimorphic response to hyperactive CCNTB1 signaling: in male mouse UGS it increased Ccnd1 mRNA abundance without increasing its protein abundance but in female UGS it increased Ccnd1 mRNA and protein abundance, suggesting a potential role for estrogens in stabilizing CCND1 protein. Treating wild type male UGS explants with androgen and either 17β-estradiol or a proteasome inhibitor increased CCND1 protein and KI67 labeling in prostatic bud epithelium. Together, our results are consistent with an epithelial proliferative growth mechanism linking CTNNB1-driven Ccnd1 transcription and estrogen-mediated CCND1 protein stabilization.
Collapse
Affiliation(s)
- William A Mulligan
- George M. O'Brien Benign Urology Center, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA; School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Kyle A Wegner
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Kimberly P Keil
- School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Vatsal Mehta
- School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - M Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University Yoshida-Konoé-cho, Sakyo, Kyoto 606-8501, Japan
| | - Chad M Vezina
- George M. O'Brien Benign Urology Center, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA; School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA; Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
18
|
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: 2.0] [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
|
19
|
Bolt CC, Negi S, Guimarães-Camboa N, Zhang H, Troy JM, Lu X, Kispert A, Evans SM, Stubbs L. Tbx18 Regulates the Differentiation of Periductal Smooth Muscle Stroma and the Maintenance of Epithelial Integrity in the Prostate. PLoS One 2016; 11:e0154413. [PMID: 27120339 PMCID: PMC4847854 DOI: 10.1371/journal.pone.0154413] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 04/13/2016] [Indexed: 11/18/2022] Open
Abstract
The T-box transcription factor TBX18 is essential to mesenchymal cell differentiation in several tissues and Tbx18 loss-of-function results in dramatic organ malformations and perinatal lethality. Here we demonstrate for the first time that Tbx18 is required for the normal development of periductal smooth muscle stromal cells in prostate, particularly in the anterior lobe, with a clear impact on prostate health in adult mice. Prostate abnormalities are only subtly apparent in Tbx18 mutants at birth; to examine postnatal prostate development we utilized a relatively long-lived hypomorphic mutant and a novel conditional Tbx18 allele. Similar to the ureter, cells that fail to express Tbx18 do not condense normally into smooth muscle cells of the periductal prostatic stroma. However, in contrast to ureter, the periductal stromal cells in mutant prostate assume a hypertrophic, myofibroblastic state and the adjacent epithelium becomes grossly disorganized. To identify molecular events preceding the onset of this pathology, we compared gene expression in the urogenital sinus (UGS), from which the prostate develops, in Tbx18-null and wild type littermates at two embryonic stages. Genes that regulate cell proliferation, smooth muscle differentiation, prostate epithelium development, and inflammatory response were significantly dysregulated in the mutant urogenital sinus around the time that Tbx18 is first expressed in the wild type UGS, suggesting a direct role in regulating those genes. Together, these results argue that Tbx18 is essential to the differentiation and maintenance of the prostate periurethral mesenchyme and that it indirectly regulates epithelial differentiation through control of stromal-epithelial signaling.
Collapse
Affiliation(s)
- C. Chase Bolt
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
| | - Soumya Negi
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
| | - Nuno Guimarães-Camboa
- Skaggs School of Pharmacy, Department of Medicine, and Department of Pharmacology, University of California San Diego, La Jolla, CA, United States of America, 92037
| | - Huimin Zhang
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
| | - Joseph M. Troy
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
| | - Xiaochen Lu
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
| | - Andreas Kispert
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Sylvia M. Evans
- Skaggs School of Pharmacy, Department of Medicine, and Department of Pharmacology, University of California San Diego, La Jolla, CA, United States of America, 92037
| | - Lisa Stubbs
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America, 61801
- * E-mail:
| |
Collapse
|
20
|
Tan PS, Nakagawa S, Goossens N, Venkatesh A, Huang T, Ward SC, Sun X, Song WM, Koh A, Canasto-Chibuque C, Deshmukh M, Nair V, Mahajan M, Zhang B, Fiel MI, Kobayashi M, Kumada H, Hoshida Y. Clinicopathological indices to predict hepatocellular carcinoma molecular classification. Liver Int 2016; 36:108-18. [PMID: 26058462 PMCID: PMC4674393 DOI: 10.1111/liv.12889] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/01/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) is the second most lethal cancer caused by lack of effective therapies. Although promising, HCC molecular classification, which enriches potential responders to specific therapies, has not yet been assessed in clinical trials of anti-HCC drugs. We aimed to overcome these challenges by developing clinicopathological surrogate indices of HCC molecular classification. METHODS Hepatocellular carcinoma classification defined in our previous transcriptome meta-analysis (S1, S2 and S3 subclasses) was implemented in an FDA-approved diagnostic platform (Elements assay, NanoString). Ninety-six HCC tumours (training set) were assayed to develop molecular subclass-predictive indices based on clinicopathological features, which were independently validated in 99 HCC tumours (validation set). Molecular deregulations associated with the histopathological features were determined by pathway analysis. Sample sizes for HCC clinical trials enriched with specific molecular subclasses were determined. RESULTS Hepatocellular carcinoma subclass-predictive indices were steatohepatitic (SH)-HCC variant and immune cell infiltrate for S1 subclass, macrotrabecular/compact pattern, lack of pseudoglandular pattern, and high serum alpha-foetoprotein (>400 ng/ml) for S2 subclass, and microtrabecular pattern, lack of SH-HCC and clear cell variants, and lower histological grade for S3 subclass. Macrotrabecular/compact pattern, a predictor of S2 subclass, was associated with the activation of therapeutically targetable oncogene YAP and stemness markers EPCAM/KRT19. BMP4 was associated with pseudoglandular pattern. Subclass-predictive indices-based patient enrichment reduced clinical trial sample sizes from 121, 184 and 53 to 30, 43 and 22 for S1, S2 and S3 subclass-targeting therapies respectively. CONCLUSIONS Hepatocellular carcinoma molecular subclasses can be enriched by clinicopathological indices tightly associated with deregulation of therapeutically targetable molecular pathways.
Collapse
Affiliation(s)
- Poh Seng Tan
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S,Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Health System, Singapore
| | - Shigeki Nakagawa
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Nicolas Goossens
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S,Division of Gastroenterology and Hepatology, Geneva University Hospital, Switzerland
| | - Anu Venkatesh
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Tiangui Huang
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Stephen C. Ward
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Xiaochen Sun
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Anna Koh
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Claudia Canasto-Chibuque
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Manjeet Deshmukh
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Venugopalan Nair
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Milind Mahajan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, U.S
| | - Maria Isabel Fiel
- Division of Gastroenterology and Hepatology, Geneva University Hospital, Switzerland
| | | | | | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, U.S
| |
Collapse
|
21
|
Gamat M, Malinowski RL, Parkhurst LJ, Steinke LM, Marker PC. Ornithine Decarboxylase Activity Is Required for Prostatic Budding in the Developing Mouse Prostate. PLoS One 2015; 10:e0139522. [PMID: 26426536 PMCID: PMC4591331 DOI: 10.1371/journal.pone.0139522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/12/2015] [Indexed: 11/23/2022] Open
Abstract
The prostate is a male accessory sex gland that produces secretions in seminal fluid to facilitate fertilization. Prostate secretory function is dependent on androgens, although the mechanism by which androgens exert their effects is still unclear. Polyamines are small cationic molecules that play pivotal roles in DNA transcription, translation and gene regulation. The rate-limiting enzyme in polyamine biosynthesis is ornithine decarboxylase, which is encoded by the gene Odc1. Ornithine decarboxylase mRNA decreases in the prostate upon castration and increases upon administration of androgens. Furthermore, testosterone administered to castrated male mice restores prostate secretory activity, whereas administering testosterone and the ornithine decarboxylase inhibitor D,L-α-difluromethylornithine (DFMO) to castrated males does not restore prostate secretory activity, suggesting that polyamines are required for androgens to exert their effects. To date, no one has examined polyamines in prostate development, which is also androgen dependent. In this study, we showed that ornithine decarboxylase protein was expressed in the epithelium of the ventral, dorsolateral and anterior lobes of the adult mouse prostate. Ornithine decarboxylase protein was also expressed in the urogenital sinus (UGS) epithelium of the male and female embryo prior to prostate development, and expression continued in prostatic epithelial buds as they emerged from the UGS. Inhibiting ornithine decarboxylase using DFMO in UGS organ culture blocked the induction of prostatic buds by androgens, and significantly decreased expression of key prostate transcription factor, Nkx3.1, by androgens. DFMO also significantly decreased the expression of developmental regulatory gene Notch1. Other genes implicated in prostatic development including Sox9, Wif1 and Srd5a2 were unaffected by DFMO. Together these results indicate that Odc1 and polyamines are required for androgens to exert their effect in mediating prostatic bud induction, and are required for the expression of a subset of prostatic developmental regulatory genes including Notch1 and Nkx3.1.
Collapse
Affiliation(s)
- Melissa Gamat
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
| | - Rita L. Malinowski
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
| | - Linnea J. Parkhurst
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
| | - Laura M. Steinke
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
| | - Paul C. Marker
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, United States of America
- * E-mail:
| |
Collapse
|
22
|
Keil KP, Vezina CM. DNA methylation as a dynamic regulator of development and disease processes: spotlight on the prostate. Epigenomics 2015; 7:413-25. [PMID: 26077429 DOI: 10.2217/epi.15.8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prostate development, benign hyperplasia and cancer involve androgen and growth factor signaling as well as stromal-epithelial interactions. We review how DNA methylation influences these and related processes in other organ systems such as how proliferation is restricted to specific cell populations during defined temporal windows, how androgens elicit their actions and how cells establish, maintain and remodel DNA methylation in a time and cell specific fashion. We also discuss mechanisms by which hormones and endocrine disrupting chemicals reprogram DNA methylation in the prostate and elsewhere and examine evidence for a reawakening of developmental epigenetic pathways as drivers of prostate cancer and benign prostate hyperplasia.
Collapse
Affiliation(s)
- Kimberly P Keil
- Comparative Biosciences, University of Wisconsin-Madison, 1656 Linden Dr., Madison, WI 53705, USA
| | - Chad M Vezina
- Comparative Biosciences, University of Wisconsin-Madison, 1656 Linden Dr., Madison, WI 53705, USA
| |
Collapse
|
23
|
Keil KP, Altmann HM, Abler LL, Hernandez LL, Vezina CM. Histone acetylation regulates prostate ductal morphogenesis through a bone morphogenetic protein-dependent mechanism. Dev Dyn 2015; 244:1404-14. [PMID: 26283270 DOI: 10.1002/dvdy.24321] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 07/01/2015] [Accepted: 08/06/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Epigenetic factors influence stem cell function and other developmental events but their role in prostate morphogenesis is not completely known. We tested the hypothesis that histone deacetylase (HDAC) activity is required for prostate morphogenesis. RESULTS We identified the presence of class I nuclear HDACs in the mouse urogenital sinus (UGS) during prostate development and found that Hdac 2 mRNA abundance diminishes as development proceeds which is especially evident in prostatic epithelium. Blockade of HDACs with the inhibitor trichostatin A (TSA) decreased the number of prostatic buds formed in UGS explant cultures but not the number of buds undergoing branching morphogenesis. In the latter, TSA promoted an extensive branching phenotype that was reversed by exogenous NOGGIN protein, which functions as a bone morphogenetic protein (BMP) inhibitor. TSA also increased Bmp2 promoter H3K27ac abundance, Bmp2 and Bmp4 mRNA abundance, and the percentage of epithelial cells marked by BMP-responsive phosphorylated SMAD1/5/8 protein. TSA exposed UGS explants grafted under the kidney capsule of untreated host mice for continued development achieved a smaller size without an obvious difference in glandular histology compared with control treated grafts. CONCLUSIONS These results are consistent with an active role for HDACs in shaping prostate morphogenesis by regulating Bmp abundance.
Collapse
Affiliation(s)
- Kimberly P Keil
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Helene M Altmann
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lisa L Abler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Laura L Hernandez
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
24
|
Bauman TM, Vezina CM, Huang W, Marker PC, Peterson RE, Ricke WA. Beta-catenin is elevated in human benign prostatic hyperplasia specimens compared to histologically normal prostate tissue. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2014; 2:313-322. [PMID: 25606577 PMCID: PMC4297327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/09/2014] [Indexed: 06/04/2023]
Abstract
Benign prostatic hyperplasia (BPH) is linked to lower urinary tract symptoms (LUTS) such as incomplete bladder emptying, urinary frequency and urgency. Mechanisms responsible for BPH are not fully known. Here, we tested whether beta-catenin (CTNNB1) immunostaining intensity and distribution differ in human glandular BPH tissue specimens compared to normal prostate tissue. Multiplex immunostaining of CTNNB1, its putative transcriptional target gene lymphoid enhancer binding factor 1 (LEF1), and the epithelial marker E-cadherin were examined in clinical human prostate specimens with or without histological BPH (pure epithelial or mixed stromal-epithelial nodules). BPH specimens were obtained from 24 men who experienced LUTS and underwent transurethral resection of the prostate surgery. Control specimens were tumor-adjacent histologically normal prostate tissue from 48 patients who underwent radical prostatectomy. The resulting multispectral images were unmixed and optical densities recorded to quantify staining abundance, cellular (membranous, cytoplasmic, and nuclear) and tissue localization (stromal versus epithelial), and determination of percentage of CTNNB1-positive cells. The following CTNNB1 indices were significantly higher in BPH compared to normal prostate tissue: overall staining intensity, staining intensity in prostate stromal cell membranes, cytoplasm and nuclei, and prostate epithelial cell nuclei. The following LEF1 indices were significantly lower in BPH compared to tumor-adjacent normal prostate tissue: stromal LEF1 staining intensity, percentage of LEF1-positive stromal cells, and intensity of LEF1 staining in stromal cell membranes, cytoplasm, and nuclei. The percentage of stromal cells with CTNNB1(+)/LEF1(-) nuclei was higher and percentage of stromal cells with CTNNB1(-)/LEF1(+) nuclei was lower in BPH compared to tumor-adjacent normal prostate tissues. These results support the hypothesis that CTNNB1 expression increases in specific BPH tissue compartments. Further, since nuclear LEF1 staining does not coincide with cytoplasmic or nuclear CTNNB1 staining, it does not appear to be a reliable index of CTNNB1 activity in adult human prostate.
Collapse
Affiliation(s)
- Tyler M Bauman
- Department of Urology and Carbone Cancer Center, University of Wisconsin School of Medicine and Public HealthMadison, WI 53705, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary MedicineMadison, WI 53706, USA
- University of Wisconsin O’Brien Urology Research CenterMadison, WI, USA
| | - Wei Huang
- University of Wisconsin O’Brien Urology Research CenterMadison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public HealthMadison, WI 53705, USA
| | - Paul C Marker
- University of Wisconsin O’Brien Urology Research CenterMadison, WI, USA
- Division of Pharmaceutical Sciences, University of Wisconsin School of PharmacyMadison, WI 53705, USA
| | - Richard E Peterson
- Division of Pharmaceutical Sciences, University of Wisconsin School of PharmacyMadison, WI 53705, USA
| | - William A Ricke
- Department of Urology and Carbone Cancer Center, University of Wisconsin School of Medicine and Public HealthMadison, WI 53705, USA
- University of Wisconsin O’Brien Urology Research CenterMadison, WI, USA
| |
Collapse
|
25
|
Keil KP, Abler LL, Laporta J, Altmann HM, Yang B, Jarrard DF, Hernandez LL, Vezina CM. Androgen receptor DNA methylation regulates the timing and androgen sensitivity of mouse prostate ductal development. Dev Biol 2014; 396:237-45. [PMID: 25446526 DOI: 10.1016/j.ydbio.2014.10.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 10/11/2014] [Accepted: 10/13/2014] [Indexed: 12/27/2022]
Abstract
Androgen receptor (AR) signaling initiates mouse prostate development by stimulating prostate ductal bud formation and specifying bud patterns. Curiously, however, prostatic bud initiation lags behind the onset of gonadal testosterone synthesis by about three days. This study's objective was to test the hypothesis that DNA methylation controls the timing and scope of prostate ductal development by regulating Ar expression in the urogenital sinus (UGS) from which the prostate derives. We determined that Ar DNA methylation decreases in UGS mesenchyme during prostate bud formation in vivo and that this change correlates with decreased DNA methyltransferase expression in the same cell population during the same time period. To examine the role of DNA methylation in prostate development, fetal UGSs were grown in serum-free medium and 5 alpha dihydrotestosterone (DHT) and the DNA methylation inhibitor 5'-aza-2'-deoxycytidine (5AzadC) were introduced into the medium at specific times. As a measure of prostate development, in situ hybridization was used to visualize and count Nkx3-1 mRNA positive prostatic buds. We determined that inhibiting DNA methylation when prostatic buds are being specified, accelerates the onset of prostatic bud development, increases bud number, and sensitizes the budding response to androgens. Inhibition of DNA methylation also reduces Ar DNA methylation in UGS explants and increases Ar mRNA and protein in UGS mesenchyme and epithelium. Together, these results support a novel mechanism whereby Ar DNA methylation regulates UGS androgen sensitivity to control the rate and number of prostatic buds formed, thereby establishing a developmental checkpoint.
Collapse
Affiliation(s)
- Kimberly P Keil
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lisa L Abler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jimena Laporta
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Helene M Altmann
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Bing Yang
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - David F Jarrard
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI, USA; Environmental and Molecular Toxicology, University of Wisconsin, Madison, WI, USA
| | - Laura L Hernandez
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
26
|
Schneider AJ, Branam AM, Peterson RE. Intersection of AHR and Wnt signaling in development, health, and disease. Int J Mol Sci 2014; 15:17852-85. [PMID: 25286307 PMCID: PMC4227194 DOI: 10.3390/ijms151017852] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/04/2014] [Accepted: 09/18/2014] [Indexed: 12/16/2022] Open
Abstract
The AHR (aryl hydrocarbon receptor) and Wnt (wingless-related MMTV integration site) signaling pathways have been conserved throughout evolution. Appropriately regulated signaling through each pathway is necessary for normal development and health, while dysregulation can lead to developmental defects and disease. Though both pathways have been vigorously studied, there is relatively little research exploring the possibility of crosstalk between these pathways. In this review, we provide a brief background on (1) the roles of both AHR and Wnt signaling in development and disease, and (2) the molecular mechanisms that characterize activation of each pathway. We also discuss the need for careful and complete experimental evaluation of each pathway and describe existing research that explores the intersection of AHR and Wnt signaling. Lastly, to illustrate in detail the intersection of AHR and Wnt signaling, we summarize our recent findings which show that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced disruption of Wnt signaling impairs fetal prostate development.
Collapse
Affiliation(s)
- Andrew J Schneider
- School of Pharmacy and Molecular and Environmental Toxicology Center University of Wisconsin, Madison, WI 53705, USA.
| | - Amanda M Branam
- School of Pharmacy and Molecular and Environmental Toxicology Center University of Wisconsin, Madison, WI 53705, USA.
| | - Richard E Peterson
- School of Pharmacy and Molecular and Environmental Toxicology Center University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
27
|
Kitagaki J, Miyauchi S, Xie CJ, Yamashita M, Yamada S, Kitamura M, Murakami S. Effects of the proteasome inhibitor, bortezomib, on cytodifferentiation and mineralization of periodontal ligament cells. J Periodontal Res 2014; 50:248-55. [PMID: 24948468 DOI: 10.1111/jre.12202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE The proteasome inhibitor, bortezomib, is known to induce osteoblastic differentiation in a number of cell lines, such as mesenchymal stem cells and osteoblastic precursor cells. As periodontal ligament (PDL) cells are multipotent, we examined whether bortezomib may induce the differentiation of PDL cells into hard-tissue-forming cells. MATERIAL AND METHODS A mouse PDL clone cell line, MPDL22 cells, was cultured in mineralization medium in the presence or absence of bortezomib. Expression of calcification-related genes and calcified-nodule formation were evaluated by real-time PCR and Alizarin Red staining, respectively. RESULTS Bortezomib increased the expression of calcification-related mRNAs, such as tissue nonspecific alkaline phosphatase isoenzyme (ALPase), bone sialoprotein (Bsp), runt-related transcription factor 2 (Runx2) and osteopontin, and calcified-nodule formation in MPDL22 cells. These effects were induced, in part, by increasing the cytosolic accumulation and nuclear translocation of β-catenin, leading to an increase in expression of bone morphogenetic protein (Bmp)-2, -4 and -6 mRNAs. In addition, bortezomib enhanced BMP-2-induced expression of Bsp and osteopontin mRNAs and increased calcified-nodule formation in MPDL22 cells. CONCLUSION Bortezomib induced cytodifferentiation and mineralization of PDL cells by enhancing the accumulation of β-catenin within the cytosol and the nucleus and increasing the expression of Bmp-2, -4 and -6 mRNAs. Moreover, bortezomib enhanced the BMP-2-induced cytodifferentiation and mineralization of PDL cells, suggesting that bortezomib may be efficacious for use in periodontal regeneration therapy.
Collapse
Affiliation(s)
- J Kitagaki
- Department of Periodontology, Division of Oral Biology and Disease Control, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan; Challenge to Intractable Oral Diseases, Center for Translational Dental Research, Osaka University Dental Hospital, Suita, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Schneider AJ, Moore RW, Branam AM, Abler LL, Keil KP, Mehta V, Vezina CM, Peterson RE. In utero exposure to TCDD alters Wnt signaling during mouse prostate development: linking ventral prostate agenesis to downregulated β-catenin signaling. Toxicol Sci 2014; 141:176-87. [PMID: 24928892 DOI: 10.1093/toxsci/kfu116] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes ventral prostate agenesis in C57BL/6J mice by preventing ventral prostatic budding in the embryonic urogenital sinus (UGS). TCDD (5 μg/kg, po) administered to pregnant dams on embryonic day 15.5 (E15.5) activates the aryl hydrocarbon receptor in the UGS mesenchyme, disrupting the mesenchymally derived paracrine signaling that instructs epithelial prostatic budding. How TCDD alters the mesenchymal milieu is not well understood. We previously showed that TCDD disrupts some aspects of Wnt signaling in UGSs grown in vitro. Here we provide the first comprehensive, in vivo characterization of Wnt signaling in male E16.5 UGSs during normal development, and after in utero TCDD exposure. Vehicle- and TCDD-exposed UGSs were probed by in situ hybridization to assess relative abundance and localization of RNA from 46 genes that regulate Wnt signaling. TCDD altered the staining pattern of five genes, increasing staining for Wnt10a and Wnt16 and decreasing staining for Ror2, Rspo2, and Wif1. We also used immunohistochemistry to show, for the first time, activation of β-catenin (CTNNB1) signaling in ventral basal epithelium of control UGSs at E16.5. This onset of CTNNB1 signaling occurred immediately prior to the initiation of ventral prostatic budding and is characterized by a pronounced increase in CTNNB1 nuclear localization and subsequent expression of the CTNNB1 signaling target gene, Lef1. In utero TCDD exposure prevented the onset of CTNNB1 signaling and LEF1 expression in the ventral basal epithelium, thereby elucidating a likely mechanism by which TCDD contributes to failed prostatic budding in the ventral UGS.
Collapse
Affiliation(s)
| | - Robert W Moore
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705
| | - Amanda M Branam
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705
| | - Lisa L Abler
- School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | - Kimberly P Keil
- School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | - Vatsal Mehta
- School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | - Chad M Vezina
- School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin 53706
| | | |
Collapse
|
29
|
Keil KP, Abler LL, Mehta V, Altmann HM, Laporta J, Plisch EH, Suresh M, Hernandez LL, Vezina CM. DNA methylation of E-cadherin is a priming mechanism for prostate development. Dev Biol 2014; 387:142-53. [PMID: 24503032 DOI: 10.1016/j.ydbio.2014.01.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/20/2013] [Accepted: 01/23/2014] [Indexed: 12/31/2022]
Abstract
In prostate and other epithelial cancers, E-cadherin (CDH1) is downregulated inappropriately by DNA methylation to promote an invasive phenotype. Though cancer frequently involves a reawakening of developmental signaling pathways, whether DNA methylation of Cdh1 occurs during organogenesis has not been determined. Here we show that DNA methylation of Cdh1 mediates outgrowth of developing prostate ducts. During the three-day gestational window leading up to and including prostate ductal initiation, Cdh1 promoter methylation increases and its mRNA and protein abundance decreases in epithelium giving rise to prostatic buds. DNA methylation is required for prostate specification, ductal outgrowth, and branching morphogenesis. All three endpoints are impaired by a DNA methylation inhibitor, which also decreases Cdh1 promoter methylation and increases Cdh1 mRNA and protein abundance. A CDH1 function-blocking antibody restores prostatic identity, bud outgrowth, and potentiates epithelial differentiation in the presence of the DNA methylation inhibitor. This is the first study to mechanistically link acquired changes in DNA methylation to the normal process of prostate organogenesis. We propose a novel mechanism whereby Cdh1 promoter methylation restricts Cdh1 abundance in developing prostate epithelium to create a permissive environment for prostatic bud outgrowth. Thus, DNA methylation primes the prostate primordium to respond to developmental cues mediating outgrowth, differentiation and maturation of the ductal network.
Collapse
Affiliation(s)
- Kimberly P Keil
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Lisa L Abler
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Vatsal Mehta
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Helene M Altmann
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA
| | - Jimena Laporta
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Erin H Plisch
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - M Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Laura L Hernandez
- Department of Dairy Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
30
|
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: 32] [Impact Index Per Article: 2.9] [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
|
31
|
Branam AM, Davis NM, Moore RW, Schneider AJ, Vezina CM, Peterson RE. TCDD inhibition of canonical Wnt signaling disrupts prostatic bud formation in mouse urogenital sinus. Toxicol Sci 2013; 133:42-53. [PMID: 23429912 DOI: 10.1093/toxsci/kft027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In mice, in utero exposure to 2,3,7,8-tetrachlorodibenzo-p- dioxin (TCDD) reduces the number of dorsolateral prostatic buds resulting in a smaller dorsolateral prostate and prevents formation of ventral buds culminating in ventral prostate agenesis. The genes and signaling pathways affected by TCDD that are responsible for disrupting prostate development are largely unknown. Here we show that treatment of urogenital sinus (UGS) organ cultures with known inhibitors of canonical Wnt signaling also inhibits prostatic bud formation. In support of the hypothesis that TCDD decreases canonical Wnt signaling, we identify inhibitory effects of TCDD on multiple components of the canonical Wnt signaling pathway in the UGS that temporally coincide with the inhibitory effect of TCDD on prostatic bud formation: (1) expression of R-spondins (Rspo2 and Rspo3) that promote canonical Wnt signaling is reduced; (2) expression of Lef1, Tcf1, and Wif1, established canonical Wnt target genes, is decreased; (3) expression of Lgr5, a RSPO receptor that activates canonical Wnt signaling, is reduced; and (4) expression of Dickkopfs (Dkks), inhibitors of canonical Wnt signaling, is not increased by TCDD. Thus, the TCDD-induced reduction in canonical Wnt signaling is associated with a decrease in activators (Rspo2 and Rspo3) rather than an increase in inhibitors (Dkk1 and Dkk2) of the pathway. This study focuses on determining whether treatment of TCDD-exposed UGS organ cultures with RSPO2 and/or RSPO3 is capable of rescuing the inhibitory effects of TCDD on canonical Wnt signaling and prostatic bud formation. We discovered that each RSPO alone or in combination partially rescues TCDD inhibition of both canonical Wnt signaling and prostatic bud formation.
Collapse
Affiliation(s)
- Amanda M Branam
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, USA
| | | | | | | | | | | |
Collapse
|