51
|
Pigat N, Reyes-Gomez E, Boutillon F, Palea S, Barry Delongchamps N, Koch E, Goffin V. Combined Sabal and Urtica Extracts (WS ® 1541) Exert Anti-proliferative and Anti-inflammatory Effects in a Mouse Model of Benign Prostate Hyperplasia. Front Pharmacol 2019; 10:311. [PMID: 30984003 PMCID: PMC6450068 DOI: 10.3389/fphar.2019.00311] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 03/14/2019] [Indexed: 12/19/2022] Open
Abstract
WS® 1541 is a phytopharmaceutical drug combination containing a lipophilic extract from fruits of Sabal serrulata (WS® 1473) and an aqueous ethanolic extract from roots of Urtica dioica (WS® 1031). It is approved in several countries worldwide for the treatment of lower urinary tract syndrome (LUTS) linked to benign prostate hyperplasia (BPH). Clinical studies have demonstrated the efficacy of this unique combination in the treatment of BPH-related LUTS. However, its mechanisms of action in vivo remain partly uncharacterized. The aim of this study was to take advantage of a validated mouse model of BPH to better characterize its growth-inhibitory and anti-inflammatory properties. We used the probasin–prolactin (Pb-PRL) transgenic mouse model in which prostate-specific overexpression of PRL results in several features of the human disease including tissue hypertrophy, epithelial hyperplasia, increased stromal cellularity, inflammation, and LUTS. Six-month-old heterozygous Pb-PRL male mice were randomly distributed to five groups (11–12 animals/group) orally treated for 28 consecutive days with WS® 1541 (300, 600, or 900 mg/kg/day), the 5α-reductase inhibitor finasteride used as reference (5 mg/kg/day) or vehicle (olive oil 5 ml/kg/day). Administration of WS® 1541 was well tolerated and caused a dose-dependent reduction of prostate weight (vs. vehicle) that was statistically significant at the two highest doses. This effect was accompanied by a reduction in prostate cell proliferation as assessed by lower Ki-67 expression (qPCR and immunohistochemistry). In contrast, finasteride had no or only a mild effect on these parameters. The growth-inhibitory activity of WS® 1541 was accompanied by a strong anti-inflammatory effect as evidenced by the reduced infiltration of cells expressing the leukocyte common antigen CD45. In sharp contrast, finasteride significantly increased the prostate inflammatory status according to this readout. Molecular profiling (qPCR) of 23 selected pro-inflammatory genes confirmed the strong anti-inflammatory potency of WS® 1541 compared to finasteride. Since treatment of WS® 1541 did not interfere with transgene expression and activity in the prostate of Pb-PRL mice, the effects observed in this study are entirely attributable to the intrinsic pharmacological action of the drug combination.
Collapse
Affiliation(s)
- Natascha Pigat
- PRL/GH Pathophysiology Laboratory, Institut Necker Enfants Malades, Unit 1151, Inserm, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| | - Edouard Reyes-Gomez
- Unité d'Histologie et d'Anatomie Pathologique, Laboratoire d'Anatomo-Cytopathologie, Biopôle Alfort, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France.,Inserm, U955 - IMRB, Ecole Nationale Vétérinaire d'Alfort, UPEC, Maisons-Alfort, France
| | - Florence Boutillon
- PRL/GH Pathophysiology Laboratory, Institut Necker Enfants Malades, Unit 1151, Inserm, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| | | | - Nicolas Barry Delongchamps
- PRL/GH Pathophysiology Laboratory, Institut Necker Enfants Malades, Unit 1151, Inserm, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France.,Urology Department, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Egon Koch
- Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany
| | - Vincent Goffin
- PRL/GH Pathophysiology Laboratory, Institut Necker Enfants Malades, Unit 1151, Inserm, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| |
Collapse
|
52
|
Yang BY, Jiang CY, Dai CY, Zhao RZ, Wang XJ, Zhu YP, Qian YX, Yin FL, Fu XY, Jing YF, Han BM, Xia SJ, Ruan Y. 5-ARI induces autophagy of prostate epithelial cells through suppressing IGF-1 expression in prostate fibroblasts. Cell Prolif 2019; 52:e12590. [PMID: 30883989 PMCID: PMC6536403 DOI: 10.1111/cpr.12590] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/11/2019] [Accepted: 01/27/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives 5α‐reductase inhibitor (5‐ARI) is a commonly used medicine in the treatment of lower urinary tract symptoms (LUTS) associated with benign prostatic hyperplasia (BPH). Our study mainly focuses on the mechanism of BPH development after 5ARI treatment. Materials and Methods Prostate specimens from patients were collected. Insulin‐like growth factor 1 (IGF‐1), Beclin‐1, LC3 levels, was analysed by immunohistochemistry. The role IGF‐1 on autophagic flux in prostate epithelial cells was studied. Additionally, effect of autophagy on recombinant grafts consisting of prostate stromal and epithelial cells in nude mice was investigated. Results We demonstrated that IGF‐1 expression is down‐regulated in prostate fibroblasts after long‐term 5‐ARI application. A decrease in IGF‐1 levels was found to activate autophagic flux through the mTOR pathway in prostate epithelial cells, while the inhibition of IGF‐1 receptor function induced autophagy in prostate epithelial cells. In addition, we revealed that blocking autophagic flux initiation can reduce the volume of recombinant grafts in vivo. Finally, our findings suggest that long‐term 5‐ARI application reduces IGF‐1 secretion by prostatic stromal cells, thereby inducing autophagy of prostatic epithelial cells, which is one of the mechanisms underlying BPH pathogenesis and progression. Conclusions Focusing on the autophagy induced by low levels of IGF‐1 in prostatic epithelial cells, after elucidating AR signalling impairment of prostate stromal cells, might provide a novel strategy for the treatment and prevention of BPH development.
Collapse
Affiliation(s)
- Bo-Yu Yang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Yi Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Yun Dai
- Institute of Translational Medicine, Shanghai General Hospital, Shanghai, China
| | - Rui-Zhe Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xing-Jie Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Ping Zhu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Xin Qian
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fu-Li Yin
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang-Yu Fu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Feng Jing
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bang-Min Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu-Jie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Ruan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
53
|
Arriaga JM, Abate-Shen C. Genetically Engineered Mouse Models of Prostate Cancer in the Postgenomic Era. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a030528. [PMID: 29661807 DOI: 10.1101/cshperspect.a030528] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Recent genomic sequencing analyses have unveiled the spectrum of genomic alterations that occur in primary and advanced prostate cancer, raising the question of whether the corresponding genes are functionally relevant for prostate tumorigenesis, and whether such functions are associated with particular disease stages. In this review, we describe genetically engineered mouse models (GEMMs) of prostate cancer, focusing on those that model genomic alterations known to occur in human prostate cancer. We consider whether the phenotypes of GEMMs based on gain or loss of function of the relevant genes provide reliable counterparts to study the predicted consequences of the corresponding genomic alterations as occur in human prostate cancer, and we discuss exceptions in which the GEMMs do not fully emulate the expected phenotypes. Last, we highlight future directions for the generation of new GEMMs of prostate cancer and consider how we can use GEMMs most effectively to decipher the biological and molecular mechanisms of disease progression, as well as to tackle clinically relevant questions.
Collapse
Affiliation(s)
- Juan M Arriaga
- Departments of Urology, Medicine, Systems Biology, and Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| | - Cory Abate-Shen
- Departments of Urology, Medicine, Systems Biology, and Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
54
|
Abstract
Comprehensive knowledge of the normal prostate epithelial lineage hierarchy is a prerequisite to investigate the identity of the cells of origin for prostate cancer. The basal and luminal cells constitute most of the prostate epithelium and have been the major focuses of the study on the cells of origin for prostate cancer. Much progress has been made during the past few decades, mainly using mouse models, to understand the inter-lineage relationship and intra-lineage heterogeneity in adults as well as the lineage plasticity during conditions of stress. These studies have concluded that the adult mouse prostate basal and luminal cells are largely independently sustained under physiological conditions, but both types of cells possess the capacity for bipotent differentiation under stress or artificial experimental conditions. However, the existence or the identity of the putative progenitors within each lineage warrants further investigation. Whether the human prostate lineage hierarchy is completely the same as that of the mouse remains uncertain. Experiments from independent groups have demonstrated that both types of cells in mice and humans can serve as targets for transformation. But controversies remain whether the disease from distinct cells of origin display different clinical behaviors. Further investigation of the intra-lineage heterogeneity will provide new insights into this issue. Understanding the identity of the cells of origin for prostate cancer will help identify novel prognostic markers for early detection of aggressive prostate cancers, provide insights into the therapeutic vulnerability of these tumors, and inspire novel therapeutic strategies.
Collapse
|
55
|
Barros-Silva JD, Linn DE, Steiner I, Guo G, Ali A, Pakula H, Ashton G, Peset I, Brown M, Clarke NW, Bronson RT, Yuan GC, Orkin SH, Li Z, Baena E. Single-Cell Analysis Identifies LY6D as a Marker Linking Castration-Resistant Prostate Luminal Cells to Prostate Progenitors and Cancer. Cell Rep 2018; 25:3504-3518.e6. [PMID: 30566873 PMCID: PMC6315111 DOI: 10.1016/j.celrep.2018.11.069] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 09/26/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022] Open
Abstract
The exact identity of castrate-resistant (CR) cells and their relation to CR prostate cancer (CRPC) is unresolved. We use single-cell gene profiling to analyze the molecular heterogeneity in basal and luminal compartments. Within the luminal compartment, we identify a subset of cells intrinsically resistant to castration with a bi-lineage gene expression pattern. We discover LY6D as a marker of CR prostate progenitors with multipotent differentiation and enriched organoid-forming capacity. Lineage tracing further reveals that LY6D+ CR luminal cells can produce LY6D- luminal cells. In contrast, in luminal cells lacking PTEN, LY6D+ cells predominantly give rise to LY6D+ tumor cells, contributing to high-grade PIN lesions. Gene expression analyses in patients' biopsies indicate that LY6D expression correlates with early disease progression, including progression to CRPC. Our studies thus identify a subpopulation of luminal progenitors characterized by LY6D expression and intrinsic castration resistance. LY6D may serve as a prognostic maker for advanced prostate cancer.
Collapse
Affiliation(s)
- João D Barros-Silva
- Prostate Oncobiology, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK
| | - Douglas E Linn
- Division of Genetics, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ivana Steiner
- Prostate Oncobiology, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK
| | - Guoji Guo
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Adnan Ali
- Prostate Oncobiology, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK
| | - Hubert Pakula
- Division of Genetics, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Garry Ashton
- Histology Unit, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK
| | - Isabel Peset
- Imaging Unit, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK
| | - Michael Brown
- Genito-Urinary Cancer Research, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Cancer Research Centre, Wilmslow Road, Manchester M20 4GJ, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK
| | - Noel W Clarke
- Genito-Urinary Cancer Research, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Cancer Research Centre, Wilmslow Road, Manchester M20 4GJ, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK; Department of Surgery, The Christie Hospital, Department of Urology, Salford Royal Hospitals, Manchester, UK
| | | | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, MA 02115, USA
| | - Stuart H Orkin
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| | - Zhe Li
- Division of Genetics, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Esther Baena
- Prostate Oncobiology, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park SK10 4TG, UK.
| |
Collapse
|
56
|
Zhang D, Zhao S, Li X, Kirk JS, Tang DG. Prostate Luminal Progenitor Cells in Development and Cancer. Trends Cancer 2018; 4:769-783. [PMID: 30352679 PMCID: PMC6212301 DOI: 10.1016/j.trecan.2018.09.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) has a predominantly luminal phenotype. Basal cells were previously identified as a cell of origin for PCa, but increasing evidence implicates luminal cells as a preferred cell of origin for PCa, as well as key drivers of tumor development and progression. Prostate luminal cells are understudied compared with basal cells. In this review, we describe the contribution of prostate luminal progenitor (LP) cells to luminal cell development and their role in prostate development, androgen-mediated regeneration of castrated prostate, and tumorigenesis. We also discuss the potential value of LP transcriptomics to identify new targets and therapies to treat aggressive PCa. Finally, we propose future research directions focusing on molecular mechanisms underlying LP cell biology and heterogeneity in normal and diseased prostate.
Collapse
Affiliation(s)
- Dingxiao Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Jason S Kirk
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
57
|
Staal J, Beyaert R. Inflammation and NF-κB Signaling in Prostate Cancer: Mechanisms and Clinical Implications. Cells 2018; 7:E122. [PMID: 30158439 PMCID: PMC6162478 DOI: 10.3390/cells7090122] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer is a highly prevalent form of cancer that is usually slow-developing and benign. Due to its high prevalence, it is, however, still the second most common cause of death by cancer in men in the West. The higher prevalence of prostate cancer in the West might be due to elevated inflammation from metabolic syndrome or associated comorbidities. NF-κB activation and many other signals associated with inflammation are known to contribute to prostate cancer malignancy. Inflammatory signals have also been associated with the development of castration resistance and resistance against other androgen depletion strategies, which is a major therapeutic challenge. Here, we review the role of inflammation and its link with androgen signaling in prostate cancer. We further describe the role of NF-κB in prostate cancer cell survival and proliferation, major NF-κB signaling pathways in prostate cancer, and the crosstalk between NF-κB and androgen receptor signaling. Several NF-κB-induced risk factors in prostate cancer and their potential for therapeutic targeting in the clinic are described. A better understanding of the inflammatory mechanisms that control the development of prostate cancer and resistance to androgen-deprivation therapy will eventually lead to novel treatment options for patients.
Collapse
Affiliation(s)
- Jens Staal
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
58
|
Thomas-Jardin SE, Kanchwala MS, Jacob J, Merchant S, Meade RK, Gahnim NM, Nawas AF, Xing C, Delk NA. Identification of an IL-1-induced gene expression pattern in AR + PCa cells that mimics the molecular phenotype of AR - PCa cells. Prostate 2018; 78. [PMID: 29527701 PMCID: PMC5893432 DOI: 10.1002/pros.23504] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND In immunosurveillance, bone-derived immune cells infiltrate the tumor and secrete inflammatory cytokines to destroy cancer cells. However, cancer cells have evolved mechanisms to usurp inflammatory cytokines to promote tumor progression. In particular, the inflammatory cytokine, interleukin-1 (IL-1), is elevated in prostate cancer (PCa) patient tissue and serum, and promotes PCa bone metastasis. IL-1 also represses androgen receptor (AR) accumulation and activity in PCa cells, yet the cells remain viable and tumorigenic; suggesting that IL-1 may also contribute to AR-targeted therapy resistance. Furthermore, IL-1 and AR protein levels negatively correlate in PCa tumor cells. Taken together, we hypothesize that IL-1 reprograms AR positive (AR+ ) PCa cells into AR negative (AR- ) PCa cells that co-opt IL-1 signaling to ensure AR-independent survival and tumor progression in the inflammatory tumor microenvironment. METHODS LNCaP and PC3 PCa cells were treated with IL-1β or HS-5 bone marrow stromal cell (BMSC) conditioned medium and analyzed by RNA sequencing and RT-QPCR. To verify genes identified by RNA sequencing, LNCaP, MDA-PCa-2b, PC3, and DU145 PCa cell lines were treated with the IL-1 family members, IL-1α or IL-1β, or exposed to HS-5 BMSC in the presence or absence of Interleukin-1 Receptor Antagonist (IL-1RA). Treated cells were analyzed by western blot and/or RT-QPCR. RESULTS Comparative analysis of sequencing data from the AR+ LNCaP PCa cell line versus the AR- PC3 PCa cell line reveals an IL-1-conferred gene suite in LNCaP cells that is constitutive in PC3 cells. Bioinformatics analysis of the IL-1 regulated gene suite revealed that inflammatory and immune response pathways are primarily elicited; likely facilitating PCa cell survival and tumorigenicity in an inflammatory tumor microenvironment. CONCLUSIONS Our data supports that IL-1 reprograms AR+ PCa cells to mimic AR- PCa gene expression patterns that favor AR-targeted treatment resistance and cell survival.
Collapse
Affiliation(s)
| | - Mohammed S. Kanchwala
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Joan Jacob
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Sana Merchant
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Rachel K. Meade
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Nagham M. Gahnim
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Afshan F. Nawas
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Chao Xing
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nikki A. Delk
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| |
Collapse
|
59
|
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: 1.6] [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
|
60
|
Chua CW, Epsi NJ, Leung EY, Xuan S, Lei M, Li BI, Bergren SK, Hibshoosh H, Mitrofanova A, Shen MM. Differential requirements of androgen receptor in luminal progenitors during prostate regeneration and tumor initiation. eLife 2018; 7:28768. [PMID: 29334357 PMCID: PMC5807048 DOI: 10.7554/elife.28768] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022] Open
Abstract
Master regulatory genes of tissue specification play key roles in stem/progenitor cells and are often important in cancer. In the prostate, androgen receptor (AR) is a master regulator essential for development and tumorigenesis, but its specific functions in prostate stem/progenitor cells have not been elucidated. We have investigated AR function in CARNs (CAstration-Resistant Nkx3.1-expressing cells), a luminal stem/progenitor cell that functions in prostate regeneration. Using genetically--engineered mouse models and novel prostate epithelial cell lines, we find that progenitor properties of CARNs are largely unaffected by AR deletion, apart from decreased proliferation in vivo. Furthermore, AR loss suppresses tumor formation after deletion of the Pten tumor suppressor in CARNs; however, combined Pten deletion and activation of oncogenic Kras in AR-deleted CARNs result in tumors with focal neuroendocrine differentiation. Our findings show that AR modulates specific progenitor properties of CARNs, including their ability to serve as a cell of origin for prostate cancer.
Collapse
Affiliation(s)
- Chee Wai Chua
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Nusrat J Epsi
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers, The State University of New Jersey, Newark, United States.,Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, United States
| | - Eva Y Leung
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Shouhong Xuan
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Ming Lei
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Bo I Li
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Sarah K Bergren
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States
| | - Antonina Mitrofanova
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers, The State University of New Jersey, Newark, United States.,Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, United States
| | - Michael M Shen
- Department of Medicine, Columbia University Medical Center, New York, United States.,Department of Genetics and Development, Columbia University Medical Center, New York, United States.,Department of Urology, Columbia University Medical Center, New York, United States.,Department of Systems Biology, Columbia University Medical Center, New York, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, United States
| |
Collapse
|
61
|
Albini A, Bruno A, Bassani B, D’Ambrosio G, Pelosi G, Consonni P, Castellani L, Conti M, Cristoni S, Noonan DM. Serum Steroid Ratio Profiles in Prostate Cancer: A New Diagnostic Tool Toward a Personalized Medicine Approach. Front Endocrinol (Lausanne) 2018; 9:110. [PMID: 29674995 PMCID: PMC5895774 DOI: 10.3389/fendo.2018.00110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/05/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Serum steroids are crucial molecules altered in prostate cancer (PCa). Mass spectrometry (MS) is currently the elected technology for the analysis of steroids in diverse biological samples. Steroids have complex biological pathways and stoichiometry and it is important to evaluate their quantitative ratio. MS applications to patient hormone profiling could lead to a diagnostic approach. METHODS Here, we employed the Surface Activated Chemical Ionization-Electrospray-NIST (SANIST) developed in our laboratories, to obtain quantitative serum steroid ratio relationship profiles with a machine learning Bayesian model to discriminate patients with PCa. The approach is focused on steroid relationship profiles and disease association. RESULTS A pilot study on patients affected by PCa, benign prostate hypertrophy (BPH), and control subjects [prostate-specific antigen (PSA) lower than 2.5 ng/mL] was done in order to investigate the classification performance of the SANIST platform. The steroid profiles of 71 serum samples (31 controls, 20 patients with PCa and 20 subjects with benign prostate hyperplasia) were evaluated. The levels of 10 steroids were quantitated on the SANIST platform: Aldosterone, Corticosterone, Cortisol, 11-deoxycortisol, Androstenedione, Testosterone, dehydroepiandrosterone, dehydroepiandrosterone sulfate (DHEAS), 17-OH-Progesterone and Progesterone. We performed both traditional and a machine learning analysis. CONCLUSION We show that the machine learning approach based on the steroid relationships developed here was much more accurate than the PSA, DHEAS, and direct absolute value match method in separating the PCa, BPH and control subjects, increasing the sensitivity to 90% and specificity to 84%. This technology, if applied in the future to a larger number of samples will be able to detect the individual enzymatic disequilibrium associated with the steroid ratio and correlate it with the disease. This learning machine approach could be valid in a personalized medicine setting.
Collapse
Affiliation(s)
- Adriana Albini
- IRCCS MultiMedica, Milan, Italy
- Department of Medicine and Surgery, University Milano-Bicocca, Milan, Italy
| | | | | | | | - Giuseppe Pelosi
- IRCCS MultiMedica, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | | | | | - Simone Cristoni
- I.S.B.—Ion Source & Biotechnologies, Bresso, Italy
- *Correspondence: Simone Cristoni,
| | - Douglas M. Noonan
- IRCCS MultiMedica, Milan, Italy
- Department of Biotechnologies and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
62
|
Histone 2B-GFP Label-Retaining Prostate Luminal Cells Possess Progenitor Cell Properties and Are Intrinsically Resistant to Castration. Stem Cell Reports 2017; 10:228-242. [PMID: 29276153 PMCID: PMC5768933 DOI: 10.1016/j.stemcr.2017.11.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 12/19/2022] Open
Abstract
The existence of slow-cycling luminal cells in the prostate has been suggested, but their identity and functional properties remain unknown. Using a bigenic mouse model to earmark, isolate, and characterize the quiescent stem-like cells, we identify a label-retaining cell (LRC) population in the luminal cell layer as luminal progenitors. Molecular and biological characterizations show that these luminal LRCs are significantly enriched in the mouse proximal prostate, exhibit relative dormancy, display bipotency in both in vitro and in vivo assays, and express a stem/progenitor gene signature with resemblance to aggressive prostate cancer. Importantly, these LRCs, compared with bulk luminal cells, maintain a lower level of androgen receptor (AR) expression and are less androgen dependent and also castration resistant in vivo. Finally, analysis of phenotypic markers reveals heterogeneity within the luminal progenitor cell pool. Our study establishes luminal LRCs as progenitors that may serve as a cellular origin for castration-resistant prostate cancer. A bigenic mouse model to study prostatic slow-cycling luminal epithelial cells Prostate label-retaining cells (LRCs) exhibit stem/progenitor cell activities Luminal LRCs are developmentally bipotent and display a progenitor gene signature Luminal LRCs resist castration and molecularly resemble aggressive prostate cancer
Collapse
|
63
|
Wang Z, Hu L, Salari K, Bechis SK, Ge R, Wu S, Rassoulian C, Pham J, Wu CL, Tabatabaei S, Strand DW, Olumi AF. Androgenic to oestrogenic switch in the human adult prostate gland is regulated by epigenetic silencing of steroid 5α-reductase 2. J Pathol 2017; 243:457-467. [PMID: 28940538 DOI: 10.1002/path.4985] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/19/2017] [Accepted: 09/10/2017] [Indexed: 12/25/2022]
Abstract
Benign prostatic hyperplasia is the most common proliferative abnormality of the prostate. All men experience some prostatic growth as they age, but the rate of growth varies among individuals. Steroid 5α-reductase 2 (SRD5A2) is a critical enzyme for prostatic development and growth. Previous work indicates that one-third of adult prostatic samples do not express SRD5A2, secondary to epigenetic modifications. Here we show that the level of oestradiol is dramatically elevated, concomitant with significant upregulation of oestrogen response genes, in prostatic samples with methylation at the SRD5A2 promoter. The phosphorylation of oestrogen receptor-α in prostatic stroma is upregulated when SRD5A2 expression is absent. We show that tumour necrosis factor (TNF)-α suppresses SRD5A2 mRNA and protein expression, and simultaneously promotes expression of aromatase, the enzyme responsible for conversion of testosterone to oestradiol. Concomitant suppression of SRD5A2 and treatment with TNF-α synergistically upregulate the aromatase levels. The data suggest that, in the absence of prostatic SRD5A2, there is an androgenic to oestrogenic switch. These findings have broad implications for choosing appropriate classes of medications for the management of benign and malignant prostatic diseases. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Libing Hu
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Urology, Yan'an Affiliated hospital of Kunming Medical University, Yunnan, PR China
| | - Keyan Salari
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Seth K Bechis
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rongbin Ge
- Department of Pathology, University of Massachusetts Medical Center, Worcester, MA, USA
| | - Shulin Wu
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cyrus Rassoulian
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan Pham
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chin-Lee Wu
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shahin Tabatabaei
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Douglas W Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Aria F Olumi
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
64
|
Rycaj K, Tang DG. Molecular determinants of prostate cancer metastasis. Oncotarget 2017; 8:88211-88231. [PMID: 29152153 PMCID: PMC5675705 DOI: 10.18632/oncotarget.21085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 08/31/2017] [Indexed: 12/12/2022] Open
Abstract
Metastatic cancer remains largely incurable and fatal. The general course of cancer, from the initiation of primary tumor formation and progression to metastasis, is a multistep process wherein tumor cells at each step must display specific phenotypic features. Distinctive capabilities required for primary tumor initiation and growth form the foundation, and sometimes may remain critical, for subsequent metastases. These phenotypic features must remain easily malleable during the acquisition of additional capabilities unique and essential to the metastatic process such as dissemination to distant tissues wherein tumor cells interact with foreign microenvironments. Thus, the metastatic phenotype is a culmination of multiple genetic and epigenetic alterations and subsequent selection for favorable traits under the pressure of ever-changing tumor microenvironments. Although our understanding of the molecular programs that drive cancer metastasis are incomplete, increasing evidence suggests that successful metastatic colonization relies on the dissemination of cancer stem cells (CSCs) with tumor-regenerating capacity and adaptive programs for survival in distant organs. In the past 2-3 years, a myriad of novel molecular regulators and determinants of prostate cancer metastasis have been reported, and in this Perspective, we comprehensively review this body of literature and summarize recent findings regarding cell autonomous molecular mechanisms critical for prostate cancer metastasis.
Collapse
Affiliation(s)
- Kiera Rycaj
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Dean G. Tang
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
- Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
65
|
Boivin FJ, Schmidt-Ott KM. Transcriptional mechanisms coordinating tight junction assembly during epithelial differentiation. Ann N Y Acad Sci 2017. [PMID: 28636799 DOI: 10.1111/nyas.13367] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epithelial tissues form a selective barrier via direct cell-cell interactions to separate and establish concentration gradients between the different compartments of the body. Proper function and formation of this barrier rely on the establishment of distinct intercellular junction complexes. These complexes include tight junctions, adherens junctions, desmosomes, and gap junctions. The tight junction is by far the most diverse junctional complex in the epithelial barrier. Its composition varies greatly across different epithelial tissues to confer various barrier properties. Thus, epithelial cells rely on tightly regulated transcriptional mechanisms to ensure proper formation of the epithelial barrier and to achieve tight junction diversity. Here, we review different transcriptional mechanisms utilized during embryogenesis and disease development to promote tight junction assembly and maintenance of intercellular barrier integrity. We focus particularly on the Grainyhead-like transcription factors and ligand-activated nuclear hormone receptors, two central families of proteins in epithelialization.
Collapse
Affiliation(s)
- Felix J Boivin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology, Charité Medical University, Berlin, Germany
| |
Collapse
|
66
|
Henry GH, Loof N, Strand DW. OMIP-040: Optimized gating of human prostate cellular subpopulations. Cytometry A 2017; 91:1147-1149. [PMID: 28834328 DOI: 10.1002/cyto.a.23187] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Gervaise H Henry
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nicolas Loof
- The Moody Foundation Flow Cytometry Facility, Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Douglas W Strand
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
67
|
Strand DW, Costa DN, Francis F, Ricke WA, Roehrborn CG. Targeting phenotypic heterogeneity in benign prostatic hyperplasia. Differentiation 2017; 96:49-61. [PMID: 28800482 DOI: 10.1016/j.diff.2017.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 02/07/2023]
Abstract
Benign prostatic hyperplasia and associated lower urinary tract symptoms remain difficult to treat medically, resulting in hundreds of thousands of surgeries performed annually in elderly males. New therapies have not improved clinical outcomes since alpha blockers and 5 alpha reductase inhibitors were introduced in the 1990s. An underappreciated confounder to identifying novel targets is pathological heterogeneity. Individual patients display unique phenotypes, composed of distinct cell types. We have yet to develop a cellular or molecular understanding of these unique phenotypes, which has led to failure in developing targeted therapies for personalized medicine. This review covers the strategic experimental approach to unraveling the cellular pathogenesis of discrete BPH phenotypes and discusses how to incorporate these findings into the clinic to improve outcomes.
Collapse
Affiliation(s)
- Douglas W Strand
- Department of Urology, University of Texas Southwestern Medical Center, USA.
| | - Daniel N Costa
- Department of Radiology, University of Texas Southwestern Medical Center, USA
| | - Franto Francis
- Department of Pathology, University of Texas Southwestern Medical Center, USA
| | - William A Ricke
- Department of Urology, University of Wisconsin School of Medicine, USA
| | - Claus G Roehrborn
- Department of Urology, University of Texas Southwestern Medical Center, USA
| |
Collapse
|
68
|
Henry G, Malewska A, Mauck R, Gahan J, Hutchinson R, Torrealba J, Francis F, Roehrborn C, Strand D. Molecular pathogenesis of human prostate basal cell hyperplasia. Prostate 2017; 77:1344-1355. [PMID: 28795417 PMCID: PMC5580247 DOI: 10.1002/pros.23394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/12/2017] [Indexed: 11/09/2022]
Abstract
BACKGROUND Understanding the molecular pathogenesis of distinct phenotypes in human benign prostatic hyperplasia (BPH) is essential to improving therapeutic intervention. Current therapies target smooth muscle and luminal epithelia for relief of lower urinary tract symptoms (LUTS) due to BPH, but basal cell hyperplasia (BCH) remains untargeted. The incidence of has been reported at 8-10%, but a molecular and cellular characterization has not been performed on this phenotype. METHODS Using freshly digested tissue from surgical specimens, we performed RNA-seq analysis of flow cytometry-purified basal epithelia from 3 patients with and 4 patients without a majority BCH phenotype. qPCR was performed on 28 genes identified as significant from 13 non-BCH and 7 BCH specimens to confirm transcriptomic analysis. IHC was performed on several non-BCH and BCH specimens for 3 proteins identified as significant by transcriptomic analysis. RESULTS A total of 141 human BPH specimens were analyzed for the presence of BCH. Clinical characteristics of non-BCH and BCH cohorts revealed no significant differences in age, PSA, prostate volume, medical treatment, or comorbidities. Quantitation of cellular subsets by flow cytometry in 11 BCH patients vs. 11 non-BCH patients demonstrated a significant increase in the ratio of basal to luminal epithelia in patients with BCH (P <0.05), but no significant differences in the total number of leukocytes. RNA-seq data from flow cytometry isolated basal epithelia from patients with and without BCH were subjected to gene set enrichment analysis of differentially expressed genes, which revealed increased expression of members of the epidermal differentiation complex. Transcriptomic data were complemented by immunohistochemistry for members of the epidermal differentiation complex, revealing a morphological similarity to other stratified squamous epithelial layers. CONCLUSIONS Increased expression of epidermal differentiation complex members and altered epithelial stratification resembles the progression of other metaplastic diseases. These data provide insight into the plasticity of the human prostate epithelium and suggest a classification of basal cell hyperplasia as a metaplasia.
Collapse
Affiliation(s)
- Gervaise Henry
- Department of Urology, UT Southwestern Medical Center, Dallas,
Texas
| | - Alicia Malewska
- Department of Urology, UT Southwestern Medical Center, Dallas,
Texas
| | - Ryan Mauck
- Department of Urology, UT Southwestern Medical Center, Dallas,
Texas
| | - Jeffrey Gahan
- Department of Urology, UT Southwestern Medical Center, Dallas,
Texas
| | - Ryan Hutchinson
- Department of Urology, UT Southwestern Medical Center, Dallas,
Texas
| | - Jose Torrealba
- Department of Pathology, UT Southwestern Medical Center, Dallas,
Texas
| | - Franto Francis
- Department of Pathology, UT Southwestern Medical Center, Dallas,
Texas
| | - Claus Roehrborn
- Department of Urology, UT Southwestern Medical Center, Dallas,
Texas
| | - Douglas Strand
- Department of Urology, UT Southwestern Medical Center, Dallas,
Texas
| |
Collapse
|
69
|
Young MRI. Redirecting the focus of cancer immunotherapy to premalignant conditions. Cancer Lett 2017; 391:83-88. [PMID: 28130162 PMCID: PMC5925415 DOI: 10.1016/j.canlet.2017.01.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/18/2022]
Abstract
Much progress has been made in introducing immunological treatment approaches for cancer, with lessons learned from both the successes and failures of immunotherapy. Among the challenges of immunotherapeutic approaches for cancer are the multitudes of mechanisms by which cancers are known to subvert the immune defenses. This has led to the incorporation into the immunotherapeutic arsenal strategies by which to overcome the cancer’s immunological blockades. What has been only superficially explored is the immunological milieu of premalignant lesions and the possibility of immunological approaches for the treatment of premalignant lesions so as to prevent secondary premalignant lesions and their progression to cancer. This review discusses the immunological environment associated with premalignant lesions, and the possible missed opportunity of utilizing immunological treatment strategies in the less hostile environment of premalignant lesions as compared to the immune subversive cancer environment.
Collapse
Affiliation(s)
- M Rita I Young
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA; Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
70
|
|