1
|
Len-Tayon K, Metzger D, Laverny G. [New insights of vitamin D-based therapy for prostate cancer]. Med Sci (Paris) 2025; 41:154-159. [PMID: 40028953 DOI: 10.1051/medsci/2025011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Prostate cancer is the third leading cause of cancer-related death among men in developed countries. Hormone and chemotherapy are the standard of care for advanced prostate cancer, but most patients develop resistance. Therefore, new therapeutic strategies are needed to improve the clinical management of prostate cancer. Low circulating levels of the secosteroid hormone vitamin D, and reduced expression of its receptor in prostate epithelial cells correlate with prostate cancer severity. However, the mechanisms underlying the anticancer effects of vitamin D are poorly understood. Our recent work demonstrates the therapeutic effects of a vitamin D analog on the tumor microenvironment in Pten(i)pe-/- mice, a mouse model of prostate cancer. In addition, we have shown that the combination of a vitamin D analog with the chemotherapeutic agent docetaxel, overcomes chemoresistance in primary prostate cancer spheroids, and in xenografts derived from a patient with prostate cancer resistant to docetaxel and to androgen deprivation. Therefore, therapeutic strategies based on the use of vitamin D analogs represent new avenues for advanced prostate cancer.
Collapse
Affiliation(s)
- Kateryna Len-Tayon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch-Graffenstaden, France - CNRS UMR 7104, Illkirch-Graffenstaden, France - Inserm U1258, Illkirch-Graffenstaden, France - Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch-Graffenstaden, France - CNRS UMR 7104, Illkirch-Graffenstaden, France - Inserm U1258, Illkirch-Graffenstaden, France - Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Gilles Laverny
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch-Graffenstaden, France - CNRS UMR 7104, Illkirch-Graffenstaden, France - Inserm U1258, Illkirch-Graffenstaden, France - Université de Strasbourg, Illkirch-Graffenstaden, France
| |
Collapse
|
2
|
Chen H, Fang S, Zhu X, Liu H. Cancer-associated fibroblasts and prostate cancer stem cells: crosstalk mechanisms and implications for disease progression. Front Cell Dev Biol 2024; 12:1412337. [PMID: 39092186 PMCID: PMC11291335 DOI: 10.3389/fcell.2024.1412337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
The functional heterogeneity and ecological niche of prostate cancer stem cells (PCSCs), which are major drivers of prostate cancer development and treatment resistance, have attracted considerable research attention. Cancer-associated fibroblasts (CAFs), which are crucial components of the tumor microenvironment (TME), substantially affect PCSC stemness. Additionally, CAFs promote PCSC growth and survival by releasing signaling molecules and modifying the surrounding environment. Conversely, PCSCs may affect the characteristics and behavior of CAFs by producing various molecules. This crosstalk mechanism is potentially crucial for prostate cancer progression and the development of treatment resistance. Using organoids to model the TME enables an in-depth study of CAF-PCSC interactions, providing a valuable preclinical tool to accurately evaluate potential target genes and design novel treatment strategies for prostate cancer. The objective of this review is to discuss the current research on the multilevel and multitarget regulatory mechanisms underlying CAF-PCSC interactions and crosstalk, aiming to inform therapeutic approaches that address challenges in prostate cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Hao Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Sharkey C, Long X, Al-Faouri R, Strand D, Olumi AF, Wang Z. Enhanced prostatic Esr1 + luminal epithelial cells in the absence of SRD5A2. J Pathol 2024; 263:300-314. [PMID: 38606616 PMCID: PMC11166526 DOI: 10.1002/path.6283] [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: 12/15/2023] [Revised: 02/07/2024] [Accepted: 03/07/2024] [Indexed: 04/13/2024]
Abstract
Steroid 5α reductase 2 (SRD5A2) converts testosterone to dihydrotestosterone and is crucial for prostatic development. 5α reductase inhibitors (5ARI) reduce prostate size in benign prostate hyperplasia (BPH) and ameliorate lower urinary tract symptoms secondary to BPH. However, the mechanisms of 5ARI functioning are still not fully understood. Here, we used a Srd5a2-/- mouse model and employed single-cell RNA sequencing to explore the impact of SRD5A2 absence on prostate cellular heterogeneity. Significant alterations in luminal epithelial cell (LE) populations were observed, alongside an increased proportion and proliferative phenotype of estrogen receptor 1 (ESR1)+ LE2 cells, following an SRD5A2-independent ESR1 differentiation trajectory. LE2 cells exhibited enhanced estrogen response gene signatures, suggesting an alternative pathway for prostate growth when SRD5A2 is absent. Human prostate biopsy analysis revealed an inverse correlation between the expressions of SRD5A2 and LE2 markers (ESR1/PKCα), and an inverse correlation between SRD5A2 and the clinical efficiency of 5ARI. These findings provide insights into 5ARI resistance mechanisms and potential alternative therapies for BPH-related lower urinary tract symptoms. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Christina Sharkey
- Department of Surgery, Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xingbo Long
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Ra’ad Al-Faouri
- Department of Surgery, Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Douglas Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Aria F. Olumi
- Department of Surgery, Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zongwei Wang
- Department of Surgery, Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Sha K, Zhang R, Maolake A, Singh S, Chatta G, Eng KH, Nastiuk KL, Krolewski JJ. Androgen deprivation triggers a cytokine signaling switch to induce immune suppression and prostate cancer recurrence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.01.569685. [PMID: 38405929 PMCID: PMC10888871 DOI: 10.1101/2023.12.01.569685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Androgen deprivation therapy (ADT) is an effective but not curative treatment for advanced and recurrent prostate cancer (PC). We investigated the mechanisms controlling the response to androgen-deprivation by surgical castration in genetically-engineered mouse models (GEMM) of PC, using high frequency ultrasound imaging to rigorously measure tumor volume. Castration initially causes almost all tumors to shrink in volume, but many tumors subsequently recur within 5-10 weeks. Blockade of tumor necrosis factor (TNF) signaling a few days in advance of castration surgery, using a TNFR2 ligand trap, prevents regression in a PTEN-deficient GEMM. Following tumor regression, a basal stem cell-like population within the tumor increases along with TNF protein levels. Tumor cell lines in culture recapitulate these in vivo observations, suggesting that basal stem cells are the source of TNF. When TNF signaling blockade is administered immediately prior to castration, tumors regress but recurrence is prevented, implying that a late wave of TNF secretion within the tumor, which coincides with the expression of NFkB regulated genes, drives recurrence. The inhibition of signaling downstream of one NFkB-regulated protein, chemokine C-C motif ligand 2 (CCL2), prevents post-castration tumor recurrence, phenocopying post-castration (late) TNF signaling blockade. CCL2 was originally identified as a macrophage chemoattractant and indeed at late times after castration gene sets related to chemotaxis and migration are up-regulated. Importantly, enhanced CCL2 signaling during the tumor recurrence phase coincides with an increase in pro-tumorigenic macrophages and a decrease in CD8 T cells, suggesting that recurrence is driven at least in part by tumor immunosuppression. In summary, we demonstrate that a therapy-induced switch in TNF signaling, a consequence of the increased stem cell-like character of the residual tumor cells surviving ADT, induces an immunosuppressive tumor microenvironment and concomitant tumor recurrence.
Collapse
|
5
|
Dos Santos L, Carbone F, Pacreau E, Diarra S, Luka M, Pigat N, Baures M, Navarro E, Anract J, Barry Delongchamps N, Cagnard N, Bost F, Nemazanyy I, Petitjean O, Hamaï A, Ménager M, Palea S, Guidotti JE, Goffin V. Cell Plasticity in a Mouse Model of Benign Prostate Hyperplasia Drives Amplification of Androgen-Independent Epithelial Cell Populations Sensitive to Antioxidant Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:30-51. [PMID: 37827216 DOI: 10.1016/j.ajpath.2023.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/23/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Benign prostate hyperplasia (BPH) is caused by the nonmalignant enlargement of the transition zone of the prostate gland, leading to lower urinary tract symptoms. Although current medical treatments are unsatisfactory in many patients, the limited understanding of the mechanisms driving disease progression prevents the development of alternative therapeutic strategies. The probasin-prolactin (Pb-PRL) transgenic mouse recapitulates many histopathological features of human BPH. Herein, these alterations parallel urodynamic disturbance reminiscent of lower urinary tract symptoms. Single-cell RNA-sequencing analysis of Pb-PRL mouse prostates revealed that their epithelium mainly includes low-androgen signaling cell populations analogous to Club/Hillock cells enriched in the aged human prostate. These intermediate cells are predicted to result from the reprogramming of androgen-dependent luminal cells. Pb-PRL mouse prostates exhibited increased vulnerability to oxidative stress due to reduction of antioxidant enzyme expression. One-month treatment of Pb-PRL mice with anethole trithione (ATT), a specific inhibitor of mitochondrial ROS production, reduced prostate weight and voiding frequency. In human BPH-1 epithelial cells, ATT decreased mitochondrial metabolism, cell proliferation, and stemness features. ATT prevented the growth of organoids generated by sorted Pb-PRL basal and LSCmed cells, the two major BPH-associated, androgen-independent epithelial cell compartments. Taken together, these results support cell plasticity as a driver of BPH progression and therapeutic resistance to androgen signaling inhibition, and identify antioxidant therapy as a promising treatment of BPH.
Collapse
Affiliation(s)
- Leïla Dos Santos
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France
| | - Francesco Carbone
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Imagine Institute, Université Paris Cité, Atip-Avenir Team, INSERM UMR 1163, Paris, France; Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Emeline Pacreau
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France
| | - Sekou Diarra
- Humana Biosciences SAS, Prologue Biotech, Labège, France
| | - Marine Luka
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France; Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Imagine Institute, Université Paris Cité, Atip-Avenir Team, INSERM UMR 1163, Paris, France
| | - Natascha Pigat
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France
| | - Manon Baures
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France
| | - Emilie Navarro
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France
| | - Julien Anract
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France; Urology Department, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Nicolas Barry Delongchamps
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France; Urology Department, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Nicolas Cagnard
- Bioinformatics Core Platform, Université Paris Cité, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, Paris, France
| | - Frédéric Bost
- C3M, INSERM U1065, Université Côte d'Azur, Equipe Labélisée Ligue Nationale contre le Cancer, Nice, France
| | - Ivan Nemazanyy
- Metabolomics Core Facility, Université de Paris-Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, Paris, France
| | | | - Ahmed Hamaï
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France
| | - Mickaël Ménager
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Imagine Institute, Université Paris Cité, Atip-Avenir Team, INSERM UMR 1163, Paris, France; Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Stefano Palea
- Humana Biosciences SAS, Prologue Biotech, Labège, France
| | - Jacques-Emmanuel Guidotti
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France
| | - Vincent Goffin
- Institut Necker Enfants Malades, Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Paris, France.
| |
Collapse
|
6
|
Germanos AA, Arora S, Zheng Y, Goddard ET, Coleman IM, Ku AT, Wilkinson S, Song H, Brady NJ, Amezquita RA, Zager M, Long A, Yang YC, Bielas JH, Gottardo R, Rickman DS, Huang FW, Ghajar CM, Nelson PS, Sowalsky AG, Setty M, Hsieh AC. Defining cellular population dynamics at single-cell resolution during prostate cancer progression. eLife 2022; 11:e79076. [PMID: 36511483 PMCID: PMC9747158 DOI: 10.7554/elife.79076] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Advanced prostate malignancies are a leading cause of cancer-related deaths in men, in large part due to our incomplete understanding of cellular drivers of disease progression. We investigate prostate cancer cell dynamics at single-cell resolution from disease onset to the development of androgen independence in an in vivo murine model. We observe an expansion of a castration-resistant intermediate luminal cell type that correlates with treatment resistance and poor prognosis in human patients. Moreover, transformed epithelial cells and associated fibroblasts create a microenvironment conducive to pro-tumorigenic immune infiltration, which is partially androgen responsive. Androgen-independent prostate cancer leads to significant diversification of intermediate luminal cell populations characterized by a range of androgen signaling activity, which is inversely correlated with proliferation and mRNA translation. Accordingly, distinct epithelial populations are exquisitely sensitive to translation inhibition, which leads to epithelial cell death, loss of pro-tumorigenic signaling, and decreased tumor heterogeneity. Our findings reveal a complex tumor environment largely dominated by castration-resistant luminal cells and immunosuppressive infiltrates.
Collapse
Affiliation(s)
- Alexandre A Germanos
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
- University of Washington Molecular and Cellular Biology ProgramSeattleUnited States
| | - Sonali Arora
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Ye Zheng
- Division of Vaccine and infectious Diseases, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Erica T Goddard
- Division of Public Health Sciences, Translational Research Program, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Anson T Ku
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Hanbing Song
- Division of Hematology/Oncology, Department of Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Nicholas J Brady
- Department of Pathology and Laboratory Medicine, Weill Cornell MedicineNew YorkUnited States
| | - Robert A Amezquita
- Division of Vaccine and infectious Diseases, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Michael Zager
- Center for Data Visualization, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Annalysa Long
- Division of Public Health Sciences, Translational Research Program, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Yu Chi Yang
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Jason H Bielas
- Division of Public Health Sciences, Translational Research Program, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Raphael Gottardo
- Division of Vaccine and infectious Diseases, Fred Hutchinson Cancer CenterSeattleUnited States
- Division of Public Health Sciences, Translational Research Program, Fred Hutchinson Cancer CenterSeattleUnited States
| | - David S Rickman
- Department of Pathology and Laboratory Medicine, Weill Cornell MedicineNew YorkUnited States
| | - Franklin W Huang
- Division of Hematology/Oncology, Department of Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Cyrus M Ghajar
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
- Division of Public Health Sciences, Translational Research Program, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
- University of Washington Departments of Medicine and Genome SciencesSeattleUnited States
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIHBethesdaUnited States
| | - Manu Setty
- Translational Data Science Integrated Research Center, Fred Hutchinson Cancer CenterSeattleUnited States
- Division of Basic Sciences, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Andrew C Hsieh
- Division of Human Biology, Fred Hutchinson Cancer CenterSeattleUnited States
- University of Washington Departments of Medicine and Genome SciencesSeattleUnited States
| |
Collapse
|
7
|
Yan Q, Wang M, Xia H, Dai C, Diao T, Wang Y, Hou H, Zhang H, Liu M, Long X. Single-cell RNA-sequencing technology demonstrates the heterogeneity between aged prostate peripheral and transitional zone. Clin Transl Med 2022; 12:e1084. [PMID: 36245324 PMCID: PMC9574492 DOI: 10.1002/ctm2.1084] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/29/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Identifying cellular and functional heterogeneity within aged prostate is critical for understanding the spatial distribution of prostate diseases. METHODS Aged human prostate peripheral zone (PZ) and transitional zone (TZ) tissues were used for single-cell RNA-sequencing. Results were validated by immunofluorescence staining. RESULTS We found that club/hillock epithelial cells, compared with other epithelial cells, had significantly higher NOTCH signaling activity and expressed higher levels of neuro-stems but lower androgen-related genes. These cells were primarily found in the TZ and provided a stem-like niche around the proximal prostate ducts. Significant heterogeneity was observed in the aged luminal population. A novel TFF3+ luminal subtype with elevated MYC and E2F pathway activities was observed, primarily in the PZ. Further analysis revealed that epithelial cells in the TZ had higher levels of stem- and inflammation-related pathway activities but lower androgen/lineage-related pathway activities than those in the PZ. Notably, the activation of MYC, E2F and DNA repair pathways significantly increased in PZ luminal cells. In the immune landscape, we found that the immune microenvironment in the TZ is more complex and disordered with more infiltration of NK and Treg cells. CD8 T cell and macrophage in the TZ exhibit both inflammation activation and suppression phenotypes. In the stroma, the TZ had a higher fibroblast density, and fibroblasts in the TZ exhibited stronger transcriptome activity in immunity and proliferation. Ligand-receptor interaction analysis revealed that fibroblasts could contribute to a NOTCH signaling niche for club/hillock cells in the TZ and balance the prostate immune microenvironment. The activation of stem properties, inflammatory infiltration and loss of androgen/lineage activity are prominent features distinguishing the TZ from PZ. CONCLUSIONS Our study explains the heterogeneity between the TZ and PZ of aged prostate, which may help understand the spatial distribution of prostate diseases and establish a foundation for novel target discovery.
Collapse
Affiliation(s)
- Qiuxia Yan
- Peking University Fifth School of Clinical MedicineBeijingChina,Department of UrologyBeijing HospitalNational Center of GerontologyBeijingChina
| | - Miao Wang
- Department of UrologyBeijing HospitalNational Center of GerontologyBeijingChina
| | - Haoran Xia
- Department of UrologyBeijing HospitalNational Center of GerontologyBeijingChina
| | - Cao Dai
- Department of General SurgeryThe Third Affiliated Hospital Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Tongxiang Diao
- Department of UrologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | | | - Huimin Hou
- Department of UrologyBeijing HospitalNational Center of GerontologyBeijingChina
| | - Hong Zhang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular SciencesPeking University Health Science CenterBeijingChina
| | - Ming Liu
- Department of UrologyBeijing HospitalNational Center of GerontologyBeijingChina
| | - Xingbo Long
- Department of UrologySun Yat‐sen University Cancer CenterGuangzhouGuangdongChina,State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| |
Collapse
|
8
|
Transcriptomic Signature and Growth Factor Regulation of Castration-Tolerant Prostate Luminal Progenitor Cells. Cancers (Basel) 2022; 14:cancers14153775. [PMID: 35954439 PMCID: PMC9367377 DOI: 10.3390/cancers14153775] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/28/2022] [Indexed: 02/05/2023] Open
Abstract
Background: The molecular and cellular mechanisms that drive castration-resistant prostate cancer (CRPC) remain poorly understood. LSCmed cells defines an FACS-enriched population of castration-tolerant luminal progenitor cells that has been proposed to promote tumorigenesis and CRPC in Pten-deficient mice. The goals of this study were to assess the relevance of LSCmed cells through the analysis of their molecular proximity with luminal progenitor-like cell clusters identified by single-cell (sc)RNA-seq analyses of mouse and human prostates, and to investigate their regulation by in silico-predicted growth factors present in the prostatic microenvironment. Methods: Several bioinformatic pipelines were used for pan-transcriptomic analyses. LSCmed cells isolated by cell sorting from healthy and malignant mouse prostates were characterized using RT-qPCR, immunofluorescence and organoid assays. Results: LSCmed cells match (i) mouse luminal progenitor cell clusters identified in scRNA-seq analyses for which we provide a common 15-gene signature including the previously identified LSCmed marker Krt4, and (ii) Club/Hillock cells of the human prostate. This transcriptional overlap was maintained in cancer contexts. EGFR/ERBB4, IGF-1R and MET pathways were identified as autocrine/paracrine regulators of progenitor, proliferation and differentiation properties of LSCmed cells. The functional redundancy of these signaling pathways allows them to bypass the effect of receptor-targeted pharmacological inhibitors. Conclusions: Based on transcriptomic profile and pharmacological resistance to monotherapies that failed in CRPC patients, this study supports LSCmed cells as a relevant model to investigate the role of castration-tolerant progenitor cells in human prostate cancer progression.
Collapse
|
9
|
Abu el Maaty MA, Terzic J, Keime C, Rovito D, Lutzing R, Yanushko D, Parisotto M, Grelet E, Namer IJ, Lindner V, Laverny G, Metzger D. Hypoxia-mediated stabilization of HIF1A in prostatic intraepithelial neoplasia promotes cell plasticity and malignant progression. SCIENCE ADVANCES 2022; 8:eabo2295. [PMID: 35867798 PMCID: PMC9307253 DOI: 10.1126/sciadv.abo2295] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Prostate cancer (PCa) is a leading cause of cancer-related deaths. The slow evolution of precancerous lesions to malignant tumors provides a broad time frame for preventing PCa. To characterize prostatic intraepithelial neoplasia (PIN) progression, we conducted longitudinal studies on Pten(i)pe-/- mice that recapitulate prostate carcinogenesis in humans. We found that early PINs are hypoxic and that hypoxia-inducible factor 1 alpha (HIF1A) signaling is activated in luminal cells, thus enhancing malignant progression. Luminal HIF1A dampens immune surveillance and drives luminal plasticity, leading to the emergence of cells that overexpress Transglutaminase 2 (TGM2) and have impaired androgen signaling. Elevated TGM2 levels in patients with PCa are associated with shortened progression-free survival after prostatectomy. Last, we show that pharmacologically inhibiting HIF1A impairs cell proliferation and induces apoptosis in PINs. Therefore, our study demonstrates that HIF1A is a target for PCa prevention and that TGM2 is a promising prognostic biomarker of early relapse after prostatectomy.
Collapse
Affiliation(s)
- Mohamed A. Abu el Maaty
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Julie Terzic
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Daniela Rovito
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Régis Lutzing
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Darya Yanushko
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Maxime Parisotto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Elise Grelet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Izzie Jacques Namer
- Université de Strasbourg, Strasbourg, France
- ICube, CNRS, UMR 7357, Strasbourg, France
| | - Véronique Lindner
- Département de Pathologie, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Gilles Laverny
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
- Corresponding author. (D.M.); (G.L.)
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
- Corresponding author. (D.M.); (G.L.)
| |
Collapse
|
10
|
Tang DG. Understanding and targeting prostate cancer cell heterogeneity and plasticity. Semin Cancer Biol 2022; 82:68-93. [PMID: 34844845 PMCID: PMC9106849 DOI: 10.1016/j.semcancer.2021.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) is a prevalent malignancy that occurs primarily in old males. Prostate tumors in different patients manifest significant inter-patient heterogeneity with respect to histo-morphological presentations and molecular architecture. An individual patient tumor also harbors genetically distinct clones in which PCa cells display intra-tumor heterogeneity in molecular features and phenotypic marker expression. This inherent PCa cell heterogeneity, e.g., in the expression of androgen receptor (AR), constitutes a barrier to the long-term therapeutic efficacy of AR-targeting therapies. Furthermore, tumor progression as well as therapeutic treatments induce PCa cell plasticity such that AR-positive PCa cells may turn into AR-negative cells and prostate tumors may switch lineage identity from adenocarcinomas to neuroendocrine-like tumors. This induced PCa cell plasticity similarly confers resistance to AR-targeting and other therapies. In this review, I first discuss PCa from the perspective of an abnormal organ development and deregulated cellular differentiation, and discuss the luminal progenitor cells as the likely cells of origin for PCa. I then focus on intrinsic PCa cell heterogeneity in treatment-naïve tumors with the presence of prostate cancer stem cells (PCSCs). I further elaborate on PCa cell plasticity induced by genetic alterations and therapeutic interventions, and present potential strategies to therapeutically tackle PCa cell heterogeneity and plasticity. My discussions will make it clear that, to achieve enduring clinical efficacy, both intrinsic PCa cell heterogeneity and induced PCa cell plasticity need to be targeted with novel combinatorial approaches.
Collapse
Affiliation(s)
- Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Experimental Therapeutics (ET) Graduate Program, The University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
11
|
Han H, Wang Y, Curto J, Gurrapu S, Laudato S, Rumandla A, Chakraborty G, Wang X, Chen H, Jiang Y, Kumar D, Caggiano EG, Capogiri M, Zhang B, Ji Y, Maity SN, Hu M, Bai S, Aparicio AM, Efstathiou E, Logothetis CJ, Navin N, Navone NM, Chen Y, Giancotti FG. Mesenchymal and stem-like prostate cancer linked to therapy-induced lineage plasticity and metastasis. Cell Rep 2022; 39:110595. [PMID: 35385726 PMCID: PMC9414743 DOI: 10.1016/j.celrep.2022.110595] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 09/18/2021] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Bioinformatic analysis of 94 patient-derived xenografts (PDXs), cell lines, and organoids (PCOs) identifies three intrinsic transcriptional subtypes of metastatic castration-resistant prostate cancer: androgen receptor (AR) pathway + prostate cancer (PC) (ARPC), mesenchymal and stem-like PC (MSPC), and neuroendocrine PC (NEPC). A sizable proportion of castration-resistant and metastatic stage PC (M-CRPC) cases are admixtures of ARPC and MSPC. Analysis of clinical datasets and mechanistic studies indicates that MSPC arises from ARPC as a consequence of therapy-induced lineage plasticity. AR blockade with enzalutamide induces (1) transcriptional silencing of TP53 and hence dedifferentiation to a hybrid epithelial and mesenchymal and stem-like state and (2) inhibition of BMP signaling, which promotes resistance to AR inhibition. Enzalutamide-tolerant LNCaP cells re-enter the cell cycle in response to neuregulin and generate metastasis in mice. Combined inhibition of HER2/3 and AR or mTORC1 exhibits efficacy in models of ARPC and MSPC or MSPC, respectively. These results define MSPC, trace its origin to therapy-induced lineage plasticity, and reveal its sensitivity to HER2/3 inhibition.
Collapse
Affiliation(s)
- Hyunho Han
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yan Wang
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; Herbert Irving Comprehensive Cancer Center and Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Josue Curto
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; Herbert Irving Comprehensive Cancer Center and Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Sreeharsha Gurrapu
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; Herbert Irving Comprehensive Cancer Center and Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Sara Laudato
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA
| | - Alekya Rumandla
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; UT MDACC UT Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | | | - Xiaobo Wang
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; Herbert Irving Comprehensive Cancer Center and Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; UT MDACC UT Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Hong Chen
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA
| | - Yan Jiang
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA
| | - Dhiraj Kumar
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; Herbert Irving Comprehensive Cancer Center and Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Emily G Caggiano
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; UT MDACC UT Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Monica Capogiri
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA
| | - Boyu Zhang
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA
| | - Yan Ji
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA
| | - Sankar N Maity
- Department of GU Oncology, UT MDACC, Houston, TX 77054, USA
| | - Min Hu
- Department of Genetics, UT MDACC, Houston, TX 77054, USA
| | - Shanshan Bai
- Department of Genetics, UT MDACC, Houston, TX 77054, USA
| | - Ana M Aparicio
- Department of GU Oncology, UT MDACC, Houston, TX 77054, USA
| | | | | | - Nicholas Navin
- Department of Genetics, UT MDACC, Houston, TX 77054, USA
| | - Nora M Navone
- Department of GU Oncology, UT MDACC, Houston, TX 77054, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program and Department of Medicine, MSKCC, New York, NY 10065, USA
| | - Filippo G Giancotti
- Department of Cancer Biology, UT MDACC, Houston, TX 77054, USA; Herbert Irving Comprehensive Cancer Center and Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
12
|
High Keratin-7 Expression in Benign Peri-Tumoral Prostatic Glands Is Predictive of Bone Metastasis Onset and Prostate Cancer-Specific Mortality. Cancers (Basel) 2022; 14:cancers14071623. [PMID: 35406395 PMCID: PMC8997075 DOI: 10.3390/cancers14071623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 12/10/2022] Open
Abstract
BACKGROUND New predictive biomarkers are needed to accurately predict metastasis-free survival (MFS) and cancer-specific survival (CSS) in localized prostate cancer (PC). Keratin-7 (KRT7) overexpression has been associated with poor prognosis in several cancers and is described as a novel prostate progenitor marker in the mouse prostate. METHODS KRT7 expression was evaluated in prostatic cell lines and in human tissue by immunohistochemistry (IHC, on advanced PC, n = 91) and immunofluorescence (IF, on localized PC, n = 285). The KRT7 mean fluorescence intensity (MFI) was quantified in different compartments by digital analysis and correlated to clinical endpoints in the localized PC cohort. RESULTS KRT7 is expressed in prostatic cell lines and found in the basal and supra-basal compartment from healthy prostatic glands and benign peri-tumoral glands from localized PC. The KRT7 staining is lost in luminal cells from localized tumors and found as an aberrant sporadic staining (2.2%) in advanced PC. In the localized PC cohort, high KRT7 MFI above the 80th percentile in the basal compartment was significantly and independently correlated with MFS and CSS, and with hypertrophic basal cell phenotype. CONCLUSION High KRT7 expression in benign glands is an independent biomarker of MFS and CSS, and its expression is lost in tumoral cells. These results require further validation on larger cohorts.
Collapse
|
13
|
Prostate luminal progenitor cells: from mouse to human, from health to disease. Nat Rev Urol 2022; 19:201-218. [DOI: 10.1038/s41585-021-00561-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/11/2022]
|
14
|
Abu El Maaty MA, Grelet E, Keime C, Rerra AI, Gantzer J, Emprou C, Terzic J, Lutzing R, Bornert JM, Laverny G, Metzger D. Single-cell analyses unravel cell type-specific responses to a vitamin D analog in prostatic precancerous lesions. SCIENCE ADVANCES 2021; 7:7/31/eabg5982. [PMID: 34330705 PMCID: PMC8324049 DOI: 10.1126/sciadv.abg5982] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/15/2021] [Indexed: 05/04/2023]
Abstract
Epidemiological data have linked vitamin D deficiency to the onset and severity of various cancers, including prostate cancer, and although in vitro studies have demonstrated anticancer activities for vitamin D, clinical trials provided conflicting results. To determine the impact of vitamin D signaling on prostatic precancerous lesions, we treated genetically engineered Pten(i)pe-/- mice harboring prostatic intraepithelial neoplasia (PIN) with Gemini-72, a vitamin D analog with reported anticancer activities. We show that this analog induces apoptosis in senescent PINs, normalizes extracellular matrix remodeling by stromal fibroblasts, and reduces the prostatic infiltration of immunosuppressive myeloid-derived suppressor cells. Moreover, single-cell RNA-sequencing analysis demonstrates that while a subset of luminal cells expressing Krt8, Krt4, and Tacstd2 (termed luminal-C cells) is lost by such a treatment, antiapoptotic pathways are induced in persistent luminal-C cells. Therefore, our findings delineate the distinct responses of PINs and the microenvironment to Gemini-72, and shed light on mechanisms that limit treatment's efficacy.
Collapse
Affiliation(s)
- Mohamed A Abu El Maaty
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Elise Grelet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Anna-Isavella Rerra
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Justine Gantzer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Camille Emprou
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Julie Terzic
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Régis Lutzing
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Jean-Marc Bornert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Gilles Laverny
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| |
Collapse
|
15
|
Li W, Shen MM. Prostate cancer cell heterogeneity and plasticity: Insights from studies of genetically-engineered mouse models. Semin Cancer Biol 2021; 82:60-67. [PMID: 34147640 DOI: 10.1016/j.semcancer.2021.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/21/2022]
Abstract
Although prostate adenocarcinoma lacks distinguishable histopathological subtypes, prostate cancer displays significant inter- and intratumor heterogeneity at the molecular level and with respect to disease prognosis and treatment response. In principle, understanding the basis for prostate cancer heterogeneity can help distinguish aggressive from indolent disease, and help overcome castration-resistance in advanced prostate cancer. In this review, we will discuss recent advances in understanding the cell types of origin, putative cancer stem cells, and tumor plasticity in prostate cancer, focusing on insights from studies of genetically engineered mouse models (GEMMs). We will also outline future directions for investigating tumor heterogeneity using mouse models of prostate cancer.
Collapse
Affiliation(s)
- Weiping Li
- 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
|
Lee MW, Miljanic M, Triplett T, Ramirez C, Aung KL, Eckhardt SG, Capasso A. Current methods in translational cancer research. Cancer Metastasis Rev 2021; 40:7-30. [PMID: 32929562 PMCID: PMC7897192 DOI: 10.1007/s10555-020-09931-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022]
Abstract
Recent developments in pre-clinical screening tools, that more reliably predict the clinical effects and adverse events of candidate therapeutic agents, has ushered in a new era of drug development and screening. However, given the rapid pace with which these models have emerged, the individual merits of these translational research tools warrant careful evaluation in order to furnish clinical researchers with appropriate information to conduct pre-clinical screening in an accelerated and rational manner. This review assesses the predictive utility of both well-established and emerging pre-clinical methods in terms of their suitability as a screening platform for treatment response, ability to represent pharmacodynamic and pharmacokinetic drug properties, and lastly debates the translational limitations and benefits of these models. To this end, we will describe the current literature on cell culture, organoids, in vivo mouse models, and in silico computational approaches. Particular focus will be devoted to discussing gaps and unmet needs in the literature as well as current advancements and innovations achieved in the field, such as co-clinical trials and future avenues for refinement.
Collapse
Affiliation(s)
- Michael W Lee
- Department of Medical Education, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Mihailo Miljanic
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Todd Triplett
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Craig Ramirez
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Kyaw L Aung
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - S Gail Eckhardt
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Anna Capasso
- Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
- Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
17
|
Joseph DB, Turco AE, Vezina CM, Strand DW. Progenitors in prostate development and disease. Dev Biol 2021; 473:50-58. [PMID: 33529704 DOI: 10.1016/j.ydbio.2020.11.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/21/2022]
Abstract
The prostate develops by epithelial budding and branching processes that occur during fetal and postnatal stages. The adult prostate demonstrates remarkable regenerative capacity, with the ability to regrow to its original size over multiple cycles of castration and androgen administration. This capacity for controlled regeneration prompted the search for an androgen-independent epithelial progenitor in benign prostatic hyperplasia (BPH) and prostate cancer (PCa). BPH is hypothesized to be a reawakening of ductal branching, resulting in the formation of new proximal glands, all while androgen levels are decreasing in the aging male. Advanced prostate cancer can be slowed with androgen deprivation, but resistance eventually occurs, suggesting the existence of an androgen-independent progenitor. Recent studies indicate that there are multiple castration-insensitive epithelial cell types in the proximal area of the prostate, but not all act as progenitors during prostate development or regeneration. This review highlights how recent cellular and anatomical studies are changing our perspective on the identity of the prostate progenitor.
Collapse
Affiliation(s)
- Diya B Joseph
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Anne E Turco
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Chad M Vezina
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Douglas W Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
18
|
Joseph DB, Henry GH, Malewska A, Iqbal NS, Ruetten HM, Turco AE, Abler LL, Sandhu SK, Cadena MT, Malladi VS, Reese JC, Mauck RJ, Gahan JC, Hutchinson RC, Roehrborn CG, Baker LA, Vezina CM, Strand DW. Urethral luminal epithelia are castration-insensitive cells of the proximal prostate. Prostate 2020; 80:872-884. [PMID: 32497356 PMCID: PMC7339731 DOI: 10.1002/pros.24020] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Castration-insensitive epithelial progenitors capable of regenerating the prostate have been proposed to be concentrated in the proximal region based on facultative assays. Functional characterization of prostate epithelial populations isolated with individual cell surface markers has failed to provide a consensus on the anatomical and transcriptional identity of proximal prostate progenitors. METHODS Here, we use single-cell RNA sequencing to obtain a complete transcriptomic profile of all epithelial cells in the mouse prostate and urethra to objectively identify cellular subtypes. Pan-transcriptomic comparison to human prostate cell types identified a mouse equivalent of human urethral luminal cells, which highly expressed putative prostate progenitor markers. Validation of the urethral luminal cell cluster was performed using immunostaining and flow cytometry. RESULTS Our data reveal that previously identified facultative progenitors marked by Trop2, Sca-1, KRT4, and PSCA are actually luminal epithelial cells of the urethra that extend into the proximal region of the prostate, and are resistant to castration-induced androgen deprivation. Mouse urethral luminal cells were identified to be the equivalent of previously identified human club and hillock cells that similarly extend into proximal prostate ducts. Benign prostatic hyperplasia (BPH) has long been considered an "embryonic reawakening," but the cellular origin of the hyperplastic growth concentrated in the periurethral region is unclear. We demonstrate an increase in urethral luminal cells within glandular nodules from BPH patients. Urethral luminal cells are further increased in patients treated with a 5-α reductase inhibitor. CONCLUSIONS Our data demonstrate that cells of the proximal prostate that express putative progenitor markers, and are enriched by castration in the proximal prostate, are urethral luminal cells and that these cells may play an important role in the etiology of human BPH.
Collapse
Affiliation(s)
- Diya B. Joseph
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Gervaise H. Henry
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, Texas
| | - Alicia Malewska
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Nida S. Iqbal
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Hannah M. Ruetten
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Anne E. Turco
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lisa L. Abler
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Simran K. Sandhu
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Mark T. Cadena
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Venkat S. Malladi
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, Texas
| | | | - Ryan J. Mauck
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Jeffrey C. Gahan
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | | | | | - Linda A. Baker
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Chad M. Vezina
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Douglas W. Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
19
|
Testa U, Castelli G, Pelosi E. Cellular and Molecular Mechanisms Underlying Prostate Cancer Development: Therapeutic Implications. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E82. [PMID: 31366128 PMCID: PMC6789661 DOI: 10.3390/medicines6030082] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most frequent nonskin cancer and second most common cause of cancer-related deaths in man. Prostate cancer is a clinically heterogeneous disease with many patients exhibiting an aggressive disease with progression, metastasis, and other patients showing an indolent disease with low tendency to progression. Three stages of development of human prostate tumors have been identified: intraepithelial neoplasia, adenocarcinoma androgen-dependent, and adenocarcinoma androgen-independent or castration-resistant. Advances in molecular technologies have provided a very rapid progress in our understanding of the genomic events responsible for the initial development and progression of prostate cancer. These studies have shown that prostate cancer genome displays a relatively low mutation rate compared with other cancers and few chromosomal loss or gains. The ensemble of these molecular studies has led to suggest the existence of two main molecular groups of prostate cancers: one characterized by the presence of ERG rearrangements (~50% of prostate cancers harbor recurrent gene fusions involving ETS transcription factors, fusing the 5' untranslated region of the androgen-regulated gene TMPRSS2 to nearly the coding sequence of the ETS family transcription factor ERG) and features of chemoplexy (complex gene rearrangements developing from a coordinated and simultaneous molecular event), and a second one characterized by the absence of ERG rearrangements and by the frequent mutations in the E3 ubiquitin ligase adapter SPOP and/or deletion of CDH1, a chromatin remodeling factor, and interchromosomal rearrangements and SPOP mutations are early events during prostate cancer development. During disease progression, genomic and epigenomic abnormalities accrued and converged on prostate cancer pathways, leading to a highly heterogeneous transcriptomic landscape, characterized by a hyperactive androgen receptor signaling axis.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
20
|
Mao N, Gao D, Hu W, Hieronymus H, Wang S, Lee YS, Lee C, Choi D, Gopalan A, Chen Y, Carver BS. Aberrant Expression of ERG Promotes Resistance to Combined PI3K and AR Pathway Inhibition through Maintenance of AR Target Genes. Mol Cancer Ther 2019; 18:1577-1586. [PMID: 31296553 DOI: 10.1158/1535-7163.mct-18-1386] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/02/2019] [Accepted: 07/01/2019] [Indexed: 11/16/2022]
Abstract
On the basis of our previous work defining the molecular rationale for combined targeting of the PI3K and AR pathways in PTEN loss prostate cancer, the first clinical trial was recently reported demonstrating a significant benefit for combination therapy in patients with metastatic prostate cancer. In this phase II trial, loss of PTEN was a biomarker predictive of response to combined AKT and AR inhibition. Given that PTEN loss prostate cancers are significantly enriched for ERG genomic rearrangements, we evaluated how the aberrant expression of ERG may impact response to PI3K/AR-targeted therapy. Here, we show that overexpression of ERG in the setting of Pten loss promotes resistance to combined PI3K and AR pathway inhibition with associated maintenance of AR target gene expression. Importantly, following AR knockout in the setting of ERG overexpression, there is maintenance of a subset of AR lineage-specific target genes, making AR dispensable in this context. This has important clinical implications as even in the setting of the androgen-regulated TMPRSS2:ERG genomic rearrangement, ERG expression is never abolished following AR inhibition and may allow for cell survival following AR (lineage)-targeted therapies.
Collapse
Affiliation(s)
- Ninghui Mao
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dong Gao
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wenhuo Hu
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Haley Hieronymus
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shangqian Wang
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Young Sun Lee
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cindy Lee
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Danielle Choi
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anuradha Gopalan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yu Chen
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brett S Carver
- Human Oncogenesis and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Division of Urology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
21
|
STAT5a/b Deficiency Delays, but does not Prevent, Prolactin-Driven Prostate Tumorigenesis in Mice. Cancers (Basel) 2019; 11:cancers11070929. [PMID: 31269779 PMCID: PMC6678910 DOI: 10.3390/cancers11070929] [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: 06/11/2019] [Revised: 06/28/2019] [Accepted: 06/28/2019] [Indexed: 11/28/2022] Open
Abstract
The canonical prolactin (PRL) Signal Transducer and Activator of Transcription (STAT) 5 pathway has been suggested to contribute to human prostate tumorigenesis via an autocrine/paracrine mechanism. The probasin (Pb)-PRL transgenic mouse models this mechanism by overexpressing PRL specifically in the prostate epithelium leading to strong STAT5 activation in luminal cells. These mice exhibit hypertrophic prostates harboring various pre-neoplastic lesions that aggravate with age and accumulation of castration-resistant stem/progenitor cells. As STAT5 signaling is largely predominant over other classical PRL-triggered pathways in Pb-PRL prostates, we reasoned that Pb-Cre recombinase-driven genetic deletion of a floxed Stat5a/b locus should prevent prostate tumorigenesis in so-called Pb-PRLΔSTAT5 mice. Anterior and dorsal prostate lobes displayed the highest Stat5a/b deletion efficiency with no overt compensatory activation of other PRLR signaling cascade at 6 months of age; hence the development of tumor hallmarks was markedly reduced. Stat5a/b deletion also reversed the accumulation of stem/progenitor cells, indicating that STAT5 signaling regulates prostate epithelial cell hierarchy. Interestingly, ERK1/2 and AKT, but not STAT3 and androgen signaling, emerged as escape mechanisms leading to delayed tumor development in aged Pb-PRLΔSTAT5 mice. Unexpectedly, we found that Pb-PRL prostates spontaneously exhibited age-dependent decline of STAT5 signaling, also to the benefit of AKT and ERK1/2 signaling. As a consequence, both Pb-PRL and Pb-PRLΔSTAT5 mice ultimately displayed similar pathological prostate phenotypes at 18 months of age. This preclinical study provides insight on STAT5-dependent mechanisms of PRL-induced prostate tumorigenesis and alternative pathways bypassing STAT5 signaling down-regulation upon prostate neoplasia progression.
Collapse
|
22
|
Abstract
Stem/progenitor cells play central roles in processes of organogenesis and tissue maintenance, whereas cancer stem cells (CSCs) are thought to drive tumor malignancy. Here, we review recent progress in the identification and analysis of normal prostate stem/progenitor cells as well as putative CSCs in both genetically engineered mouse models as well as in human tissue. We also discuss studies that have investigated the cell type of origin for prostate cancer. In addition, we provide a critical assessment of methodologies used in stem cell analyses and outline directions for future research.
Collapse
Affiliation(s)
- Jia J Li
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Department Genetics and Development, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Department of Urology, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Department of Systems Biology, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Michael M Shen
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Department Genetics and Development, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Department of Urology, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Department of Systems Biology, Columbia University College of Physicians and Surgeons, New York, New York 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York, New York 10032
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Goffin V. Prolactin receptor targeting in breast and prostate cancers: New insights into an old challenge. Pharmacol Ther 2017; 179:111-126. [DOI: 10.1016/j.pharmthera.2017.05.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|