1
|
Noorani R, Botting-Provost S, Barsoum GK, Laurie C, El-Zein M, Franco EL. Comprehensive appraisal of the association between sexually transmitted infections and prostate cancer: A scoping review of empirical studies, reviews, and meta-analyses. Cancer Epidemiol 2025; 96:102781. [PMID: 40031094 DOI: 10.1016/j.canep.2025.102781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/09/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
We performed a scoping review on the association of sexually transmitted infections (STIs) with prostate cancer and identified knowledge gaps. Searching four databases (Medline, Embase, Scopus, and Cochrane) identified 286 eligible records. Most empirical studies (n = 191) were cross-sectional (n = 66) and case-control (n = 52). The most studied STIs were human papillomavirus (HPV) (n = 82), human immunodeficiency virus (HIV) (n = 52), and herpes simplex virus (HSV) (n = 30). We included 68 narrative reviews, 10 systematic reviews, and 17 meta-analyses. Most effect estimates (odds ratios, hazard ratios, risk ratios and standardised incidence ratios) did not support an association between STIs and prostate cancer: 373 and 218 of 591 effect estimates were above and below the null, respectively, except for HIV where 74 of 108 estimates were below the null. Knowledge gaps included case-control studies, insights into HIV-related mechanisms for a lower risk for prostate cancer, studies on Mycoplasma and Ureaplasma, studies adjusting for co-infection with other STIs, and studies assessing whether STIs predispose men to a more aggressive form of prostate cancer. A key research priority identified is the need for more evidence on the biological mechanisms driving infection-mediated prostate carcinogenesis.
Collapse
Affiliation(s)
- Rodrigo Noorani
- Division of Cancer Epidemiology, McGill University, Montréal, QC, Canada.
| | | | - George Kas Barsoum
- Division of Cancer Epidemiology, McGill University, Montréal, QC, Canada
| | - Cassandra Laurie
- Division of Cancer Epidemiology, McGill University, Montréal, QC, Canada
| | - Mariam El-Zein
- Division of Cancer Epidemiology, McGill University, Montréal, QC, Canada
| | - Eduardo L Franco
- Division of Cancer Epidemiology, McGill University, Montréal, QC, Canada
| |
Collapse
|
2
|
Hu J, Liu F, Zhang J, Yin L, Cao W, Xu W, Chang Y, Wang Y, Wang J, Hou Y, Liu L, Chen S, Zhu G, Jiang J, Wang Z, Wei GH, He HH, Gu D, Chen K, Ren S. Spatially resolved transcriptomic analysis of the adult human prostate. Nat Genet 2025; 57:922-933. [PMID: 40169792 DOI: 10.1038/s41588-025-02139-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/21/2025] [Indexed: 04/03/2025]
Abstract
The prostate is an organ characterized by significant spatial heterogeneity. To better understand its intricate structure and cellular composition, we constructed a comprehensive single-cell atlas of the adult human prostate. Our high-resolution mapping effort identified 253,381 single cells and 34,876 nuclei sampled from 11 patients who underwent radical resection of bladder cancer, which were categorized into 126 unique subpopulations. This work revealed various new cell types in the human prostate and their specific spatial localization. Notably, we discovered four distinct acini, two of which were tightly associated with E-twenty-six transcription factor family (ETS)-fusion-negative prostate cancer. Through the integration of spatial, single-cell and bulk-seq analyses, we propose that two specific luminal cell types could serve as the common origins of prostate cancer. Additionally, our findings suggest that zone-specific fibroblasts may contribute to the observed heterogeneity among luminal cells. This atlas will serve as a valuable reference for studying prostate biology and diseases such as prostate cancer.
Collapse
Affiliation(s)
- Junyi Hu
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Liu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jing Zhang
- Department of Pathology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lei Yin
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wanli Cao
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Weidong Xu
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yifan Chang
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Ye Wang
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jian Wang
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yaxin Hou
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lilong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sujun Chen
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Guanghui Zhu
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Junhui Jiang
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo, China
| | - Zixian Wang
- MOE Key Laboratory of Metabolism and Molecular Medicine and Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Gong-Hong Wei
- MOE Key Laboratory of Metabolism and Molecular Medicine and Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer Center, Shanghai Medical College of Fudan University, Shanghai, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Housheng Hansen He
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Di Gu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shancheng Ren
- Department of Urology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
3
|
Sugiki S, Horie T, Kunii K, Sakamoto T, Nakamura Y, Chikazawa I, Morita N, Ishigaki Y, Miyazawa K. Integrated Bioinformatic Analyses Reveal Thioredoxin as a Putative Marker of Cancer Stem Cells and Prognosis in Prostate Cancer. Cancer Inform 2025; 24:11769351251319872. [PMID: 40008390 PMCID: PMC11851766 DOI: 10.1177/11769351251319872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Objectives Prostate cancer stem cells (CSCs) play an important role in cancer cell survival, proliferation, metastasis, and recurrence; thus, removing CSCs is important for complete cancer removal. However, the mechanisms underlying CSC functions remain largely unknown, making it difficult to develop new anticancer drugs targeting CSCs. Herein, we aimed to identify novel factors that regulate stemness and predict prognosis. Methods We reanalyzed 2 single-cell RNA sequencing data of prostate cancer (PCa) tissues using Seurat. We used gene set enrichment analysis (GSEA) to estimate CSCs and identified common upregulated genes in CSCs between these datasets. To investigate whether its expression levels change over CSC differentiation, we performed a trajectory analysis using monocle 3. In addition, GSEA helped us understand how the identified genes regulate stemness. Finally, to assess their clinical significance, we used the Cancer Genome Atlas database to evaluate their impact on prognosis. Results The expression of thioredoxin (TXN), a redox enzyme, was approximately 1.2 times higher in prostate CSCs than in PCa cells (P < 1 × 10-10), and TXN expression decreased over CSC differentiation. In addition, GSEA suggested that intracellular signaling pathways, including MYC, may be involved in stemness regulation by TXN. Furthermore, TXN expression correlated with poor prognosis (P < .05) in PCa patients with high stemness. Conclusions Despite the limited sample size in our study and the need for further in vitro and in vivo experiments to demonstrate whether TXN functionally regulates prostate CSCs, our findings suggest that TXN may serve as a novel therapeutic target against CSCs. Moreover, TXN expression in CSCs could be a useful marker for predicting the prognosis of PCa patients.
Collapse
Affiliation(s)
- Shigeru Sugiki
- Department of Urology, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Tetsuhiro Horie
- Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, Japan
- Department of Pharmacy, Kanazawa Medical University Hospital, Kahoku, Ishikawa, Japan
| | - Kenshiro Kunii
- Department of Urology, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Takuya Sakamoto
- Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, Japan
- Department of Pharmacy, Kanazawa Medical University Hospital, Kahoku, Ishikawa, Japan
| | - Yuka Nakamura
- Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Ippei Chikazawa
- Department of Urology, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Nobuyo Morita
- Department of Urology, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Katsuhito Miyazawa
- Department of Urology, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| |
Collapse
|
4
|
Zhu Z, Xuan W, Wang C, Li C. Long noncoding RNA mediates enzalutamide resistance and transformation in neuroendocrine prostate cancer. Front Oncol 2024; 14:1481777. [PMID: 39655078 PMCID: PMC11625809 DOI: 10.3389/fonc.2024.1481777] [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: 08/16/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Prostate cancer is a malignant tumor caused by the malignant proliferation of epithelial cells, which is highly heterogeneous and drug-resistant, and neuroendocrine prostate cancer (NEPC) is an essential cause of drug resistance in its late stage. Elucidating the evolution of NEPC and the resistance process of enzalutamide, a novel antiandrogen, will be of great help in improving the prognosis of patients. As a research hotspot in the field of molecular biology in recent years, the wide range of biological functions of long noncoding RNAs (lncRNAs) has demonstrated their position in the therapeutic process of many diseases, and a large number of studies have revealed their critical roles in tumor progression and drug resistance. Therefore, elucidating the involvement of lncRNAs in the formation of NEPCs and their interrelationship with enzalutamide resistance may provide new ideas for a deeper understanding of the development of this disease and the occurrence of enzalutamide resistance and give a new direction for reversing the therapeutic dilemma of advanced prostate cancer. This article focuses on lncRNAs that regulate enzalutamide resistance and the neuroendocrine transition of prostate cancer through epigenetic, androgen receptor (AR) signaling, and non-AR pathways that act as "molecular sponges" interacting with miRNAs. Some insights into these mechanisms are used to provide some help for subsequent research in this area.
Collapse
Affiliation(s)
- Zhe Zhu
- Department of Urology, Anhui No.2 Provincial People’s Hospital, HeFei, China
| | - Wenjing Xuan
- Department of Obstetrics, Anhui No.2 Provincial People’s Hospital, HeFei, China
| | - Chaohui Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chancan Li
- Department of Urology, Anhui No.2 Provincial People’s Hospital, HeFei, China
| |
Collapse
|
5
|
Kiviaho A, Eerola SK, Kallio HML, Andersen MK, Hoikka M, Tiihonen AM, Salonen I, Spotbeen X, Giesen A, Parker CTA, Taavitsainen S, Hantula O, Marttinen M, Hermelo I, Ismail M, Midtbust E, Wess M, Devlies W, Sharma A, Krossa S, Häkkinen T, Afyounian E, Vandereyken K, Kint S, Kesseli J, Tolonen T, Tammela TLJ, Viset T, Størkersen Ø, Giskeødegård GF, Rye MB, Murtola T, Erickson A, Latonen L, Bova GS, Mills IG, Joniau S, Swinnen JV, Voet T, Mirtti T, Attard G, Claessens F, Visakorpi T, Rautajoki KJ, Tessem MB, Urbanucci A, Nykter M. Single cell and spatial transcriptomics highlight the interaction of club-like cells with immunosuppressive myeloid cells in prostate cancer. Nat Commun 2024; 15:9949. [PMID: 39550375 PMCID: PMC11569175 DOI: 10.1038/s41467-024-54364-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
Prostate cancer treatment resistance is a significant challenge facing the field. Genomic and transcriptomic profiling have partially elucidated the mechanisms through which cancer cells escape treatment, but their relation toward the tumor microenvironment (TME) remains elusive. Here we present a comprehensive transcriptomic landscape of the prostate TME at multiple points in the standard treatment timeline employing single-cell RNA-sequencing and spatial transcriptomics data from 120 patients. We identify club-like cells as a key epithelial cell subtype that acts as an interface between the prostate and the immune system. Tissue areas enriched with club-like cells have depleted androgen signaling and upregulated expression of luminal progenitor cell markers. Club-like cells display a senescence-associated secretory phenotype and their presence is linked to increased polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) activity. Our results indicate that club-like cells are associated with myeloid inflammation previously linked to androgen deprivation therapy resistance, providing a rationale for their therapeutic targeting.
Collapse
Affiliation(s)
- Antti Kiviaho
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Sini K Eerola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Heini M L Kallio
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Maria K Andersen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Miina Hoikka
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Aliisa M Tiihonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Iida Salonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Xander Spotbeen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Alexander Giesen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Sinja Taavitsainen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Olli Hantula
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Mikael Marttinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Ismaïl Hermelo
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | | | - Elise Midtbust
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Maximilian Wess
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Wout Devlies
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Molecular Endocrinology Laboratory, Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Abhibhav Sharma
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sebastian Krossa
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Central staff, St. Olavs Hospital HF, 7006, Trondheim, Norway
| | - Tomi Häkkinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Ebrahim Afyounian
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Katy Vandereyken
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Sam Kint
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Juha Kesseli
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Teemu Tolonen
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
- Department of Pathology, Fimlab Laboratories, Ltd, Tampere University Hospital, Tampere, Finland
| | - Teuvo L J Tammela
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Trond Viset
- Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Øystein Størkersen
- Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Guro F Giskeødegård
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Morten B Rye
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Teemu Murtola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Andrew Erickson
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- ICAN-Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - G Steven Bova
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Patrick G Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast, UK
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Thierry Voet
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Tuomas Mirtti
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- ICAN-Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Pathology, University of Helsinki & Helsinki University Hospital, Helsinki, Finland
| | - Gerhardt Attard
- University College London Cancer Institute, London, UK
- University College London Hospitals, London, UK
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Tapio Visakorpi
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
- Fimlab Laboratories, Ltd, Tampere University Hospital, Tampere, Finland
| | - Kirsi J Rautajoki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - May-Britt Tessem
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Alfonso Urbanucci
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland.
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| | - Matti Nykter
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland.
| |
Collapse
|
6
|
Mu J, Li R, Zheng Y, Lu Y, Ma L, Yin L, Zhang M, Ma W, Chang M, Liu A, Li J, Zhu H, Wang D. Human intermediate prostate cancer stem cells contribute to the initiation and development of prostate adenocarcinoma. Stem Cell Res Ther 2024; 15:296. [PMID: 39256886 PMCID: PMC11389492 DOI: 10.1186/s13287-024-03917-8] [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: 03/24/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Intermediate cells are present in the early stages of human prostate development and adenocarcinoma. While primary cells isolated from benign human prostate tissues or tumors exhibit an intermediate phenotype in vitro, they cannot form tumors in vivo unless genetically modified. It is unclear about the stem cell properties and tumorigenicity of intermediate cells. METHODS We developed a customized medium to culture primary human intermediate prostate cells, which were transplanted into male immunodeficient NCG mice to examine tumorigenicity in vivo. We treated the cells with different concentrations of dihydrotestosterone (DHT) and enzalutamide in vitro and surgically castrated the mice after cell transplantation in vivo. Immunostaining, qRT-PCR, RNA sequencing, and western blotting were performed to characterize the cells in tissues and 2D and 3D cultures. RESULTS We found intermediate cells expressing AR+PSA+CK8+CK5+ in the luminal compartment of human prostate adenocarcinoma by immunostaining. We cultured the primary intermediate cells in vitro, which expressed luminal (AR+PSA+CK8+CK18+), basal (CK5+P63+), intermediate (IVL+), and stem cell (CK4+CK13+PSCA+SOX2+) markers. These cells resisted castration in vitro by upregulating the expression of AR, PSA, and proliferation markers KI67 and PCNA. The intermediate cells had high tumorigenicity in vivo, forming tumors in immunodeficient NCG mice in a month without any genetic modification or co-transplantation with embryonic urogenital sinus mesenchyme (UGSM) cells. We named these cells human castration-resistant intermediate prostate cancer stem cells or CriPCSCs and defined the xenograft model as patient primary cell-derived xenograft (PrDX). Human CriPCSCs resisted castration in vitro and in vivo by upregulating AR expression. Furthermore, human CriPCSCs differentiated into amplifying adenocarcinoma cells of luminal phenotype in PrDX tumors in vivo, which can dedifferentiate into CriPCSCs in vitro. CONCLUSIONS Our study identified and established methods for culturing human CriPCSCs, which had high tumorigenicity in vivo without any genetic modification or UGSM co-transplantation. Human CriPCSCs differentiated into amplifying adenocarcinoma cells of luminal phenotype in the fast-growing tumors in vivo, which hold the potential to dedifferentiate into intermediate stem cells. These cells resisted castration by upregulating AR expression. The human CriPCSC and PrDX methods hold significant potential for advancing prostate cancer research and precision medicine.
Collapse
Affiliation(s)
- Jie Mu
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Ruizhi Li
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Yu Zheng
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266011, China
| | - Yi Lu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266011, China
| | - Lei Ma
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Lin Yin
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Miao Zhang
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
| | - Wenyu Ma
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
| | - Mengjia Chang
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
| | - Aihua Liu
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China.
- College of Life Sciences, Qingdao University, Qingdao, 266071, China.
| | - Jing Li
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China.
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266011, China.
| | - Dong Wang
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
7
|
Zhu T, Tong H, Du Z, Beck S, Teschendorff AE. An improved epigenetic counter to track mitotic age in normal and precancerous tissues. Nat Commun 2024; 15:4211. [PMID: 38760334 PMCID: PMC11101651 DOI: 10.1038/s41467-024-48649-8] [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: 09/24/2023] [Accepted: 05/09/2024] [Indexed: 05/19/2024] Open
Abstract
The cumulative number of stem cell divisions in a tissue, known as mitotic age, is thought to be a major determinant of cancer-risk. Somatic mutational and DNA methylation (DNAm) clocks are promising tools to molecularly track mitotic age, yet their relationship is underexplored and their potential for cancer risk prediction in normal tissues remains to be demonstrated. Here we build and validate an improved pan-tissue DNAm counter of total mitotic age called stemTOC. We demonstrate that stemTOC's mitotic age proxy increases with the tumor cell-of-origin fraction in each of 15 cancer-types, in precancerous lesions, and in normal tissues exposed to major cancer risk factors. Extensive benchmarking against 6 other mitotic counters shows that stemTOC compares favorably, specially in the preinvasive and normal-tissue contexts. By cross-correlating stemTOC to two clock-like somatic mutational signatures, we confirm the mitotic-like nature of only one of these. Our data points towards DNAm as a promising molecular substrate for detecting mitotic-age increases in normal tissues and precancerous lesions, and hence for developing cancer-risk prediction strategies.
Collapse
Affiliation(s)
- Tianyu Zhu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Huige Tong
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Zhaozhen Du
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Stephan Beck
- Medical Genomics Group, UCL Cancer Institute, University College London, 72 Huntley Street, WC1E 6BT, London, UK
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
| |
Collapse
|
8
|
Feng DC, Zhu WZ, Wang J, Li DX, Shi X, Xiong Q, You J, Han P, Qiu S, Wei Q, Yang L. The implications of single-cell RNA-seq analysis in prostate cancer: unraveling tumor heterogeneity, therapeutic implications and pathways towards personalized therapy. Mil Med Res 2024; 11:21. [PMID: 38605399 PMCID: PMC11007901 DOI: 10.1186/s40779-024-00526-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
In recent years, advancements in single-cell and spatial transcriptomics, which are highly regarded developments in the current era, particularly the emerging integration of single-cell and spatiotemporal transcriptomics, have enabled a detailed molecular comprehension of the complex regulation of cell fate. The insights obtained from these methodologies are anticipated to significantly contribute to the development of personalized medicine. Currently, single-cell technology is less frequently utilized for prostate cancer compared with other types of tumors. Starting from the perspective of RNA sequencing technology, this review outlined the significance of single-cell RNA sequencing (scRNA-seq) in prostate cancer research, encompassing preclinical medicine and clinical applications. We summarize the differences between mouse and human prostate cancer as revealed by scRNA-seq studies, as well as a combination of multi-omics methods involving scRNA-seq to highlight the key molecular targets for the diagnosis, treatment, and drug resistance characteristics of prostate cancer. These studies are expected to provide novel insights for the development of immunotherapy and other innovative treatment strategies for castration-resistant prostate cancer. Furthermore, we explore the potential clinical applications stemming from other single-cell technologies in this review, paving the way for future research in precision medicine.
Collapse
Affiliation(s)
- De-Chao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Surgery & Interventional Science, University College London, London, WC1E 6BT, UK.
| | - Wei-Zhen Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Deng-Xiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xu Shi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiao Xiong
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jia You
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shi Qiu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Feng W, Ladewig E, Salsabeel N, Zhao H, Lee YS, Gopalan A, Lange M, Luo H, Kang W, Fan N, Rosiek E, de Stanchina E, Chen Y, Carver BS, Leslie CS, Sawyers CL. ERG activates a stem-like proliferation-differentiation program in prostate epithelial cells with mixed basal-luminal identity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.15.540839. [PMID: 38585869 PMCID: PMC10996491 DOI: 10.1101/2023.05.15.540839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
To gain insight into how ERG translocations cause prostate cancer, we performed single cell transcriptional profiling of an autochthonous mouse model at an early stage of disease initiation. Despite broad expression of ERG in all prostate epithelial cells, proliferation was enriched in a small, stem-like population with mixed-luminal basal identity (called intermediate cells). Through a series of lineage tracing and primary prostate tissue transplantation experiments, we find that tumor initiating activity resides in a subpopulation of basal cells that co-express the luminal genes Tmprss2 and Nkx3.1 (called BasalLum) but not in the larger population of classical Krt8+ luminal cells. Upon ERG activation, BasalLum cells give rise to the highly proliferative intermediate state, which subsequently transitions to the larger population of Krt8+ luminal cells characteristic of ERG-positive human cancers. Furthermore, this proliferative population is characterized by an ERG-specific chromatin state enriched for NFkB, AP-1, STAT and NFAT binding, with implications for TF cooperativity. The fact that the proliferative potential of ERG is enriched in a small stem-like population implicates the chromatin context of these cells as a critical variable for unmasking its oncogenic activity.
Collapse
Affiliation(s)
- Weiran Feng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Erik Ladewig
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Nazifa Salsabeel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Huiyong Zhao
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Young Sun Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Anuradha Gopalan
- Department of Pathology, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Matthew Lange
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Hanzhi Luo
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Wenfei Kang
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Ning Fan
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Eric Rosiek
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Brett S. Carver
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
- Division of Urology, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Christina S. Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| | - Charles L. Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center; New York, NY 10065, USA
| |
Collapse
|
10
|
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
|
11
|
Chen J, Zheng Q, Hicks JL, Trabzonlu L, Ozbek B, Jones T, Vaghasia AM, Larman TC, Wang R, Markowski MC, Denmeade SR, Pienta KJ, Hruban RH, Antonarakis ES, Gupta A, Dang CV, Yegnasubramanian S, De Marzo AM. MYC-driven increases in mitochondrial DNA copy number occur early and persist throughout prostatic cancer progression. JCI Insight 2023; 8:e169868. [PMID: 37971875 PMCID: PMC10807718 DOI: 10.1172/jci.insight.169868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
Increased mitochondrial function may render some cancers vulnerable to mitochondrial inhibitors. Since mitochondrial function is regulated partly by mitochondrial DNA copy number (mtDNAcn), accurate measurements of mtDNAcn could help reveal which cancers are driven by increased mitochondrial function and may be candidates for mitochondrial inhibition. However, prior studies have employed bulk macrodissections that fail to account for cell type-specific or tumor cell heterogeneity in mtDNAcn. These studies have often produced unclear results, particularly in prostate cancer. Herein, we developed a multiplex in situ method to spatially quantify cell type-specific mtDNAcn. We show that mtDNAcn is increased in luminal cells of high-grade prostatic intraepithelial neoplasia (HGPIN), is increased in prostatic adenocarcinomas (PCa), and is further elevated in metastatic castration-resistant prostate cancer. Increased PCa mtDNAcn was validated by 2 orthogonal methods and is accompanied by increases in mtRNAs and enzymatic activity. Mechanistically, MYC inhibition in prostate cancer cells decreases mtDNA replication and expression of several mtDNA replication genes, and MYC activation in the mouse prostate leads to increased mtDNA levels in the neoplastic prostate cells. Our in situ approach also revealed elevated mtDNAcn in precancerous lesions of the pancreas and colon/rectum, demonstrating generalization across cancer types using clinical tissue samples.
Collapse
Affiliation(s)
- Jiayu Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qizhi Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jessica L. Hicks
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Levent Trabzonlu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Busra Ozbek
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tracy Jones
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Tatianna C. Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Sam R. Denmeade
- Department of Oncology and
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kenneth J. Pienta
- Department of Oncology and
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ralph H. Hruban
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Emmanuel S. Antonarakis
- Department of Oncology and
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Chi V. Dang
- Department of Oncology and
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Srinivasan Yegnasubramanian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology and
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology and
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
García-Vílchez R, Añazco-Guenkova AM, López J, Dietmann S, Tomé M, Jimeno S, Azkargorta M, Elortza F, Bárcena L, Gonzalez-Lopez M, Aransay AM, Sánchez-Martín MA, Huertas P, Durán RV, Blanco S. N7-methylguanosine methylation of tRNAs regulates survival to stress in cancer. Oncogene 2023; 42:3169-3181. [PMID: 37660182 PMCID: PMC10589097 DOI: 10.1038/s41388-023-02825-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 07/27/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2023]
Abstract
Tumour progression and therapy tolerance are highly regulated and complex processes largely dependent on the plasticity of cancer cells and their capacity to respond to stress. The higher plasticity of cancer cells highlights the need for identifying targetable molecular pathways that challenge cancer cell survival. Here, we show that N7-guanosine methylation (m7G) of tRNAs, mediated by METTL1, regulates survival to stress conditions in cancer cells. Mechanistically, we find that m7G in tRNAs protects them from stress-induced cleavage and processing into 5' tRNA fragments. Our analyses reveal that the loss of tRNA m7G methylation activates stress response pathways, sensitising cancer cells to stress. Furthermore, we find that the loss of METTL1 reduces tumour growth and increases cytotoxic stress in vivo. Our study uncovers the role of m7G methylation of tRNAs in stress responses and highlights the potential of targeting METTL1 to sensitise cancer cells to chemotherapy.
Collapse
Affiliation(s)
- Raquel García-Vílchez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007, Salamanca, Spain
| | - Ana M Añazco-Guenkova
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007, Salamanca, Spain
| | - Judith López
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007, Salamanca, Spain
| | - Sabine Dietmann
- Washington University School of Medicine in St. Louis, 660S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Mercedes Tomé
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Sevilla, Spain
| | - Sonia Jimeno
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Sevilla, Spain
- Departamento de Genética, Universidad de Sevilla, Sevilla, Spain
| | - Mikel Azkargorta
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 801 bld., 48160, Derio, Bizkaia, Spain
- Carlos III Networked Proteomics Platform (ProteoRed-ISCIII), Madrid, Spain
| | - Félix Elortza
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 801 bld., 48160, Derio, Bizkaia, Spain
- Carlos III Networked Proteomics Platform (ProteoRed-ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Laura Bárcena
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 801 bld., 48160, Derio, Bizkaia, Spain
| | - Monika Gonzalez-Lopez
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 801 bld., 48160, Derio, Bizkaia, Spain
| | - Ana M Aransay
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 801 bld., 48160, Derio, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Manuel A Sánchez-Martín
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007, Salamanca, Spain
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Pablo Huertas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Sevilla, Spain
- Departamento de Genética, Universidad de Sevilla, Sevilla, Spain
| | - Raúl V Durán
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Sevilla, Spain
| | - Sandra Blanco
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain.
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
13
|
van Amerongen R, Bentires-Alj M, van Boxtel AL, Clarke RB, Fre S, Suarez EG, Iggo R, Jechlinger M, Jonkers J, Mikkola ML, Koledova ZS, Sørlie T, Vivanco MDM. Imagine beyond: recent breakthroughs and next challenges in mammary gland biology and breast cancer research. J Mammary Gland Biol Neoplasia 2023; 28:17. [PMID: 37450065 PMCID: PMC10349020 DOI: 10.1007/s10911-023-09544-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/25/2023] [Indexed: 07/18/2023] Open
Abstract
On 8 December 2022 the organizing committee of the European Network for Breast Development and Cancer labs (ENBDC) held its fifth annual Think Tank meeting in Amsterdam, the Netherlands. Here, we embraced the opportunity to look back to identify the most prominent breakthroughs of the past ten years and to reflect on the main challenges that lie ahead for our field in the years to come. The outcomes of these discussions are presented in this position paper, in the hope that it will serve as a summary of the current state of affairs in mammary gland biology and breast cancer research for early career researchers and other newcomers in the field, and as inspiration for scientists and clinicians to move the field forward.
Collapse
Affiliation(s)
- Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| | - Mohamed Bentires-Alj
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Antonius L van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Robert B Clarke
- Manchester Breast Centre, Division of Cancer Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Silvia Fre
- Institut Curie, Genetics and Developmental Biology Department, PSL Research University, CNRS UMR3215, U93475248, InsermParis, France
| | - Eva Gonzalez Suarez
- Transformation and Metastasis Laboratory, Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Richard Iggo
- INSERM U1312, University of Bordeaux, 33076, Bordeaux, France
| | - Martin Jechlinger
- Cell Biology and Biophysics Department, EMBL, Heidelberg, Germany
- Molit Institute of Personalized Medicine, Heilbronn, Germany
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Marja L Mikkola
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, P.O.B. 56, 00014, Helsinki, Finland
| | - Zuzana Sumbalova Koledova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Therese Sørlie
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Maria dM Vivanco
- Cancer Heterogeneity Lab, CIC bioGUNE, Basque Research and Technology Alliance, BRTA, Technological Park Bizkaia, 48160, Derio, Spain
| |
Collapse
|
14
|
Pitzen SP, Dehm SM. Basal epithelial cells in prostate development, tumorigenesis, and cancer progression. Cell Cycle 2023; 22:1303-1318. [PMID: 37098827 PMCID: PMC10228417 DOI: 10.1080/15384101.2023.2206502] [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: 10/05/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 04/27/2023] Open
Abstract
The prostate epithelium is composed of two predominant cell populations: luminal and basal epithelial cells. Luminal cells have a secretory function that supports male fertility while basal cells function in regeneration and maintenance of epithelial tissue. Recent studies in humans and mice have expanded our knowledge of the role and regulation of luminal and basal cells in prostate organogenesis, development, and homeostasis. The insights from healthy prostate biology can inform studies focused on the origins of prostate cancer, progression of the disease, and development of resistance to targeted hormonal therapies. In this review, we discuss a critical role for basal cells in the development and maintenance of healthy prostate tissue. Additionally, we provide evidence supporting a role for basal cells in oncogenesis and therapeutic resistance mechanisms of prostate cancer. Finally, we describe basal cell regulators that may promote lineage plasticity and basal cell identity in prostate cancers that have developed therapeutic resistance. These regulators could serve as therapeutic targets to inhibit or delay resistance and thereby improve outcomes for prostate cancer patients.
Collapse
Affiliation(s)
- Samuel P. Pitzen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Molecular, Cellular, and Developmental Biology and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
15
|
Beshiri M, Agarwal S, Yin JJ, Kelly K. Prostate organoids: emerging experimental tools for translational research. J Clin Invest 2023; 133:169616. [PMID: 37183816 PMCID: PMC10178834 DOI: 10.1172/jci169616] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Organoid technology has provided new translational research opportunities in oncology, in part by enabling the development of patient-representative living biobanks. Prostate cancer research historically has been constrained to a small number of in vitro models, limiting the ability to translate experimental conclusions for contemporary, heterogeneous patient populations. The facility of organoid culture methods to maintain luminal prostate epithelia, the common lineage of prostate cancers, has greatly expanded the phenotypic and genotypic diversity of available tractable models, including luminal stem/progenitor cells and progressive patient-derived cancers. Biobanks of patient prostate cancer organoids enable increased accuracy in predicting therapeutic efficacy and informative clinical trial designs. Here, we discuss how prostate organoid technology is currently being used, the promising areas of future therapeutic applications, and the current obstacles to be overcome.
Collapse
|
16
|
Zhou Y, Li T, Jia M, Dai R, Wang R. The Molecular Biology of Prostate Cancer Stem Cells: From the Past to the Future. Int J Mol Sci 2023; 24:ijms24087482. [PMID: 37108647 PMCID: PMC10140972 DOI: 10.3390/ijms24087482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Prostate cancer (PCa) continues to rank as the second leading cause of cancer-related mortality in western countries, despite the golden treatment using androgen deprivation therapy (ADT) or anti-androgen therapy. With decades of research, scientists have gradually realized that the existence of prostate cancer stem cells (PCSCs) successfully explains tumor recurrence, metastasis and therapeutic failure of PCa. Theoretically, eradication of this small population may improve the efficacy of current therapeutic approaches and prolong PCa survival. However, several characteristics of PCSCs make their diminishment extremely challenging: inherent resistance to anti-androgen and chemotherapy treatment, over-activation of the survival pathway, adaptation to tumor micro-environments, escape from immune attack and being easier to metastasize. For this end, a better understanding of PCSC biology at the molecular level will definitely inspire us to develop PCSC targeted approaches. In this review, we comprehensively summarize signaling pathways responsible for homeostatic regulation of PCSCs and discuss how to eliminate these fractional cells in clinical practice. Overall, this study deeply pinpoints PCSC biology at the molecular level and provides us some research perspectives.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Tian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Man Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Rongyang Dai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Ronghao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
17
|
Wissmiller K, Bilekova S, Franko A, Lutz SZ, Katsburg M, Gulde S, Pellegata NS, Stenzl A, Heni M, Berti L, Häring HU, Lickert H. Inceptor correlates with markers of prostate cancer progression and modulates insulin/IGF1 signaling and cancer cell migration. Mol Metab 2023; 71:101706. [PMID: 36931467 PMCID: PMC10074927 DOI: 10.1016/j.molmet.2023.101706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/21/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
OBJECTIVE The insulin/insulin-like growth factor 1 (IGF1) pathway is emerging as a crucial component of prostate cancer progression. Therefore, we investigated the role of the novel insulin/IGF1 signaling modulator inceptor in prostate cancer. METHODS We analyzed the expression of inceptor in human samples of benign prostate epithelium and prostate cancer. Further, we performed signaling and functional assays using prostate cancer cell lines. RESULTS We found that inceptor was expressed in human benign and malignant prostate tissue and its expression positively correlated with various genes of interest, including genes involved in androgen signaling. In vitro, total levels of inceptor were increased upon androgen deprivation and correlated with high levels of androgen receptor in the nucleus. Inceptor overexpression was associated with increased cell migration, altered IGF1R trafficking and higher IGF1R activation. CONCLUSIONS Our in vitro results showed that inceptor expression was associated with androgen status, increased migration, and IGF1R signaling. In human samples, inceptor expression was significantly correlated with markers of prostate cancer progression. Taken together, these data provide a basis for investigation of inceptor in the context of prostate cancer.
Collapse
Affiliation(s)
- Katharina Wissmiller
- Institute of Diabetes and Regeneration Research at the Helmholtz Center Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, Ismaninger Str. 22, 81675, Munich, Germany
| | - Sara Bilekova
- Institute of Diabetes and Regeneration Research at the Helmholtz Center Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, Ismaninger Str. 22, 81675, Munich, Germany
| | - Andras Franko
- German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; Institute of Diabetes and Metabolic Disease at the Helmholtz Center Munich, Ottfried-Müller-Str. 10, 72076, Tübingen, Germany; Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Ottfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Stefan Z Lutz
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Ottfried-Müller-Str. 10, 72076, Tübingen, Germany; Clinic for Geriatric and Orthopedic Rehabilitation Bad Sebastiansweiler, Hechinger Str. 26, 72116, Mössingen, Germany
| | - Miriam Katsburg
- Institute of Diabetes and Regeneration Research at the Helmholtz Center Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Sebastian Gulde
- Institute of Diabetes and Cancer at the Helmholtz Center Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Natalia S Pellegata
- Institute of Diabetes and Cancer at the Helmholtz Center Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Martin Heni
- German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; Institute of Diabetes and Metabolic Disease at the Helmholtz Center Munich, Ottfried-Müller-Str. 10, 72076, Tübingen, Germany; Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Ottfried-Müller-Str. 10, 72076, Tübingen, Germany; Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Lucia Berti
- German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; Institute of Diabetes and Metabolic Disease at the Helmholtz Center Munich, Ottfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; Institute of Diabetes and Metabolic Disease at the Helmholtz Center Munich, Ottfried-Müller-Str. 10, 72076, Tübingen, Germany; Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Ottfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research at the Helmholtz Center Munich, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany; Technical University of Munich, School of Medicine, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
18
|
Chen J, Zheng Q, Hicks JL, Trabzonlu L, Ozbek B, Jones T, Vaghasia A, Larman TC, Wang R, Markowski MC, Denmeade SR, Pienta KJ, Hruban RH, Antonarakis ES, Gupta A, Dang CV, Yegnasubramanian S, De Marzo AM. MYC-driven increases in mitochondrial DNA copy number occur early and persist throughout prostatic cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.20.529259. [PMID: 36865273 PMCID: PMC9979994 DOI: 10.1101/2023.02.20.529259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Increased mitochondrial function may render some cancers vulnerable to mitochondrial inhibitors. Since mitochondrial function is regulated partly by mitochondrial DNA copy number (mtDNAcn), accurate measurements of mtDNAcn could help reveal which cancers are driven by increased mitochondrial function and may be candidates for mitochondrial inhibition. However, prior studies have employed bulk macrodissections that fail to account for cell type-specific or tumor cell heterogeneity in mtDNAcn. These studies have often produced unclear results, particularly in prostate cancer. Herein, we developed a multiplex in situ method to spatially quantify cell type specific mtDNAcn. We show that mtDNAcn is increased in luminal cells of high-grade prostatic intraepithelial neoplasia (HGPIN), is increased in prostatic adenocarcinomas (PCa), and is further elevated in metastatic castration-resistant prostate cancer. Increased PCa mtDNAcn was validated by two orthogonal methods and is accompanied by increases in mtRNAs and enzymatic activity. Mechanistically, MYC inhibition in prostate cancer cells decreases mtDNA replication and expression of several mtDNA replication genes, and MYC activation in the mouse prostate leads to increased mtDNA levels in the neoplastic prostate cells. Our in situ approach also revealed elevated mtDNAcn in precancerous lesions of the pancreas and colon/rectum, demonstrating generalization across cancer types using clinical tissue samples.
Collapse
Affiliation(s)
- Jiayu Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qizhi Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jessica L. Hicks
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Levent Trabzonlu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Busra Ozbek
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tracy Jones
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ajay Vaghasia
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tatianna C. Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rulin Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark C. Markowski
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sam R. Denmeade
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kenneth J. Pienta
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ralph H. Hruban
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Emmanuel S. Antonarakis
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anuj Gupta
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chi V Dang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Srinivasan Yegnasubramanian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Luo F, Tshering LF, Tutuska K, Szenk M, Rubel D, Rail JG, Russ S, Liu J, Nemajerova A, Balázsi G, Talos F. A luminal intermediate cell state maintains long-term prostate homeostasis and contributes to tumorigenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529762. [PMID: 36909551 PMCID: PMC10002646 DOI: 10.1101/2023.02.24.529762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Cellular heterogeneity poses tremendous challenges for developing cell-targeted therapies and biomarkers of clinically significant prostate cancer. The origins of this heterogeneity within normal adult and aging tissue remain unknown, leaving cellular states and transcriptional programs that allow expansions of malignant clones unidentified. To define cell states that contribute to early cancer development, we performed clonal analyses and single cell transcriptomics of normal prostate from genetically-engineered mouse models. We uncovered a luminal transcriptional state with a unique "basal-like" Wnt/p63 signaling ( luminal intermediate , LumI) which contributes to the maintenance of long-term prostate homeostasis. Moreover, LumI cells greatly expand during early stages of tumorigenesis in several mouse models of prostate cancer. Genetic ablation of p63 in vivo in luminal cells reduced the formation of aggressive clones in mouse prostate tumor models. Finally, the LumI cells and Wnt signaling appear to significantly increase in human aging prostate and prostate cancer samples, highlighting the importance of this hybrid cell state for human pathologies with potential translational impact.
Collapse
|
20
|
Hirz T, Mei S, Sarkar H, Kfoury Y, Wu S, Verhoeven BM, Subtelny AO, Zlatev DV, Wszolek MW, Salari K, Murray E, Chen F, Macosko EZ, Wu CL, Scadden DT, Dahl DM, Baryawno N, Saylor PJ, Kharchenko PV, Sykes DB. Dissecting the immune suppressive human prostate tumor microenvironment via integrated single-cell and spatial transcriptomic analyses. Nat Commun 2023; 14:663. [PMID: 36750562 PMCID: PMC9905093 DOI: 10.1038/s41467-023-36325-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
The treatment of low-risk primary prostate cancer entails active surveillance only, while high-risk disease requires multimodal treatment including surgery, radiation therapy, and hormonal therapy. Recurrence and development of metastatic disease remains a clinical problem, without a clear understanding of what drives immune escape and tumor progression. Here, we comprehensively describe the tumor microenvironment of localized prostate cancer in comparison with adjacent normal samples and healthy controls. Single-cell RNA sequencing and high-resolution spatial transcriptomic analyses reveal tumor context dependent changes in gene expression. Our data indicate that an immune suppressive tumor microenvironment associates with suppressive myeloid populations and exhausted T-cells, in addition to high stromal angiogenic activity. We infer cell-to-cell relationships from high throughput ligand-receptor interaction measurements within undissociated tissue sections. Our work thus provides a highly detailed and comprehensive resource of the prostate tumor microenvironment as well as tumor-stromal cell interactions.
Collapse
Affiliation(s)
- Taghreed Hirz
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| | - Shenglin Mei
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
| | - Hirak Sarkar
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Youmna Kfoury
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Shulin Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bronte M Verhoeven
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Alexander O Subtelny
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dimitar V Zlatev
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew W Wszolek
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keyan Salari
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Evan Murray
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Evan Z Macosko
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Douglas M Dahl
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Philip J Saylor
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Peter V Kharchenko
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Altos Labs, San Diego, CA, USA
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
21
|
Han S, Chen X, Li Z. Innate Immune Program in Formation of Tumor-Initiating Cells from Cells-of-Origin of Breast, Prostate, and Ovarian Cancers. Cancers (Basel) 2023; 15:757. [PMID: 36765715 PMCID: PMC9913549 DOI: 10.3390/cancers15030757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Tumor-initiating cells (TICs), also known as cancer stem cells (CSCs), are cancer cells that can initiate a tumor, possess self-renewal capacity, and can contribute to tumor heterogeneity. TICs/CSCs are developed from their cells-of-origin. In breast, prostate, and ovarian cancers, progenitor cells for mammary alveolar cells, prostate luminal (secretory) cells, and fallopian tube secretory cells are the preferred cellular origins for their corresponding cancer types. These luminal progenitors (LPs) express common innate immune program (e.g., Toll-like receptor (TLR) signaling)-related genes. Microbes such as bacteria are now found in breast, prostate, and fallopian tube tissues and their corresponding cancer types, raising the possibility that their LPs may sense the presence of microbes and trigger their innate immune/TLR pathways, leading to an inflammatory microenvironment. Crosstalk between immune cells (e.g., macrophages) and affected epithelial cells (e.g., LPs) may eventually contribute to formation of TICs/CSCs from their corresponding LPs, in part via STAT3 and/or NFκB pathways. As such, TICs/CSCs can inherit expression of innate-immunity/TLR-pathway-related genes from their cells-of-origin; the innate immune program may also represent their unique vulnerability, which can be explored therapeutically (e.g., by enhancing immunotherapy via augmenting TLR signaling).
Collapse
Affiliation(s)
- Sen Han
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Xueqing Chen
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Zhe Li
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
22
|
Seo E, Kang M. Current status and clinical application of patient-derived tumor organoid model in kidney and prostate cancers. BMB Rep 2023; 56:24-31. [PMID: 36476272 PMCID: PMC9887101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 01/28/2023] Open
Abstract
Urological cancers such as kidney, bladder, prostate, and testicular cancers are the most common types of cancers worldwide with high mortality and morbidity. To date, traditional cell lines and animal models have been broadly used to study pre-clinical applications and underlying molecular mechanisms of urological cancers. However, they cannot reflect biological phenotypes of real tissues and clinical diversities of urological cancers in vitro system. In vitro models cannot be utilized to reflect the tumor microenvironment or heterogeneity. Cancer organoids in three-dimensional culture have emerged as a promising platform for simulating tumor microenvironment and revealing heterogeneity. In this review, we summarize recent advances in prostate and kidney cancer organoids regarding culture conditions, advantages, and applications of these cancer organoids. [BMB Reports 2023; 56(1): 24-31].
Collapse
Affiliation(s)
- Eunjeong Seo
- Molecular Pharmacology, OliPass Corporation, Yongin 17015, Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Seoul 06351, Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
23
|
Seo E, Kang M. Current status and clinical application of patient-derived tumor organoid model in kidney and prostate cancers. BMB Rep 2023; 56:24-31. [PMID: 36476272 PMCID: PMC9887101 DOI: 10.5483/bmbrep.2022-0200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 11/30/2023] Open
Abstract
Urological cancers such as kidney, bladder, prostate, and testicular cancers are the most common types of cancers worldwide with high mortality and morbidity. To date, traditional cell lines and animal models have been broadly used to study pre-clinical applications and underlying molecular mechanisms of urological cancers. However, they cannot reflect biological phenotypes of real tissues and clinical diversities of urological cancers in vitro system. In vitro models cannot be utilized to reflect the tumor microenvironment or heterogeneity. Cancer organoids in three-dimensional culture have emerged as a promising platform for simulating tumor microenvironment and revealing heterogeneity. In this review, we summarize recent advances in prostate and kidney cancer organoids regarding culture conditions, advantages, and applications of these cancer organoids. [BMB Reports 2023; 56(1): 24-31].
Collapse
Affiliation(s)
- Eunjeong Seo
- Molecular Pharmacology, OliPass Corporation, Yongin 17015, Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Seoul 06351, Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
24
|
Kukkonen K, Autio-Kimura B, Rauhala H, Kesseli J, Nykter M, Latonen L, Visakorpi T. Nonmalignant AR-positive prostate epithelial cells and cancer cells respond differently to androgen. Endocr Relat Cancer 2022; 29:717-733. [PMID: 36219867 PMCID: PMC9644224 DOI: 10.1530/erc-22-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
Prostate cancer research suffers from the lack of suitable models to study the role of normal cells in prostate carcinogenesis. To address this challenge, we developed a cell line model mimicking luminal prostate epithelial cells by modifying the immortalized prostate epithelial cell line RWPE-1 to constitutively express the androgen receptor (AR). RWPE-1-AR cells express known AR target genes, and exhibit coexpression of luminal and basal markers characteristic of transient amplifying cells, and an RNA signature resembling prostate luminal progenitor cells. Under unstimulated conditions, constitutive AR expression does not have a biologically significant effect on the proliferation of RWPE-1 cells, but when stimulated by androgens, growth is retarded. The transcriptional response of RWPE-1-AR cells to androgen stimulation involves suppression of the growth-related KRAS pathway and is thus markedly different from that of the prostate cancer cell line LNCaP and its derivative AR-overexpressing LNCaP-ARhi cells, in which growth- and cancer-related pathways are upregulated. Hence, the nonmalignant AR-positive RWPE-1-AR cell line model could be used to study the transformation of the prostate epithelium.
Collapse
Affiliation(s)
- Konsta Kukkonen
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Bryn Autio-Kimura
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Hanna Rauhala
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Juha Kesseli
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Matti Nykter
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Leena Latonen
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Tapio Visakorpi
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
- Fimlab Laboratories Ltd, Tampere, Finland
| |
Collapse
|
25
|
Pramanik SD, Kumar Halder A, Mukherjee U, Kumar D, Dey YN, R M. Potential of histone deacetylase inhibitors in the control and regulation of prostate, breast and ovarian cancer. Front Chem 2022; 10:948217. [PMID: 36034650 PMCID: PMC9411967 DOI: 10.3389/fchem.2022.948217] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylases (HDACs) are enzymes that play a role in chromatin remodeling and epigenetics. They belong to a specific category of enzymes that eliminate the acetyl part of the histones' -N-acetyl lysine, causing the histones to be wrapped compactly around DNA. Numerous biological processes rely on HDACs, including cell proliferation and differentiation, angiogenesis, metastasis, gene regulation, and transcription. Epigenetic changes, specifically increased expression and activity of HDACs, are commonly detected in cancer. As a result, HDACi could be used to develop anticancer drugs. Although preclinical outcomes with HDACs as monotherapy have been promising clinical trials have had mixed results and limited success. In both preclinical and clinical trials, however, combination therapy with different anticancer medicines has proved to have synergistic effects. Furthermore, these combinations improved efficacy, decreased tumor resistance to therapy, and decreased toxicity. In the present review, the detailed modes of action, classification of HDACs, and their correlation with different cancers like prostate, breast, and ovarian cancer were discussed. Further, the different cell signaling pathways and the structure-activity relationship and pharmaco-toxicological properties of the HDACi, and their synergistic effects with other anticancer drugs observed in recent preclinical and clinical studies used in combination therapy were discussed for prostate, breast, and ovarian cancer treatment.
Collapse
Affiliation(s)
- Siddhartha Das Pramanik
- Department of Pharmaceutical Engineering and Technology, IIT-BHU, Varanasi, Uttar Pradesh, India
| | - Amit Kumar Halder
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, West Bengal, India
| | - Ushmita Mukherjee
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, West Bengal, India
| | - Dharmendra Kumar
- Department of Pharmaceutical Chemistry, Narayan Institute of Pharmacy, Gopal Narayan Singh University, Sasaram, Bihar, India
| | - Yadu Nandan Dey
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, West Bengal, India
| | - Mogana R
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI Education SDN.BHD., Kuala Lumpur, Malaysia
| |
Collapse
|
26
|
Xu Y, Mu J, Zhou Z, Leng Y, Yu Y, Song X, Liu A, Zhu H, Li J, Wang D. Expansion of mouse castration-resistant intermediate prostate stem cells in vitro. Stem Cell Res Ther 2022; 13:299. [PMID: 35841025 PMCID: PMC9284701 DOI: 10.1186/s13287-022-02978-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/06/2022] [Indexed: 11/10/2022] Open
Abstract
Background Most castration-resistant prostate cancers (CRPCs) have a luminal phenotype with high androgen receptor (AR) and prostate-specific antigen (PSA) expression. Currently, it is difficult to culture castration-resistant luminal cells with AR and PSA expression. Methods We formulated a custom-made medium and isolated primary cells from the prostate of adult wild-type (WT) and TRAMP mice. The cells were characterized by immunofluorescence staining, transcriptomic analysis, and qRT-PCR verification. Their self-renewal and differentiation potential in vitro and in vivo were examined. We treated the cells with androgen deprivation and enzalutamide and performed immunofluorescence staining and western blotting to analyze their expression of AR and PSA. Results We isolated a novel type of castration-resistant intermediate prostate stem cells (CRIPSCs) from adult WT and TRAMP mice. The mouse CRIPSCs proliferated rapidly in two-dimensional (2D) culture dishes and can be cultured for more than six months. The mouse CRIPSCs expressed luminal markers (AR, PSA, and Dsg4), basal markers (CK5 and p63), Psca, and the intermediate cell marker (Ivl). Transcriptomic analysis showed that the mouse CRIPSCs had upregulated signaling pathways related to cancer development and drug resistance. In the long-term culture, TRAMP CRIPSCs had higher expression of the genes related to stem cells and cancers than WT mice. Both WT and TRAMP CRIPSCs formed organoids in Matrigel. WT CRIPSCs did not form prostate tissues when transplanted in vivo without urogenital sinus mesenchyme (UGM) cells. In contrast, TRAMP CRIPSCs formed prostate ducts in NOG mice without UGM cells and differentiated into luminal, basal, and neuroendocrine cells. Androgens regulated AR translocation between the nucleus and cytoplasm in the mouse CRIPSCs. Treatment of androgen deprivation (ADT) and enzalutamide reduced AR expression in WT and TRAMP CRIPSCs; however, this treatment promoted PSA expression in TRAMP, while not WT CRIPSCs, similar to the clinical observations of CRPC. Conclusions Our study established a method for isolating and expanding mouse CRIPSCs in 2D culture dishes. Mouse CRIPSCs had markers of basal and luminal cells, including AR and PSA, and can differentiate into prostate organoids and tissues. TRAMP CRIPSCs had elevated PSA expression upon ADT and enzalutamide treatment. Our method can be translated into clinical settings for CRPC precision medicine.
Collapse
Affiliation(s)
- Yalan Xu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China.,School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Jie Mu
- College of Life Sciences, and School of Pharmacy, Medical College, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Zhixia Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China
| | - Yu Leng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China.,School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Yali Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China.,School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Xiuyue Song
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China.,School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Aihua Liu
- College of Life Sciences, and School of Pharmacy, Medical College, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, 266011, China.
| | - Jing Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China.
| | - Dong Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
27
|
Bleeker J, Wang ZA. Applications of Vertebrate Models in Studying Prostatitis and Inflammation-Associated Prostatic Diseases. Front Mol Biosci 2022; 9:898871. [PMID: 35865005 PMCID: PMC9294738 DOI: 10.3389/fmolb.2022.898871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/17/2022] [Indexed: 12/05/2022] Open
Abstract
It has long been postulated that the inflammatory environment favors cell proliferation, and is conducive to diseases such as cancer. In the prostate gland, clinical data implicate important roles of prostatitis in the progression of both benign prostatic hyperplasia (BPH) and prostate cancer (PCa). However, their causal relationships have not been firmly established yet due to unresolved molecular and cellular mechanisms. By accurately mimicking human disease, vertebrate animals provide essential in vivo models to address this question. Here, we review the vertebrate prostatitis models that have been developed and discuss how they may reveal possible mechanisms by which prostate inflammation promotes BPH and PCa. Recent studies, particularly those involving genetically engineered mouse models (GEMMs), suggest that such mechanisms are multifaceted, which include epithelium barrier disruption, DNA damage and cell proliferation induced by paracrine signals, and expansion of potential cells of origin for cancer. Future research using rodent prostatitis models should aim to distinguish the etiologies of BPH and PCa, and facilitate the development of novel clinical approaches for prostatic disease prevention.
Collapse
|
28
|
Liu Y, Wang J, Horton C, Yu C, Knudsen B, Stefanson J, Hu K, Stefanson O, Green J, Guo C, Xie Q, Wang ZA. Stromal AR inhibits prostate tumor progression by restraining secretory luminal epithelial cells. Cell Rep 2022; 39:110848. [PMID: 35613593 PMCID: PMC9175887 DOI: 10.1016/j.celrep.2022.110848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/03/2022] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Androgen receptor (AR) is expressed in both the prostate epithelium and the prostate stroma and plays diverse roles in prostate physiology. Although low expression of stromal AR is clinically associated with advanced cancer stage and worse outcome, whether stromal AR inhibits or promotes prostate cancer progression remains controversial. Here, we specifically delete AR in smooth muscle cells of the adult mouse prostate under two tumorigenic conditions, namely, the Hi-Myc genetic model and the T + E2 hormonal carcinogenesis model. Histology analyses show that stromal AR deletion exacerbates tumor progression phenotypes in both models. Furthermore, single-cell analyses of the tumor samples reveal that secretory luminal cells are the cell population particularly affected by stromal AR deletion, as they transition to a cellular state of potentiated PI3K-mTORC1 activities. Our results suggest that stromal AR normally inhibits prostate cancer progression by restraining secretory luminal cells and imply possible unintended negative effects of androgen deprivation therapy.
Collapse
Affiliation(s)
- Yueli Liu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jiawen Wang
- Sequencing Center, National Institute of Biological Sciences, Beijing 102206, China
| | - Corrigan Horton
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Chuan Yu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Beatrice Knudsen
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua Stefanson
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kevin Hu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Ofir Stefanson
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jonathan Green
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Charlene Guo
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Qing Xie
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Zhu A Wang
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
29
|
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]
|
30
|
Harris AE, Metzler VM, Lothion-Roy J, Varun D, Woodcock CL, Haigh DB, Endeley C, Haque M, Toss MS, Alsaleem M, Persson JL, Gudas LJ, Rakha E, Robinson BD, Khani F, Martin LM, Moyer JE, Brownlie J, Madhusudan S, Allegrucci C, James VH, Rutland CS, Fray RG, Ntekim A, de Brot S, Mongan NP, Jeyapalan JN. Exploring anti-androgen therapies in hormone dependent prostate cancer and new therapeutic routes for castration resistant prostate cancer. Front Endocrinol (Lausanne) 2022; 13:1006101. [PMID: 36263323 PMCID: PMC9575553 DOI: 10.3389/fendo.2022.1006101] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Androgen deprivation therapies (ADTs) are important treatments which inhibit androgen-induced prostate cancer (PCa) progression by either preventing androgen biosynthesis (e.g. abiraterone) or by antagonizing androgen receptor (AR) function (e.g. bicalutamide, enzalutamide, darolutamide). A major limitation of current ADTs is they often remain effective for limited durations after which patients commonly progress to a lethal and incurable form of PCa, called castration-resistant prostate cancer (CRPC) where the AR continues to orchestrate pro-oncogenic signalling. Indeed, the increasing numbers of ADT-related treatment-emergent neuroendocrine-like prostate cancers (NePC), which lack AR and are thus insensitive to ADT, represents a major therapeutic challenge. There is therefore an urgent need to better understand the mechanisms of AR action in hormone dependent disease and the progression to CRPC, to enable the development of new approaches to prevent, reverse or delay ADT-resistance. Interestingly the AR regulates distinct transcriptional networks in hormone dependent and CRPC, and this appears to be related to the aberrant function of key AR-epigenetic coregulator enzymes including the lysine demethylase 1 (LSD1/KDM1A). In this review we summarize the current best status of anti-androgen clinical trials, the potential for novel combination therapies and we explore recent advances in the development of novel epigenetic targeted therapies that may be relevant to prevent or reverse disease progression in patients with advanced CRPC.
Collapse
Affiliation(s)
- Anna E. Harris
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Veronika M. Metzler
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Jennifer Lothion-Roy
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Dhruvika Varun
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Corinne L. Woodcock
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Daisy B. Haigh
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Chantelle Endeley
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Maria Haque
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Michael S. Toss
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Mansour Alsaleem
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
- Department of Applied Medical Science, Applied College, Qassim University, Qassim, Saudi Arabia
| | - Jenny L. Persson
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Department of Biomedical Sciences, Malmö Universitet, Malmö, Sweden
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Emad Rakha
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Brian D. Robinson
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
| | - Francesca Khani
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
| | - Laura M. Martin
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Jenna E. Moyer
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Juliette Brownlie
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Srinivasan Madhusudan
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Cinzia Allegrucci
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Victoria H. James
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Catrin S. Rutland
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Rupert G. Fray
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
| | - Atara Ntekim
- Department of Oncology, University Hospital Ibadan, Ibadan, Nigeria
- *Correspondence: Jennie N. Jeyapalan, ; Nigel P. Mongan, ; ; Atara Ntekim,
| | - Simone de Brot
- Comparative Pathology Platform (COMPATH), Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Nigel P. Mongan
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Jennie N. Jeyapalan, ; Nigel P. Mongan, ; ; Atara Ntekim,
| | - Jennie N. Jeyapalan
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
- *Correspondence: Jennie N. Jeyapalan, ; Nigel P. Mongan, ; ; Atara Ntekim,
| |
Collapse
|
31
|
Flores-Téllez TDNJ, Baena E. Experimental challenges to modeling prostate cancer heterogeneity. Cancer Lett 2022; 524:194-205. [PMID: 34688843 DOI: 10.1016/j.canlet.2021.10.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/23/2021] [Accepted: 10/09/2021] [Indexed: 12/24/2022]
Abstract
Tumor heterogeneity plays a key role in prostate cancer prognosis, therapy selection, relapse, and acquisition of treatment resistance. Prostate cancer presents a heterogeneous diversity at inter- and intra-tumor and inter-patient levels which are influenced by multiple intrinsic and/or extrinsic factors. Recent studies have started to characterize the complexity of prostate tumors and these different tiers of heterogeneity. In this review, we discuss the most common factors that contribute to tumoral diversity. Moreover, we focus on the description of the in vitro and in vivo approaches, as well as high-throughput technologies, that help to model intra-tumoral diversity. Further understanding tumor heterogeneities and the challenges they present will guide enhanced patient risk stratification, aid the design of more precise therapies, and ultimately help beat this chameleon-like disease.
Collapse
Affiliation(s)
- Teresita Del N J Flores-Téllez
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG, UK
| | - Esther Baena
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG, UK.
| |
Collapse
|
32
|
Tuong ZK, Loudon KW, Berry B, Richoz N, Jones J, Tan X, Nguyen Q, George A, Hori S, Field S, Lynch AG, Kania K, Coupland P, Babbage A, Grenfell R, Barrett T, Warren AY, Gnanapragasam V, Massie C, Clatworthy MR. Resolving the immune landscape of human prostate at a single-cell level in health and cancer. Cell Rep 2021; 37:110132. [PMID: 34936871 PMCID: PMC8721283 DOI: 10.1016/j.celrep.2021.110132] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 09/01/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022] Open
Abstract
The prostate gland produces prostatic fluid, high in zinc and citrate and essential for the maintenance of spermatozoa. Prostate cancer is a common condition with limited treatment efficacy in castration-resistant metastatic disease, including with immune checkpoint inhibitors. Using single-cell RNA-sequencing to perform an unbiased assessment of the cellular landscape of human prostate, we identify a subset of tumor-enriched androgen receptor-negative luminal epithelial cells with increased expression of cancer-associated genes. We also find a variety of innate and adaptive immune cells in normal prostate that were transcriptionally perturbed in prostate cancer. An exception is a prostate-specific, zinc transporter-expressing macrophage population (MAC-MT) that contributes to tissue zinc accumulation in homeostasis but shows enhanced inflammatory gene expression in tumors, including T cell-recruiting chemokines. Remarkably, enrichment of the MAC-MT signature in cancer biopsies is associated with improved disease-free survival, suggesting beneficial antitumor functions.
Collapse
Affiliation(s)
- Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Kevin W Loudon
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Brendan Berry
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Urology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Nathan Richoz
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Xiao Tan
- Division of Genetics and Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Quan Nguyen
- Division of Genetics and Genomics, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Anne George
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Early Detection Programme, CRUK Cambridge Centre, Cambridge, UK
| | - Satoshi Hori
- Academic Urology Group, Department of Surgery, University of Cambridge, Cambridge, UK
| | | | - Andy G Lynch
- CRUK Cambridge Institute, Cambridge, UK; School of Mathematics and Statistics/School of Medicine, University of St Andrews, St Andrews, UK
| | | | | | - Anne Babbage
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Tristan Barrett
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Anne Y Warren
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Vincent Gnanapragasam
- Department of Urology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Academic Urology Group, Department of Surgery, University of Cambridge, Cambridge, UK; Cambridge Urology Translational Research and Clinical Trials, Cambridge Biomedical Campus, Cambridge, UK
| | - Charlie Massie
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; CRUK Cambridge Institute, Cambridge, UK; Early Detection Programme, CRUK Cambridge Centre, Cambridge, UK.
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK; NIHR Cambridge Biomedical Research Centre, Cambridge, UK; Cambridge Institute of Therapeutic Immunology & Infectious Diseases, Cambridge, UK.
| |
Collapse
|
33
|
Cable J, Pei D, Reid LM, Wang XW, Bhatia S, Karras P, Melenhorst JJ, Grompe M, Lathia JD, Song E, Kuo CJ, Zhang N, White RM, Ma SK, Ma L, Chin YR, Shen MM, Ng IOL, Kaestner KH, Zhou L, Sikandar S, Schmitt CA, Guo W, Wong CCL, Ji J, Tang DG, Dubrovska A, Yang C, Wiedemeyer WR, Weissman IL. Cancer stem cells: advances in biology and clinical translation-a Keystone Symposia report. Ann N Y Acad Sci 2021; 1506:142-163. [PMID: 34850398 PMCID: PMC9153245 DOI: 10.1111/nyas.14719] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022]
Abstract
The test for the cancer stem cell (CSC) hypothesis is to find a target expressed on all, and only CSCs in a patient tumor, then eliminate all cells with that target that eliminates the cancer. That test has not yet been achieved, but CSC diagnostics and targets found on CSCs and some other cells have resulted in a few clinically relevant therapies. However, it has become apparent that eliminating the subset of tumor cells characterized by self-renewal properties is essential for long-term tumor control. CSCs are able to regenerate and initiate tumor growth, recapitulating the heterogeneity present in the tumor before treatment. As great progress has been made in identifying and elucidating the biology of CSCs as well as their interactions with the tumor microenvironment, the time seems ripe for novel therapeutic strategies that target CSCs to find clinical applicability. On May 19-21, 2021, researchers in cancer stem cells met virtually for the Keystone eSymposium "Cancer Stem Cells: Advances in Biology and Clinical Translation" to discuss recent advances in the understanding of CSCs as well as clinical efforts to target these populations.
Collapse
Affiliation(s)
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, China
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
| | - Lola M Reid
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sonam Bhatia
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Panagiotis Karras
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology and Laboratory for Molecular Cancer Biology, Department of Oncology, Leuven, Belgium
| | - Jan Joseph Melenhorst
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center and Department of Pathology & Laboratory Medicine, Perelman School of Medicine and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Markus Grompe
- Department of Molecular and Medical Genetics, Department of Pediatrics, and Oregon Stem Cell Center, Oregon Health & Science University, Portland, Oregon
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute and Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center and Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory; Program of Molecular Medicine, Zhongshan School of Medicine, Sun Yat-Sen University; and Fountain-Valley Institute for Life Sciences, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Calvin J Kuo
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California
| | - Ning Zhang
- Translational Cancer Research Center, Peking University First Hospital, Beijing, China
| | - Richard M White
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stephanie Ky Ma
- School of Biomedical Sciences and State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Lichun Ma
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Y Rebecca Chin
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - 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, New York
| | - Irene Oi Lin Ng
- Department of Pathology and State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lei Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, Hong Kong
| | - Shaheen Sikandar
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, California
| | - Clemens A Schmitt
- Charité - Universitätsmedizin Berlin, Hematology/Oncology, and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany, and Johannes Kepler University, Kepler Universitätsklinikum, Hematology/Oncology, Linz, Austria
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carmen Chak-Lui Wong
- Department of Pathology and State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Junfang Ji
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, and Experimental Therapeutics (ET) Graduate Program, University at Buffalo, Buffalo, New York
| | - Anna Dubrovska
- OncoRay National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Heidelberg, Germany
| | - Chunzhang Yang
- Neuro-Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | | | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Ludwig Center for Cancer Stem Cell Research, Stanford University, Stanford, California
| |
Collapse
|
34
|
Kukkonen K, Taavitsainen S, Huhtala L, Uusi-Makela J, Granberg KJ, Nykter M, Urbanucci A. Chromatin and Epigenetic Dysregulation of Prostate Cancer Development, Progression, and Therapeutic Response. Cancers (Basel) 2021; 13:3325. [PMID: 34283056 PMCID: PMC8268970 DOI: 10.3390/cancers13133325] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
The dysregulation of chromatin and epigenetics has been defined as the overarching cancer hallmark. By disrupting transcriptional regulation in normal cells and mediating tumor progression by promoting cancer cell plasticity, this process has the ability to mediate all defined hallmarks of cancer. In this review, we collect and assess evidence on the contribution of chromatin and epigenetic dysregulation in prostate cancer. We highlight important mechanisms leading to prostate carcinogenesis, the emergence of castration-resistance upon treatment with androgen deprivation therapy, and resistance to antiandrogens. We examine in particular the contribution of chromatin structure and epigenetics to cell lineage commitment, which is dysregulated during tumorigenesis, and cell plasticity, which is altered during tumor progression.
Collapse
Affiliation(s)
- Konsta Kukkonen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Sinja Taavitsainen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Laura Huhtala
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Joonas Uusi-Makela
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Kirsi J. Granberg
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, 33520 Tampere, Finland; (K.K.); (S.T.); (L.H.); (J.U.-M.); (K.J.G.); (M.N.)
| | - Alfonso Urbanucci
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
35
|
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
|
36
|
Wang L, Gao J, Zhang Y, Kang S. Silencing miRNA-1297 suppresses the invasion and migration of prostate cancer cells via targeting modulation of PTEN and blocking of the AKT/ERK pathway. Exp Ther Med 2021; 22:768. [PMID: 34055067 PMCID: PMC8145438 DOI: 10.3892/etm.2021.10200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
Phosphatase and tensin homolog (PTEN) loss is a major contributing factor of prostate cancer (PC). miRNA-1297 was reported to serve role in various cancer types; however, the potential roles of miRNA-1297 in PC had not been investigated. In the present study, tumor and adjacent tissues were collected from patients with PC. The gene expression level of miRNA-1297 was measured via polymerase chain reaction. Results indicated that the miRNA-1297 was overexpressed in tumor tissues from PC patients and in PC cell lines. miRNA-1297 also contributed toward the progression of PC. PTEN was confirmed as the direct target of miRNA-1297 and bound with miRNA-1297 via four binding sites. The miRNA-1297 level was negatively associated with the PTEN level. Silencing miRNA-1297 or overexpression of PTEN significantly inhibited the cell migration and invasion. In addition, the AKT/ERK pathway was also inhibited following silencing of miRNA-1297 or overexpression of PTEN. Taken together, the results indicated that silencing miRNA-1297 exerted inhibitory effects on the invasion and migration of PC cells via modulating PTEN and blocking of the AKT/ERK pathway. The results of the present study provided a novel strategy for treatment of prostate cancer cells.
Collapse
Affiliation(s)
- Lei Wang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Jing Gao
- Department of Obstetrics and Gynecology, Tangshan Hongci Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yu Zhang
- Department of Intensive Care Units, Tangshan People's Hospital, Tangshan, Hebei 063000, P.R. China
| | - Shaosan Kang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
37
|
Zhu J, Liang X, Wu D, Chen S, Yang B, Mao W, Shen D. Clinicopathological characteristics and survival outcomes in neuroendocrine prostate cancer: A population-based study. Medicine (Baltimore) 2021; 100:e25237. [PMID: 33847621 PMCID: PMC8052035 DOI: 10.1097/md.0000000000025237] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 02/25/2021] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the clinicopathological features and the survival outcomes of neuroendocrine prostate cancer (NEPC). METHODS Within the Surveillance, Epidemiology, and End Results (SEER) database of the National Cancer Institute, we identified a total of 510 patients with NEPC between 2006 and 2015. Age-adjusted incidence rates were evaluated in the study by the SEER∗Stat Software version 8.3.6. Kaplan-Meier analysed assessed overall survival (OS) after stratification according to marital status, age, histologic subtype, metastatic status, and treatment. The significant differences were assessed in a log-rank test. Univariate and multivariate cox hazard regression analysis were performed to determine independent predictors of OS. RESULTS From a total of 560,124 patients with prostate cancer diagnosed between 2006 and 2015, we identified 510 cases of de novo NEPC. Regarding histology, among all the NEPC, 329 (64.5%) patients were diagnosed as small cell carcinoma, 181 (39.8%) were nonsmall cell carcinoma. The overall age-adjusted incidence of NEPC statistically significantly increased from 0.321/1,000,000 person-years in 2006 to 0.587/1,000,000 person-years in 2015. The median OS in our study cohort was 9 months (95% CI, 8-10 months). Multivariate cox regression analysis showed that age, histologic subtype, and stage were independent prognostic factors for NEPC patients. The majority of NEPC (78.2%) were metastatic at diagnosis. In terms of treatment, for metastatic tumor patients, chemotherapy was the most effective therapy. Chemotherapy increased the OS of patients with regional (distant) metastases from 8 months (5 months) to 13.5 months (9 months). CONCLUSION NEPC is extremely rare but the incidence of NEPC has been increasing in the past years. The prognosis of NEPC is poor because most cases are diagnosed at metastatic stage. The patients with metastases are typically treated with chemotherapy and chemotherapy shows survival benefits in both regional and distant metastatic tumor patients.
Collapse
Affiliation(s)
- Jiamin Zhu
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin
| | - Xiao Liang
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin
| | - Dan Wu
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin
| | - Shusen Chen
- Department of Radiation Oncology, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Baixia Yang
- Department of Radiation Oncology, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Weidong Mao
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin
| | - Dong Shen
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin
| |
Collapse
|
38
|
Zahalka AH, Brodin NP, Maryanovich M, Wang X, Watts KL, Pinho S, Guha C, Frenette PS. Using CT-guided stereotactic prostate radiation therapy (CT-SPRT) to assess sustained murine prostate ablation. Sci Rep 2021; 11:6571. [PMID: 33753857 PMCID: PMC7985301 DOI: 10.1038/s41598-021-86067-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/09/2021] [Indexed: 11/09/2022] Open
Abstract
The prostate is a hormone-responsive organ where testicular androgens drive the proliferation and survival of prostatic cells, ensuring the development and functioning of this gland throughout life. Androgen deprivation therapy leads to apoptosis of prostatic cells and organ regression, and is a cornerstone of prostate cancer and benign prostatic hypertrophy treatment. For several decades, androgen deprivation has been used as an adjuvant to external beam radiotherapy, however, emerging data suggests that the low rates of epithelial proliferation in the castrated prostate imparts radio-resistance. As proliferating cells exhibit increased sensitivity to radiation, we hypothesized that short bursts of synchronized epithelial proliferation, which can be achieved by exogeneous testosterone supplementation prior to targeted high-dose radiation, would maximize sustained prostate ablation, while minimizing damage to surrounding tissues. To test this hypothesis, we designed a novel computed-tomography (CT)-guided stereotactic prostate radiation therapy (CT-SPRT) technique to deliver a single high-dose 25 Gy fraction of X-ray radiation. Sustained prostatic cell ablation was assessed post CT-SPRT by measuring prostate weight, epithelial cell number, and relative contributions of luminal and basal epithelial populations in control and testosterone-pretreated glands. CT-SPRT was safely delivered with no observed damage to surrounding rectal and bladder tissues. Importantly, castrated mice that received a pulse of testosterone to induce synchronous cell proliferation prior to CT-SPRT exhibited significant sustained gland ablation compared to control mice. These results provide new insights in stereotactic radiotherapy sensitivity to maximize prostatic cell ablation and improve our understanding of prostate gland regeneration that can potentially lead to improved non-invasive therapies for benign prostatic hypertrophy and prostate cancer.
Collapse
Affiliation(s)
- Ali H Zahalka
- Department of Urology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA. .,Department of Cell Biology, Albert Einstein College of Medicine, 1301 Morris Park Ave, Bronx, NY, 10461, USA. .,Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1272, New York, NY, 10029, USA.
| | - N Patrik Brodin
- Department of Radiation Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, 1301 Morris Park Ave, Bronx, NY, 10461, USA.,Ruth L. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xizhe Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kara L Watts
- Department of Urology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Sandra Pinho
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Chandan Guha
- Department of Radiation Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Paul S Frenette
- Department of Cell Biology, Albert Einstein College of Medicine, 1301 Morris Park Ave, Bronx, NY, 10461, USA. .,Ruth L. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA. .,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
39
|
Yao J, Liu Y, Liang X, Shao J, Zhang Y, Yang J, Zheng M. Neuroendocrine Carcinoma as an Independent Prognostic Factor for Patients With Prostate Cancer: A Population-Based Study. Front Endocrinol (Lausanne) 2021; 12:778758. [PMID: 34956090 PMCID: PMC8692830 DOI: 10.3389/fendo.2021.778758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/17/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Neuroendocrine carcinoma (NEC) is a rare and highly malignant variation of prostate adenocarcinoma. We aimed to investigate the prognostic value of NEC in prostate cancer. METHODS A total of 530440 patients of prostate cancer, including neuroendocrine prostate cancer (NEPC) and adenocarcinoma from 2004 to 2018 were obtained from the national Surveillance, Epidemiology, and End Results (SEER) database. Propensity score matching (PSM), multivariable Cox proportional hazard model, Kaplan-Meier method and subgroup analysis were performed in our study. RESULTS NEPC patients were inclined to be older at diagnosis (Median age, 69(61-77) vs. 65(59-72), P< 0.001) and had higher rates of muscle invasive disease (30.9% vs. 9.2%, P < 0.001), lymph node metastasis (32.2% vs. 2.2%, P < 0.001), and distal metastasis (45.7% vs. 3.6%, P < 0.001) compared with prostate adenocarcinoma patients. However, the proportion of NEPC patients with PSA levels higher than 4.0 ng/mL was significantly less than adenocarcinoma patients (47.3% vs. 72.9%, P<0.001). NEPC patients had a lower rate of receiving surgery treatment (28.8% vs. 43.9%, P<0.001), but they had an obviously higher rate of receiving chemotherapy (57.9% vs. 1.0%, P<0.001). A Cox regression analysis demonstrated that the NEPC patients faced a remarkably worse OS (HR = 2.78, 95% CI = 2.34-3.31, P < 0.001) and CSS (HR = 3.07, 95% CI = 2.55-3.71, P < 0.001) compared with adenocarcinoma patients after PSM. Subgroup analyses further suggested that NEPC patients obtained significantly poorer prognosis across nearly all subgroups. CONCLUSION The prognosis of NEPC was worse than that of adenocarcinoma among patients with prostate cancer. The histological subtype of NEC is an independent prognostic factor for patients with prostate cancer.
Collapse
|
40
|
Arriaga JM, Panja S, Alshalalfa M, Zhao J, Zou M, Giacobbe A, Madubata CJ, Kim JY, Rodriguez A, Coleman I, Virk RK, Hibshoosh H, Ertunc O, Ozbek B, Fountain J, Jeffrey Karnes R, Luo J, Antonarakis ES, Nelson PS, Feng FY, Rubin MA, De Marzo AM, Rabadan R, Sims PA, Mitrofanova A, Abate-Shen C. A MYC and RAS co-activation signature in localized prostate cancer drives bone metastasis and castration resistance. NATURE CANCER 2020; 1:1082-1096. [PMID: 34085047 PMCID: PMC8171279 DOI: 10.1038/s43018-020-00125-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
Understanding the intricacies of lethal prostate cancer poses specific challenges due to difficulties in accurate modeling of metastasis in vivo. Here we show that NPK EYFP mice (for Nkx3.1 CreERT2/+ ; Pten flox/flox ; Kras LSL-G12D/+ ; R26R-CAG-LSL-EYFP/+) develop prostate cancer with a high penetrance of metastasis to bone, thereby enabling detection and tracking of bone metastasis in vivo and ex vivo. Transcriptomic and whole-exome analyses of bone metastasis from these mice revealed distinct molecular profiles conserved between human and mouse and specific patterns of subclonal branching from the primary tumor. Integrating bulk and single-cell transcriptomic data from mouse and human datasets with functional studies in vivo unravels a unique MYC/RAS co-activation signature associated with prostate cancer metastasis. Finally, we identify a gene signature with prognostic value for time to metastasis and predictive of treatment response in human patients undergoing androgen receptor therapy across clinical cohorts, thus uncovering conserved mechanisms of metastasis with potential translational significance.
Collapse
Affiliation(s)
- Juan M Arriaga
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY, USA
| | - Sukanya Panja
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Mohammed Alshalalfa
- Department of Radiation Oncology, University of California at San Francisco, San Francisco, CA, USA
| | - Junfei Zhao
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Min Zou
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY, USA
- Arvinas, New Haven, CT, USA
| | - Arianna Giacobbe
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY, USA
| | - Chioma J Madubata
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Jaime Yeji Kim
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY, USA
| | - Antonio Rodriguez
- Department for BioMedical Research, University of Bern and Inselspital, Bern, Switzerland
- Institute of Pathology, University of Bern and Inselspital, Bern, Switzerland
| | - Ilsa Coleman
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Renu K Virk
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Hanina Hibshoosh
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Onur Ertunc
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Suleyman Demirel University, Training and Research Hospital, Isparta, Turkey
| | - Büşra Ozbek
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julia Fountain
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
| | | | - Jun Luo
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emmanuel S Antonarakis
- Department of Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of California at San Francisco, San Francisco, CA, USA
- Department of Urology, University of California at San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Mark A Rubin
- Department for BioMedical Research, University of Bern and Inselspital, Bern, Switzerland
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raul Rabadan
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Antonina Mitrofanova
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers Biomedical and Health Sciences, Newark, NJ, USA.
| | - Cory Abate-Shen
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
41
|
Cellular and Molecular Progression of Prostate Cancer: Models for Basic and Preclinical Research. Cancers (Basel) 2020; 12:cancers12092651. [PMID: 32957478 PMCID: PMC7563251 DOI: 10.3390/cancers12092651] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 02/08/2023] Open
Abstract
Simple Summary The molecular progression of prostate cancer is complex and elusive. Biological research relies heavily on in vitro and in vivo models that can be used to examine gene functions and responses to the external agents in laboratory and preclinical settings. Over the years, several models have been developed and found to be very helpful in understanding the biology of prostate cancer. Here we describe these models in the context of available information on the cellular and molecular progression of prostate cancer to suggest their potential utility in basic and preclinical prostate cancer research. The information discussed herein should serve as a hands-on resource for scholars engaged in prostate cancer research or to those who are making a transition to explore the complex biology of prostate cancer. Abstract We have witnessed noteworthy progress in our understanding of prostate cancer over the past decades. This basic knowledge has been translated into efficient diagnostic and treatment approaches leading to the improvement in patient survival. However, the molecular pathogenesis of prostate cancer appears to be complex, and histological findings often do not provide an accurate assessment of disease aggressiveness and future course. Moreover, we also witness tremendous racial disparity in prostate cancer incidence and clinical outcomes necessitating a deeper understanding of molecular and mechanistic bases of prostate cancer. Biological research heavily relies on model systems that can be easily manipulated and tested under a controlled experimental environment. Over the years, several cancer cell lines have been developed representing diverse molecular subtypes of prostate cancer. In addition, several animal models have been developed to demonstrate the etiological molecular basis of the prostate cancer. In recent years, patient-derived xenograft and 3-D culture models have also been created and utilized in preclinical research. This review is an attempt to succinctly discuss existing information on the cellular and molecular progression of prostate cancer. We also discuss available model systems and their tested and potential utility in basic and preclinical prostate cancer research.
Collapse
|
42
|
Crowley L, Cambuli F, Aparicio L, Shibata M, Robinson BD, Xuan S, Li W, Hibshoosh H, Loda M, Rabadan R, Shen MM. A single-cell atlas of the mouse and human prostate reveals heterogeneity and conservation of epithelial progenitors. eLife 2020; 9:e59465. [PMID: 32915138 PMCID: PMC7529463 DOI: 10.7554/elife.59465] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/10/2020] [Indexed: 01/06/2023] Open
Abstract
Understanding the cellular constituents of the prostate is essential for identifying the cell of origin for prostate adenocarcinoma. Here, we describe a comprehensive single-cell atlas of the adult mouse prostate epithelium, which displays extensive heterogeneity. We observe distal lobe-specific luminal epithelial populations (LumA, LumD, LumL, and LumV), a proximally enriched luminal population (LumP) that is not lobe-specific, and a periurethral population (PrU) that shares both basal and luminal features. Functional analyses suggest that LumP and PrU cells have multipotent progenitor activity in organoid formation and tissue reconstitution assays. Furthermore, we show that mouse distal and proximal luminal cells are most similar to human acinar and ductal populations, that a PrU-like population is conserved between species, and that the mouse lateral prostate is most similar to the human peripheral zone. Our findings elucidate new prostate epithelial progenitors, and help resolve long-standing questions about anatomical relationships between the mouse and human prostate.
Collapse
Affiliation(s)
- Laura Crowley
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Francesco Cambuli
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Luis Aparicio
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Biomedical Informatics, Columbia University Irving Medical CenterNew YorkUnited States
| | - Maho Shibata
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell UniversityNew YorkUnited States
| | - Shouhong Xuan
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Weiping Li
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia University Irving Medical CenterNew YorkUnited States
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell UniversityNew YorkUnited States
| | - Raul Rabadan
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Biomedical Informatics, Columbia University Irving Medical CenterNew YorkUnited States
| | - Michael M Shen
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| |
Collapse
|
43
|
Tremblay M, Viala S, Shafer ME, Graham-Paquin AL, Liu C, Bouchard M. Regulation of stem/progenitor cell maintenance by BMP5 in prostate homeostasis and cancer initiation. eLife 2020; 9:54542. [PMID: 32894216 PMCID: PMC7525654 DOI: 10.7554/elife.54542] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 09/06/2020] [Indexed: 12/25/2022] Open
Abstract
Tissue homeostasis relies on the fine regulation between stem and progenitor cell maintenance and lineage commitment. In the adult prostate, stem cells have been identified in both basal and luminal cell compartments. However, basal stem/progenitor cell homeostasis is still poorly understood. We show that basal stem/progenitor cell maintenance is regulated by a balance between BMP5 self-renewal signal and GATA3 dampening activity. Deleting Gata3 enhances adult prostate stem/progenitor cells self-renewal capacity in both organoid and allograft assays. This phenotype results from a local increase in BMP5 activity in basal cells as shown by the impaired self-renewal capacity of Bmp5-deficient stem/progenitor cells. Strikingly, Bmp5 gene inactivation or BMP signaling inhibition with a small molecule inhibitor are also sufficient to delay prostate and skin cancer initiation of Pten-deficient mice. Together, these results establish BMP5 as a key regulator of basal prostate stem cell homeostasis and identifies a potential therapeutic approach against Pten-deficient cancers.
Collapse
Affiliation(s)
- Mathieu Tremblay
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada
| | - Sophie Viala
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada
| | - Maxwell Er Shafer
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada
| | - Adda-Lee Graham-Paquin
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada
| | - Chloe Liu
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada
| | - Maxime Bouchard
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada
| |
Collapse
|
44
|
Guo W, Li L, He J, Liu Z, Han M, Li F, Xia X, Zhang X, Zhu Y, Wei Y, Li Y, Aji R, Dai H, Wei H, Li C, Chen Y, Chen L, Gao D. Single-cell transcriptomics identifies a distinct luminal progenitor cell type in distal prostate invagination tips. Nat Genet 2020; 52:908-918. [PMID: 32807988 PMCID: PMC8383310 DOI: 10.1038/s41588-020-0642-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/07/2020] [Indexed: 11/09/2022]
Abstract
The identification of prostate stem/progenitor cells and characterization of the prostate epithelial cell lineage hierarchy are critical for understanding prostate cancer initiation. Here, we characterized 35,129 cells from mouse prostates, and identified a unique luminal cell type (termed type C luminal cell (Luminal-C)) marked by Tacstd2, Ck4 and Psca expression. Luminal-C cells located at the distal prostate invagination tips (termed Dist-Luminal-C) exhibited greater capacity for organoid formation in vitro and prostate epithelial duct regeneration in vivo. Lineage tracing of Luminal-C cells indicated that Dist-Luminal-C cells reconstituted distal prostate luminal lineages through self-renewal and differentiation. Deletion of Pten in Dist-Luminal-C cells resulted in prostatic intraepithelial neoplasia. We further characterized 11,374 human prostate cells and confirmed the existence of h-Luminal-C cells. Our study provides insights into the prostate lineage hierarchy, identifies Dist-Luminal-C cells as the luminal progenitor cell population in invagination tips and suggests one of the potential cellular origins of prostate cancer.
Collapse
Affiliation(s)
- Wangxin Guo
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lin Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Juan He
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhuang Liu
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ming Han
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinyi Xia
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyu Zhang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, China
| | - Yunguang Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rebiguli Aji
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Dai
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Wei
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Chunfeng Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY, USA.
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China.
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
45
|
Tiwari R, Manzar N, Ateeq B. Dynamics of Cellular Plasticity in Prostate Cancer Progression. Front Mol Biosci 2020; 7:130. [PMID: 32754615 PMCID: PMC7365877 DOI: 10.3389/fmolb.2020.00130] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
Despite the current advances in the treatment for prostate cancer, the patients often develop resistance to the conventional therapeutic interventions. Therapy-induced drug resistance and tumor progression have been associated with cellular plasticity acquired due to reprogramming at the molecular and phenotypic levels. The plasticity of the tumor cells is mainly governed by two factors: cell-intrinsic and cell-extrinsic. The cell-intrinsic factors involve alteration in the genetic or epigenetic regulators, while cell-extrinsic factors include microenvironmental cues and drug-induced selective pressure. Epithelial-mesenchymal transition (EMT) and stemness are two important hallmarks that dictate cellular plasticity in multiple cancer types including prostate. Emerging evidence has also pinpointed the role of tumor cell plasticity in driving anti-androgen induced neuroendocrine prostate cancer (NEPC), a lethal and therapy-resistant subtype. In this review, we discuss the role of cellular plasticity manifested due to genetic, epigenetic alterations and cues from the tumor microenvironment, and their role in driving therapy resistant prostate cancer.
Collapse
Affiliation(s)
| | | | - Bushra Ateeq
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
46
|
Pascal LE, Rigatti LH, Ai J, Zhang A, Zhou J, Nelson JB, Wang Z. EAF2 loss induces prostatic intraepithelial neoplasia from luminal epithelial cells in mice. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2020; 8:18-27. [PMID: 32211450 PMCID: PMC7076293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/18/2020] [Indexed: 06/10/2023]
Abstract
Defining the cell of origin for prostatic carcinogenesis is fundamentally important for understanding the mechanisms leading to prostate cancer. Lineage tracing studies have demonstrated that luminal epithelial cells are capable of self-replication in multiple organs, including the adult murine prostate, and cell of prostate cancer origin studies have shown that while both the luminal and basal murine prostate epithelial cells are capable of neoplastic transformation, luminal cells are more efficient as the origin of prostate cancer. ELL-associated factor 2 (EAF2) is an androgen responsive tumor suppressive protein expressed by prostate luminal epithelial cells that is frequently down-regulated in primary prostate tumors. EAF2 knockdown induces prostate cancer cell proliferation and invasion in vitro and mice with Eaf2 deficiency develop epithelial hyperplasia and murine prostatic intraepithelial neoplasia (mPIN) lesions. Here, we utilized an Eaf2 knockout, PSA-CreERT2 transgenic model crossed with a fluorescent reporter line to show that Eaf2 deficiency induces mPIN lesions derived from the luminal cell lineage. These results suggest that PIN lesions in the Eaf2 knockout mouse were derived from prostate luminal epithelial cells, further suggesting that the prostatic luminal epithelial cell is the major origin of prostate carcinogenesis.
Collapse
Affiliation(s)
- Laura E Pascal
- Department of Urology, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| | - Lora H Rigatti
- Division of Laboratory Animal Resources, University of Pittsburgh School of MedicinePittsburgh, PA 15216, USA
| | - Junkui Ai
- Department of Urology, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| | - Aiyuan Zhang
- Department of Urology, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| | - Jianhua Zhou
- Department of Urology, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| | - Joel B Nelson
- Department of Urology, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| | - Zhou Wang
- Department of Urology, University of Pittsburgh School of MedicinePittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of MedicinePittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| |
Collapse
|
47
|
TIP5 primes prostate luminal cells for the oncogenic transformation mediated by PTEN-loss. Proc Natl Acad Sci U S A 2020; 117:3637-3647. [PMID: 32024754 PMCID: PMC7035629 DOI: 10.1073/pnas.1911673117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The cell of origin and the temporal order of oncogenic events in tumors play important roles for disease state. This is of particular interest for PCa due to its highly variable clinical outcome. However, these features are difficult to analyze in tumors. We established an in vitro murine PCa organoid model taking into account the cell of origin and the temporal order of events. We found that TIP5 primes luminal prostate cells for Pten-loss mediated oncogenic transformation whereas it is dispensable once the transformation is established. Cross-species transcriptomic analyses revealed a PTEN-loss gene signature that identified a set of aggressive tumors with PTEN-del, or low PTEN expression, and high-TIP5 expression. This paper provides a powerful tool to elucidate PCa mechanisms. Prostate cancer (PCa) is the second leading cause of cancer death in men. Its clinical and molecular heterogeneities and the lack of in vitro models outline the complexity of PCa in the clinical and research settings. We established an in vitro mouse PCa model based on organoid technology that takes into account the cell of origin and the order of events. Primary PCa with deletion of the tumor suppressor gene PTEN (PTEN-del) can be modeled through Pten-down-regulation in mouse organoids. We used this system to elucidate the contribution of TIP5 in PCa initiation, a chromatin regulator that is implicated in aggressive PCa. High TIP5 expression correlates with primary PTEN-del PCa and this combination strongly associates with reduced prostate-specific antigen (PSA) recurrence-free survival. TIP5 is critical for the initiation of PCa of luminal origin mediated by Pten-loss whereas it is dispensable once Pten-loss mediated transformation is established. Cross-species analyses revealed a PTEN gene signature that identified a group of aggressive primary PCas characterized by PTEN-del, high-TIP5 expression, and a TIP5-regulated gene expression profile. The results highlight the modeling of PCa with organoids as a powerful tool to elucidate the role of genetic alterations found in recent studies in their time orders and cells of origin, thereby providing further optimization for tumor stratification to improve the clinical management of PCa.
Collapse
|
48
|
Saranyutanon S, Srivastava SK, Pai S, Singh S, Singh AP. Therapies Targeted to Androgen Receptor Signaling Axis in Prostate Cancer: Progress, Challenges, and Hope. Cancers (Basel) 2019; 12:cancers12010051. [PMID: 31877956 PMCID: PMC7016833 DOI: 10.3390/cancers12010051] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the mostly commonly diagnosed non-cutaneous malignancy and the second leading cause of cancer-related death affecting men in the United States. Moreover, it disproportionately affects the men of African origin, who exhibit significantly greater incidence and mortality as compared to the men of European origin. Since androgens play an important role in the growth of normal prostate and prostate tumors, targeting of androgen signaling has remained a mainstay for the treatment of aggressive prostate cancer. Over the years, multiple approaches have been evaluated to effectively target the androgen signaling pathway that include direct targeting of the androgens, androgen receptor (AR), AR co-regulators or other alternate mechanisms that impact the outcome of androgen signaling. Several of these approaches are currently in clinical practice, while some are still pending further development and clinical evaluation. This remarkable progress has resulted from extensive laboratory, pre-clinical and clinical efforts, and mechanistic learnings from the therapeutic success and failures. In this review, we describe the importance of androgen signaling in prostate cancer biology and advances made over the years to effectively target this signaling pathway. We also discuss emerging data on the resistance pathways associated with the failure of various androgen signaling- targeted therapies and potential of this knowledge for translation into future therapies for prostate cancer.
Collapse
Affiliation(s)
- Sirin Saranyutanon
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; (S.S.)
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; (S.S.)
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Correspondence: (S.K.S.); (A.P.S.); Tel.: +1-251-445-9874 (S.K.S.); +1-251-445-9843 (A.P.S.)
| | - Sachin Pai
- Department of Medical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA;
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; (S.S.)
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; (S.S.)
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
- Correspondence: (S.K.S.); (A.P.S.); Tel.: +1-251-445-9874 (S.K.S.); +1-251-445-9843 (A.P.S.)
| |
Collapse
|
49
|
O'Reilly D, Johnson P, Buchanan PJ. Hypoxia induced cancer stem cell enrichment promotes resistance to androgen deprivation therapy in prostate cancer. Steroids 2019; 152:108497. [PMID: 31521707 DOI: 10.1016/j.steroids.2019.108497] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023]
Abstract
Androgen deprivation therapy (ADT) is the main treatment to prolong survival in advance stage prostate cancer (PCa) but associated resistance leads to the development of terminal castrate resistant PCa (CRPC). Current research demonstrates that prostate cancer stem cells (PCSC) play a critical role in the development of treatment resistance and subsequent disease progression. Despite uncertainty surrounding the origin of these cells, studies clearly show they are associated with poorer outcomes and that ADT significantly enhances their numbers. Here in we highlight how activation of HIF signalling, in response to hypoxic conditions within the tumour microenvironment, results in the expression of genes associated with stemness and EMT promoting PCSC emergence which ultimately drives tumour relapse to CRPC. Hypoxic conditions are not only enhanced by ADT but the associated decrease in AR activation also promotes PI3K/AKT signalling which actively enhances HIF and its effects on PCSC's. Furthermore, emerging evidence now indicates that HIF-2α, rather than the commonly considered HIF-1α, is the main family member that drives PCSC emergence. Taken together this clearly identifies HIF and associated pathways as key targets for new therapeutic strategies that could potentially prevent or slow PCSC promoted resistance to ADT, thus holding potential to prolong patient survival.
Collapse
Affiliation(s)
- Debbie O'Reilly
- School of Nursing & Human Sciences, Dublin City University, Dublin, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Patricia Johnson
- School of Nursing & Human Sciences, Dublin City University, Dublin, Ireland
| | - Paul J Buchanan
- School of Nursing & Human Sciences, Dublin City University, Dublin, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin, Ireland.
| |
Collapse
|
50
|
Yu C, Hu K, Nguyen D, Wang ZA. From genomics to functions: preclinical mouse models for understanding oncogenic pathways in prostate cancer. Am J Cancer Res 2019; 9:2079-2102. [PMID: 31720076 PMCID: PMC6834478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/10/2019] [Indexed: 06/10/2023] Open
Abstract
Next-generation sequencing has revealed numerous genomic alterations that induce aberrant signaling activities in prostate cancer (PCa). Among them are pathways affecting multiple cancer types, including the PI3K/AKT/mTOR, p53, Rb, Ras/Raf/MAPK, Myc, FGF, and Wnt signaling pathways, as well as ones that are prominent in PCa, including alterations in genes of AR signaling, the ETS family, NKX3.1, and SPOP. Cross talk among the oncogenic pathways can confer PCa resistance to therapy, particularly in advanced tumors, which are castration-resistant or show neuroendocrine features. Various experimental models, such as cancer cell lines, animal models, and patient-derived xenografts and organoids have been utilized to dissect PCa progression mechanisms. Here, we review the current preclinical mouse models for studying the most commonly altered pathways in PCa, with an emphasis on their interplays. We highlight the power of genetically engineered mouse models (GEMMs) in translating genomic discoveries into understanding of the functions of these oncogenic events in vivo. Developing and analyzing PCa mouse models will undoubtedly continue to offer new insights into tumor biology and guide novel rationalized therapy.
Collapse
Affiliation(s)
- Chuan Yu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Kevin Hu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Daniel Nguyen
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Zhu A Wang
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| |
Collapse
|