1
|
Su H, Huang L, Zhou J, Yang G. Prostate cancer stem cells and their targeted therapies. Front Cell Dev Biol 2024; 12:1410102. [PMID: 39175878 PMCID: PMC11338935 DOI: 10.3389/fcell.2024.1410102] [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: 03/31/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
Prostate cancer (PCa) is the most common malignancy among men worldwide. Through androgen receptor signaling inhibitor (ARSI) treatment, patients eventually succumb to castration-resistant prostate cancer (CRPC). For this, the prostate cancer stem cells (PCSCs), as a minor population of tumor cells that can promote tumor relapse, ARSI resistance, and disease progression, are gaining attention. Therefore, specific therapy targeting PCSCs has momentum. This study reviewed the identification and characterization of PCSCs and PCSC-based putative biomarkers and summarized their mechanisms of action. We further discussed clinical trials of novel therapeutic interventions focused on PCSC-related pathways, the PCSC microenvironment, cutting-edge miRNA therapy, and immunotherapy approaches from a mechanistic standpoint. This review provides updated insights into PCSC plasticity, identifying new PCSC biomarkers and optimized treatments for patients with advanced PCa.
Collapse
Affiliation(s)
- Huilan Su
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liqun Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianjun Zhou
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guosheng Yang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Horton C, Liu Y, Wang J, Green J, Tsyporin J, Chen B, Wang ZA. Modulation of the canonical Wnt activity by androgen signaling in prostate epithelial basal stem cells. Stem Cell Reports 2023; 18:1355-1370. [PMID: 37172587 PMCID: PMC10277819 DOI: 10.1016/j.stemcr.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
Both the canonical Wnt and androgen receptor (AR) signaling pathways are important for prostate organogenesis and homeostasis. How they crosstalk to regulate prostate stem cell behaviors remains unclear. Here, we show in lineage-tracing mouse models that although Wnt is essential for basal stem cell multipotency, ectopic Wnt activity promotes basal cell over-proliferation and squamous phenotypes, which are counteracted by elevated levels of androgen. In prostate basal cell organoids, dihydrotestosterone (DHT) antagonizes R-spondin-stimulated growth in a concentration-dependent manner. DHT down-regulates the expressions of a Wnt reporter and target genes, and RNA sequencing (RNA-seq) analyses identify Wnt signaling as a key altered pathway. Mechanistically, DHT enhances AR and β-catenin protein binding, and CUT&RUN analyses reveal that ectopic AR sequesters β-catenin away from its Wnt-related cistrome. Our results suggest that an intermediate level of Wnt activity in prostate basal stem cells, achieved via AR-β-catenin interaction, is essential for normal prostate homeostasis.
Collapse
Affiliation(s)
- Corrigan Horton
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - 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
| | - Jonathan Green
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jeremiah Tsyporin
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Bin Chen
- 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
|
3
|
Tu WL, Chih YC, Shih YT, Yu YR, You LR, Chen CM. Context-specific roles of diphthamide deficiency in hepatocellular carcinogenesis. J Pathol 2022; 258:149-163. [PMID: 35781884 DOI: 10.1002/path.5986] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/13/2022] [Accepted: 07/01/2022] [Indexed: 11/07/2022]
Abstract
Diphthamide biosynthesis protein 1 (DPH1) is biochemically involved in the first step of diphthamide biosynthesis, a post-translational modification of eukaryotic elongation factor 2 (EEF2). Earlier studies showed that DPH1, also known as ovarian cancer-associated gene 1 (OVCA1), is involved in ovarian carcinogenesis. However, the role of DPH1 in hepatocellular carcinoma (HCC) remains unclear. To investigate the impact of DPH1 in hepatocellular carcinogenesis, we have performed data mining from The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) dataset. We found that reduced DPH1 levels were associated with advanced stages and poor survival of patients with HCC. Also, we generated hepatocyte-specific Dph1 deficient mice and showed that diphthamide deficient EEF2 resulted in a reduced translation elongation rate in the hepatocytes and let to mild liver damage with fatty accumulation. After N-diethylnitrosamine (DEN) -induced acute liver injury, p53-mediated pericentral hepatocyte death was increased, and compensatory proliferation was reduced in Dph1-deficient mice. Consistent with these effects, Dph1 deficiency decreased the incidence of DEN-induced pericentral-derived HCC and revealed a protective effect against p53 loss. In contrast, Dph1 deficiency combined with Trp53- or Trp53/Pten-deficient hepatocytes led to increased tumor loads associated with KRT19 (K19)-positive periportal-like cell expansion in mice. Further gene set enrichment analysis also revealed that HCC patients with lower levels of DPH1 and TP53 expression had enriched gene-sets related to the cell cycle and K19-upregulated HCC. Additionally, liver tumor organoids obtained from 6-month-old Pten/Trp53/Dph1-triple-mutant mice had a higher frequency of organoid re-initiation cells and higher proliferative index compared with those of the Pten/Trp53-double-mutant. Pten/Trp53/Dph1-triple-mutant liver tumor organoids showed expression of genes associated with stem/progenitor phenotypes, including Krt19 and Prominin-1 (Cd133) progenitor markers, combined with low hepatocyte-expressed fibrinogen genes. These findings indicate that diphthamide deficiency differentially regulates hepatocellular carcinogenesis, which inhibits pericentral hepatocytes-derived tumor and promotes periportal progenitors-associated liver tumors. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Wei-Ling Tu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming, Chiao Tung University, Taipei, Taiwan
| | - Yu-Chan Chih
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming, Chiao Tung University, Taipei, Taiwan
| | - Ya-Tung Shih
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming, Chiao Tung University, Taipei, Taiwan
| | - Yi-Ru Yu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming, Chiao Tung University, Taipei, Taiwan
| | - Li-Ru You
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Ming Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming, Chiao Tung University, Taipei, Taiwan.,Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
4
|
Xie J, Wang M, Xu S, Huang Z, Grant PW. The Unsupervised Feature Selection Algorithms Based on Standard Deviation and Cosine Similarity for Genomic Data Analysis. Front Genet 2021; 12:684100. [PMID: 34054930 PMCID: PMC8155687 DOI: 10.3389/fgene.2021.684100] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/16/2021] [Indexed: 01/04/2023] Open
Abstract
To tackle the challenges in genomic data analysis caused by their tens of thousands of dimensions while having a small number of examples and unbalanced examples between classes, the technique of unsupervised feature selection based on standard deviation and cosine similarity is proposed in this paper. We refer to this idea as SCFS (Standard deviation and Cosine similarity based Feature Selection). It defines the discernibility and independence of a feature to value its distinguishable capability between classes and its redundancy to other features, respectively. A 2-dimensional space is constructed using discernibility as x-axis and independence as y-axis to represent all features where the upper right corner features have both comparatively high discernibility and independence. The importance of a feature is defined as the product of its discernibility and its independence (i.e., the area of the rectangular enclosed by the feature’s coordinate lines and axes). The upper right corner features are by far the most important, comprising the optimal feature subset. Based on different definitions of independence using cosine similarity, there are three feature selection algorithms derived from SCFS. These are SCEFS (Standard deviation and Exponent Cosine similarity based Feature Selection), SCRFS (Standard deviation and Reciprocal Cosine similarity based Feature Selection) and SCAFS (Standard deviation and Anti-Cosine similarity based Feature Selection), respectively. The KNN and SVM classifiers are built based on the optimal feature subsets detected by these feature selection algorithms, respectively. The experimental results on 18 genomic datasets of cancers demonstrate that the proposed unsupervised feature selection algorithms SCEFS, SCRFS and SCAFS can detect the stable biomarkers with strong classification capability. This shows that the idea proposed in this paper is powerful. The functional analysis of these biomarkers show that the occurrence of the cancer is closely related to the biomarker gene regulation level. This fact will benefit cancer pathology research, drug development, early diagnosis, treatment and prevention.
Collapse
Affiliation(s)
- Juanying Xie
- School of Computer Science, Shaanxi Normal University, Xi'an, China
| | - Mingzhao Wang
- School of Computer Science, Shaanxi Normal University, Xi'an, China.,College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Shengquan Xu
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhao Huang
- School of Computer Science, Shaanxi Normal University, Xi'an, China
| | - Philip W Grant
- Department of Computer Science, Swansea University, Swansea, United Kingdom
| |
Collapse
|
5
|
Kwon OJ, Choi JM, Zhang L, Jia D, Li Z, Zhang Y, Jung SY, Creighton CJ, Xin L. The Sca-1 + and Sca-1 - mouse prostatic luminal cell lineages are independently sustained. Stem Cells 2020; 38:1479-1491. [PMID: 32627901 DOI: 10.1002/stem.3253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
The phenotypic and functional heterogeneity of the mouse prostate epithelial cell lineages remains incompletely characterized. We show that the Sca-1+ luminal cells at the mouse proximal prostate express Sox2. These cells are replicative quiescent, castration resistant, and do not possess secretory function. We use the Probasin-CreERT2 and Sox2-CreERT2 models in concert with a fluorescent reporter line to label the Sca-1- and Sca-1+ luminal cells, respectively. By a lineage tracing approach, we show that the two luminal cell populations are independently sustained. Sox2 is dispensable for the maintenance of the Sca-1+ luminal cells but is essential for their facultative bipotent differentiation capacity. The Sca-1+ luminal cells share molecular features with the human TACSTD2+ luminal cells. This study corroborates the heterogeneity of the mouse prostate luminal cell lineage and shows that the adult mouse prostate luminal cell lineage is maintained by distinct cellular entities rather than a single progenitor population.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Jong Min Choi
- Department of Chemistry and Biochemistry, Baylor College of Medicine, Houston, Texas, USA
| | - Li Zhang
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Deyong Jia
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Zhouyihan Li
- Department of Chemistry and Biochemistry, University of Washington, Seattle, Washington, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Sung Yun Jung
- Department of Chemistry and Biochemistry, Baylor College of Medicine, Houston, Texas, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Li Xin
- Department of Urology, University of Washington, Seattle, Washington, USA.,Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
Patel R, Brzezinska EA, Repiscak P, Ahmad I, Mui E, Gao M, Blomme A, Harle V, Tan EH, Malviya G, Mrowinska A, Loveridge CJ, Rushworth LK, Edwards J, Ntala C, Nixon C, Hedley A, Mackay G, Tardito S, Sansom OJ, Leung HY. Activation of β-Catenin Cooperates with Loss of Pten to Drive AR-Independent Castration-Resistant Prostate Cancer. Cancer Res 2020; 80:576-590. [PMID: 31719098 DOI: 10.1158/0008-5472.can-19-1684] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/04/2019] [Accepted: 11/08/2019] [Indexed: 11/16/2022]
Abstract
Inhibition of the androgen receptor (AR) is the main strategy to treat advanced prostate cancers. AR-independent treatment-resistant prostate cancer is a major unresolved clinical problem. Patients with prostate cancer with alterations in canonical WNT pathway genes, which lead to β-catenin activation, are refractory to AR-targeted therapies. Here, using clinically relevant murine prostate cancer models, we investigated the significance of β-catenin activation in prostate cancer progression and treatment resistance. β-Catenin activation, independent of the cell of origin, cooperated with Pten loss to drive AR-independent castration-resistant prostate cancer. Prostate tumors with β-catenin activation relied on the noncanonical WNT ligand WNT5a for sustained growth. WNT5a repressed AR expression and maintained the expression of c-Myc, an oncogenic effector of β-catenin activation, by mediating nuclear localization of NFκBp65 and β-catenin. Overall, WNT/β-catenin and AR signaling are reciprocally inhibited. Therefore, inhibiting WNT/β-catenin signaling by limiting WNT secretion in concert with AR inhibition may be useful for treating prostate cancers with alterations in WNT pathway genes. SIGNIFICANCE: Targeting of both AR and WNT/β-catenin signaling may be required to treat prostate cancers that exhibit alterations of the WNT pathway.
Collapse
MESH Headings
- Androgen Receptor Antagonists/pharmacology
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice
- PTEN Phosphohydrolase/deficiency
- Prognosis
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Survival Rate
- Tumor Cells, Cultured
- Wnt-5a Protein/genetics
- Wnt-5a Protein/metabolism
- Xenograft Model Antitumor Assays
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Rachana Patel
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom.
| | | | - Peter Repiscak
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| | - Imran Ahmad
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| | - Ernest Mui
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| | - Meiling Gao
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Arnaud Blomme
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Victoria Harle
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| | - Ee Hong Tan
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Gaurav Malviya
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Agata Mrowinska
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Carolyn J Loveridge
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| | - Linda K Rushworth
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| | - Joanne Edwards
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| | - Chara Ntala
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Colin Nixon
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Ann Hedley
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Gillian Mackay
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Saverio Tardito
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| | - Hing Y Leung
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom.
- Institute of Cancer Sciences, Glasgow, Scotland, United Kingdom
| |
Collapse
|
7
|
Wei X, Zhang L, Zhou Z, Kwon OJ, Zhang Y, Nguyen H, Dumpit R, True L, Nelson P, Dong B, Xue W, Birchmeier W, Taketo MM, Xu F, Creighton CJ, Ittmann MM, Xin L. Spatially Restricted Stromal Wnt Signaling Restrains Prostate Epithelial Progenitor Growth through Direct and Indirect Mechanisms. Cell Stem Cell 2019; 24:753-768.e6. [PMID: 30982770 DOI: 10.1016/j.stem.2019.03.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/11/2018] [Accepted: 03/10/2019] [Indexed: 12/31/2022]
Abstract
Cell-autonomous Wnt signaling has well-characterized functions in controlling stem cell activity, including in the prostate. While niche cells secrete Wnt ligands, the effects of Wnt signaling in niche cells per se are less understood. Here, we show that stromal cells in the proximal prostatic duct near the urethra, a mouse prostate stem cell niche, not only produce multiple Wnt ligands but also exhibit strong Wnt/β-catenin activity. The non-canonical Wnt ligand Wnt5a, secreted by proximal stromal cells, directly inhibits proliefration of prostate epithelial stem or progenitor cells whereas stromal cell-autonomous canonical Wnt/β-catenin signaling indirectly suppresses prostate stem or progenitor activity via the transforming growth factor β (TGFβ) pathway. Collectively, these pathways restrain the proliferative potential of epithelial cells in the proximal prostatic ducts. Human prostate likewise exhibits spatially restricted distribution of stromal Wnt/β-catenin activity, suggesting a conserved mechanism for tissue patterning. Thus, this study shows how distinct stromal signaling mechanisms within the prostate cooperate to regulate tissue homeostasis.
Collapse
Affiliation(s)
- Xing Wei
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA; Department of Urology, University of Washington, Seattle, WA 98109, USA
| | - Li Zhang
- Department of Urology, University of Washington, Seattle, WA 98109, USA
| | - Zhicheng Zhou
- Department of Urology, University of Washington, Seattle, WA 98109, USA
| | - Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, WA 98109, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hoang Nguyen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center of Stem Cell and Regenerative Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruth Dumpit
- Human Biology Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| | - Lawrence True
- Department of Pathology, University of Washington, Seattle, WA 98109, USA
| | - Peter Nelson
- Human Biology Division, Fred Hutch Cancer Research Center, Seattle, WA 98109, USA
| | - Baijun Dong
- Department of Urology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Xue
- Department of Urology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Walter Birchmeier
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Makoto M Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Feng Xu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael M Ittmann
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX 77030, USA
| | - Li Xin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Urology, University of Washington, Seattle, WA 98109, USA; Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
8
|
Functional Heterogeneity of Mouse Prostate Stromal Cells Revealed by Single-Cell RNA-Seq. iScience 2019; 13:328-338. [PMID: 30878879 PMCID: PMC6423355 DOI: 10.1016/j.isci.2019.02.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/23/2019] [Accepted: 02/27/2019] [Indexed: 12/26/2022] Open
Abstract
We perform a single-cell RNA sequencing analysis to investigate the phenotypic and functional heterogeneity of the adult mouse prostate stromal cells. Our analysis identifies three major cell populations representing the smooth muscle cells and two types of fibroblast cells enriched by Sca-1 and CD90. The Sca-1+CD90+ fibroblast cells are in direct contact with the epithelial cells and express growth factors and genes associated with cell motility, developmental process, and androgen biosynthesis. This suggests that they may regulate epithelial cell survival and growth. The Sca-1+CD90-/low myofibroblast-like cells highly express genes associated with the extracellular matrix and cytokine-mediated signaling pathways, indicating a role in tissue repair and immune responses. The Sca-1+CD90-/low cells significantly suppress the capacity of the basal cells for bipotent differentiation in the prostate organoid assay. Collectively, we identify the surface markers enabling physical separation of stromal subpopulations and generate the gene expression profiles implying their cellular functions. scRNA-seq reveals three distinct mouse prostate stromal cell populations Sca-1+CD90+ cells produce growth factors mediating developmental process Sca-1+CD90-/low cells express genes mediating immune response and tissue repair Sca-1+CD90-/low cells robustly suppress bipotent differentiation of basal cells
Collapse
|
9
|
Wang CY, Tang MC, Chang WC, Furushima K, Jang CW, Behringer RR, Chen CM. PiggyBac Transposon-Mediated Mutagenesis in Rats Reveals a Crucial Role of Bbx in Growth and Male Fertility. Biol Reprod 2016; 95:51. [PMID: 27465138 PMCID: PMC5394979 DOI: 10.1095/biolreprod.116.141739] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/14/2016] [Indexed: 12/14/2022] Open
Abstract
Bobby sox homolog (Bbx) is an evolutionally conserved gene, but its biological function remains elusive. Here, we characterized defects of Bbx mutant rats that were created by PiggyBac-mediated insertional mutagenesis. Smaller body size and male infertility were the two major phenotypes of homozygous Bbx mutants. Bbx expression profile analysis showed that Bbx was more highly expressed in the testis and pituitary gland than in other organs. Histology and hormonal gene expression analysis of control and Bbx-null pituitary glands showed that loss of Bbx appeared to be dispensable for pituitary histogenesis and the expression of major hormones. BBX was localized in the nuclei of postmeiotic spermatids and Sertoli cells in wild-type testes, but absent in mutant testes. An increased presence of aberrant multinuclear giant cells and apoptotic cells was observed in mutant seminiferous tubules. TUNEL-positive cells costained with CREM (round spermatid marker), but not PLZF (spermatogonia marker), gammaH2Ax (meiotic spermatocyte marker), or GATA4 (Sertoli cell marker). Finally, there were drastically reduced numbers and motility of epididymal sperm from Bbx-null rats. These results suggest that loss of BBX induces apoptosis of postmeiotic spermatids and results in spermiogenesis defects and infertility.
Collapse
Affiliation(s)
- Chieh-Ying Wang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Chu Tang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan Laboratory Animal Center, National Yang-Ming University, Taipei, Taiwan
| | - Wen-Chi Chang
- Laboratory Animal Center, National Yang-Ming University, Taipei, Taiwan
| | - Kenryo Furushima
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Chuan-Wei Jang
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Richard R Behringer
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Chun-Ming Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan Laboratory Animal Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
10
|
β-catenin activation drives thymoma initiation and progression in mice. Oncotarget 2016; 6:13978-93. [PMID: 26101855 PMCID: PMC4546445 DOI: 10.18632/oncotarget.4368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/01/2015] [Indexed: 11/25/2022] Open
Abstract
Thymoma is the most commonly identified cancer in the anterior mediastinum. To date, the causal mechanism that drives thymoma progression is not clear. Here, we generated K5-ΔN64Ctnnb1/ERT2 transgenic mice, which express an N-terminal deletion mutant of β-catenin fused to a mutated ligand-binding domain of estrogen receptor (ERT2) under the control of the bovine cytokeratin 5 (K5) promoter. The transgenic mouse lines named Tg1 and Tg4 were characterized. Forced expression of ΔN64Ctnnb1/ERT2 in the Tg1 and Tg4 mice developed small thymoma lesions in response to tamoxifen treatment. In the absence of tamoxifen, the Tg1 mice exhibited leaky activation of β-catenin, which activated the TOP-Gal transgene and Wnt/β-catenin-targeted genes. As the Tg1 mice aged in the absence of tamoxifen, manifest thymomas were found at 10-12 months. Interestingly, we detected loss of AIRE and increase of p63 in the thymomas of Tg1 mice, similar to that observed in human thymomas. Moreover, the β5t protease subunit, which was reported as a differential marker for human type B3 thymoma, was expressed in the Tg1 thymomas. Thus, the Tg1 mice generated in this study accurately mimic the characteristics of human thymomas and may serve as a model for understanding thymoma pathogenesis.
Collapse
|
11
|
|
12
|
Kwon OJ, Zhang B, Zhang L, Xin L. High fat diet promotes prostatic basal-to-luminal differentiation and accelerates initiation of prostate epithelial hyperplasia originated from basal cells. Stem Cell Res 2016; 16:682-91. [PMID: 27107344 DOI: 10.1016/j.scr.2016.04.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 01/03/2023] Open
Abstract
Recent lineage tracing studies showed that the prostate basal and luminal cells in adult mice are two independent lineages under the physiological condition, but basal cells are capable of generating luminal progenies during bacterial infection-induced prostatitis. Because acute bacterial infection in human prostate tissues is relatively rare, the disease relevance of the bacterial infection-induced basal-to-luminal differentiation is uncertain. Herein we employ a high fat diet-induced sterile prostate inflammation model to determine whether basal-to-luminal differentiation can be induced by inflammation irrespective of the underlying etiologies. A K14-CreER model and a fluorescent report line are utilized to specifically label basal cells with the green fluorescent protein. We show that high fat diet promotes immune cell infiltration into the prostate tissues and basal-to-luminal differentiation. Increased cell proliferation accompanies basal-to-luminal differentiation, suggesting a concurrent regulation of basal cell proliferation and differentiation. This study demonstrates that basal-to-luminal differentiation can be induced by different types of prostate inflammation evolved with distinct etiologies. Finally, high fat diet also accelerates initiation and progression of prostatic intraepithelial neoplasia that are originated from basal cells with loss-of-function of the tumor suppressor Pten. Because prostate cancer originated from basal cells tends to be invasive, our study also provides an alternative explanation for the association between obesity and aggressive prostate cancer.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Boyu Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Li Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Li Xin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States; Dan L. Duncan Cancer Center, Baylor College of Medicine, United States; Department of Pathology and Immunology.
| |
Collapse
|
13
|
Cellular Mechanisms Underlying Intertumoral Heterogeneity. Trends Cancer 2015; 1:15-23. [PMID: 28741558 DOI: 10.1016/j.trecan.2015.07.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/11/2015] [Accepted: 07/10/2015] [Indexed: 12/20/2022]
Abstract
Intertumoral heterogeneity is driven by a combination of intrinsic and extrinsic mechanisms. Intrinsic mechanisms include the genetic/epigenetic mutational profile of cells and the nature of the 'cell of origin'. There is accumulating evidence that distinct 'cells of origin' within an organ can give rise to different subtypes of cancer. Tissue-specific stem and progenitor cells are the predominant targets exploited for tumor initiation. Extrinsic factors imposed by the microenvironment may also directly influence the cell of origin by eliciting dedifferentiation. Identification of these target cell populations is important for earlier diagnosis, the detection of premalignant clones during relapse, and the design of prevention therapies for high-risk cancer families. Here we review recent developments in deciphering the cellular origins of solid cancers.
Collapse
|