1
|
Kirk JS, Wang J, Long M, Rosario S, Tracz A, Ji Y, Kumar R, Liu X, Jamroze A, Singh PK, Puzanov I, Chatta G, Cheng Q, Huang J, Wrana JL, Lovell J, Yu H, Liu S, Shen MM, Liu T, Tang DG. Integrated single-cell analysis defines the epigenetic basis of castration-resistant prostate luminal cells. Cell Stem Cell 2024; 31:1203-1221.e7. [PMID: 38878775 PMCID: PMC11297676 DOI: 10.1016/j.stem.2024.05.008] [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: 03/13/2023] [Revised: 02/26/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024]
Abstract
Understanding prostate response to castration and androgen receptor signaling inhibitors (ARSI) is critical to improving long-term prostate cancer (PCa) patient survival. Here, we use a multi-omics approach on 229,794 single cells to create a mouse single-cell reference atlas for interpreting mouse prostate biology and castration response. Our reference atlas refines single-cell annotations and provides a chromatin context, which, when coupled with mouse lineage tracing, demonstrates that castration-resistant luminal cells are distinct from the pre-existent urethra-proximal stem/progenitor cells. Molecular pathway analysis and therapeutic studies further implicate AP1 (JUN/FOS), WNT/β-catenin, FOXQ1, NF-κB, and JAK/STAT pathways as major drivers of castration-resistant luminal populations with relevance to human PCa. Our datasets, which can be explored through an interactive portal (https://visportal.roswellpark.org/data/tang/), can aid in developing combination treatments with ARSI for advanced PCa patients.
Collapse
Affiliation(s)
- Jason S Kirk
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Jie Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Spencer Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Amanda Tracz
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Yibing Ji
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Xiaozhuo Liu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Anmbreen Jamroze
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Prashant K Singh
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Gurkamal Chatta
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Qing Cheng
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jeffrey L Wrana
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Jonathan Lovell
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY 14260, USA
| | - Han Yu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Michael M Shen
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tao Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
2
|
Yuan L, Meng Y, Xiang J. KLF4 Induces Colorectal Cancer by Promoting EMT via STAT3 Activation. Dig Dis Sci 2024; 69:2841-2855. [PMID: 38816600 DOI: 10.1007/s10620-024-08473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE Krüppel-like factor 4 (KLF4) has been demonstrated to exert a pro-carcinogenic effect in solid tissues. However, the precise biological function and underlying mechanisms in colorectal cancer (CRC) remains elucidated. AIMS To investigate whether KLF4 participates in the proliferation and invasion of CRC. METHODS The expression of KLF4 was investigated using immunohistochemistry and immunoblotting. The clinical significance of KLF4 was evaluated. Furthermore, the effect of inhibiting or overexpressing KLF4 on tumor was examined. Immunoblotting and qPCR were used to detect Epithelial-mesenchymal transition-related proteins levels. Additionally, the molecular function of KLF4 is related to the STAT3 signaling pathway and was determined through JASPAR, GSEA analysis, and in vitro experiments. RESULTS KLF4 exhibits down-regulated expression in CRC and is part of the vessel invasion, TNM stage, and worse prognosis. In vitro studies have shown that KLF4 promotes cellular proliferation and invasion, as well as EMT processes. Xenograft tumor models confirmed the oncogenic role of KLF4 in nude mice. Furthermore, GSEA and JASPAR databases analysis reveal that the binding of KLF4 to the signal transducer and activator of transcription 3 (STAT3) promoter site induces activation of p-STAT3 signaling. Subsequent targeting of STAT3 confirmed its pivotal role in mediating the oncogenic effects exerted by KLF4. CONCLUSION The study suggests that KLF4 activates STAT3 signaling, inducing epithelial-mesenchymal transition, thereby promoting CRC progression.
Collapse
Affiliation(s)
- Lebin Yuan
- Department of Nail and Breast Surgery, Affiliated Xiangyang Central Hospital of Hubei University of Arts and Science, Xiangyang Center Hospital, Xiangyang, Hubei, China
| | - Yanqiu Meng
- Oncology Department, First Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiajia Xiang
- Molecular Centre Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
3
|
Lleshi E, Milne-Clark T, Lee Yu H, Martin HW, Hanson R, Lach R, Rossi SH, Riediger AL, Görtz M, Sültmann H, Flewitt A, Lynch AG, Gnanapragasam VJ, Massie CE, Dev HS. Prostate cancer detection through unbiased capture of methylated cell-free DNA. iScience 2024; 27:110330. [PMID: 39055933 PMCID: PMC11269940 DOI: 10.1016/j.isci.2024.110330] [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: 08/04/2023] [Revised: 05/02/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Prostate cancer screening using prostate-specific antigen (PSA) has been shown to reduce mortality but with substantial overdiagnosis, leading to unnecessary biopsies. The identification of a highly specific biomarker using liquid biopsies, represents an unmet need in the diagnostic pathway for prostate cancer. In this study, we employed a method that enriches for methylated cell-free DNA fragments coupled with a machine learning algorithm which enabled the detection of metastatic and localized cancers with AUCs of 0.96 and 0.74, respectively. The model also detected 51.8% (14/27) of localized and 88.7% (79/89) of patients with metastatic cancer in an external dataset. Furthermore, we show that the differentially methylated regions reflect epigenetic and transcriptomic changes at the tissue level. Notably, these regions are significantly enriched for biologically relevant pathways associated with the regulation of cellular proliferation and TGF-beta signaling. This demonstrates the potential of circulating tumor DNA methylation for prostate cancer detection and prognostication.
Collapse
Affiliation(s)
- Ermira Lleshi
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
- Department of Engineering, University of Cambridge, Cambridge, UK
| | - Toby Milne-Clark
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Henson Lee Yu
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Henno W. Martin
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Robert Hanson
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Radoslaw Lach
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Sabrina H. Rossi
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Anja Lisa Riediger
- University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | | | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Andrew Flewitt
- Department of Engineering, University of Cambridge, Cambridge, UK
| | - Andy G. Lynch
- School of Mathematics and Statistics, University of St Andrews, St Andrews KY16 9SS, UK
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | | | - Charlie E. Massie
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Harveer S. Dev
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| |
Collapse
|
4
|
Farhangnia P, Ghods R, Falak R, Zarnani AH, Delbandi AA. Identification of placenta-specific protein 1 (PLAC-1) expression on human PC-3 cell line-derived prostate cancer stem cells compared to the tumor parental cells. Discov Oncol 2024; 15:251. [PMID: 38943028 PMCID: PMC11213845 DOI: 10.1007/s12672-024-01121-x] [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: 01/04/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024] Open
Abstract
Placenta-specific protein 1 (PLAC-1) is a gene primarily expressed in the placenta and the testis. Interestingly, it is also found to be expressed in many solid tumors, and it is involved in malignant cell features. However, no evidence has been reported regarding the relationship between PLAC-1 and cancer stem cells (CSCs). In the current research, we explored the expression of the PLAC-1 molecule in prostate cancer stem cells (PCSCs) derived from the human PC-3 cell line. The enrichment of PCSCs was achieved using a three-dimensional cell culture technique known as the sphere-formation assay. To confirm the identity of PCSCs, we examined the expression of genes associated with stemness and pluripotency, such as SOX2, OCT4, Nanog, C-Myc, and KLF-4, as well as stem cell differentiation molecules like CD44 and CD133. These evaluations were conducted in both the PCSCs and the original tumor cells (parental cells) using real-time PCR and flow cytometry. Subsequently, we assessed the expression of the PLAC-1 molecule in both enriched cells and parental tumor cells at the gene and protein levels using the same techniques. The tumor cells from the PC-3 cell line formed spheroids with CSC characteristics in a non-adherent medium. The expression of SOX2, OCT4, Nanog, and C-Myc genes (p < 0.01), and the molecules CD44 and CD133 (p < 0.05) were significantly elevated in PCSCs compared to the parental cells. The expression of the PLAC-1 molecule in PCSCs showed a significant increase compared to the parental cells at both gene (p < 0.01) and protein (p < 0.001) levels. In conclusion, it was indicated for the first time that PLAC-1 is up-regulated in PCSCs derived from human PC-3 cell line. This study may propose PLAC-1 as a potential target in targeted therapies, which should be confirmed through further studies.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Zhao Q, Shang Y, Lü J, Liu Y, Wang T, Li D, Li J, Lu Y, Wang Z, Yu Z. miR-29a-KLF4 signaling inhibits breast tumor initiation by regulating cancer stem cells. Int Immunopharmacol 2024; 130:111797. [PMID: 38442582 DOI: 10.1016/j.intimp.2024.111797] [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: 02/08/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Cancer stem cells (CSCs) are known for their potent ability to drive tumor initiation and recurrence, yet the molecular mechanisms regulating CSCs are still unclear. Our study found a positive correlation between increased levels of miR-29a and better survival rates in early-stage breast cancer patients, but a negative correlation in late-stage patients, suggesting a dual function of miR-29a in regulating breast cancer. Furthermore, miR-29a showed significant downregulation in the ALDH+ breast cancer stem cell population compared to non-stem cancer cells. Overexpression of miR-29a in human breast cancer cells reduced the proportion of CSCs, suppressed their ability to form mammospheres, and inhibited the expression of stemness genes SOX2, KLF4, and hTERT in vitro. Conversely, knockdown of miR-29a in breast cancer cells showed opposite effects. Tumor xenograft experiments revealed that miR-29a overexpression significantly inhibited tumorigenesis initiated by MDA-MB-231 cell transplantation in nude mice. We further demonstrated that Krüppel-like factor 4 (KLF4), a key gene that regulates cell stemness, was a direct target of miR-29a in breast cancer cells. miR-29a suppressed the expression of KLF4 at both mRNA and protein levels. Reintroduction of KLF4 into breast cancer cells rescued the miR-29a-induced CSC suppression phenotype. In summary, our study is the first to demonstrate that miR-29a-KLF4 signaling inhibits breast tumor initiation by regulating CSCs, which provides novel therapeutic targets for preventing breast tumor initiation.
Collapse
Affiliation(s)
- Qian Zhao
- Medical Innovation Center and State Key Laboratory of Cardiology, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yuanyuan Shang
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jinhui Lü
- Medical Innovation Center and State Key Laboratory of Cardiology, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yu Liu
- Medical Innovation Center and State Key Laboratory of Cardiology, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; School of Basic Medical Sciences, Jinzhou Medical University, Liaoning, China
| | - Tao Wang
- Medical Innovation Center and State Key Laboratory of Cardiology, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Danni Li
- Medical Innovation Center and State Key Laboratory of Cardiology, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jiayuan Li
- Medical Innovation Center and State Key Laboratory of Cardiology, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; School of Basic Medical Sciences, Jinzhou Medical University, Liaoning, China
| | - Ying Lu
- Medical Innovation Center and State Key Laboratory of Cardiology, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Zhongrui Wang
- Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Zuoren Yu
- Medical Innovation Center and State Key Laboratory of Cardiology, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
6
|
Zhang Y, Li Y. β-hydroxybutyrate inhibits malignant phenotypes of prostate cancer cells through β-hydroxybutyrylation of indoleacetamide-N-methyltransferase. Cancer Cell Int 2024; 24:121. [PMID: 38555451 PMCID: PMC10981303 DOI: 10.1186/s12935-024-03277-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/19/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent cancers in men and is associated with high mortality and disability rates. β-hydroxybutyrate (BHB), a ketone body, has received increasing attention for its role in cancer. However, its role in PCa remains unclear. This study aimed to explore the mechanism and feasibility of BHB as a treatment alternative for PCa. METHODS Colony formation assay, flow cytometry, western blot assay, and transwell assays were performed to determine the effect of BHB on the proliferation and metastasis of PCa cells. Tumor sphere formation and aldehyde dehydrogenase assays were used to identify the impact of BHB or indoleacetamide-N-methyltransferase (INMT) on the stemness of PCa cells. N6-methyladenosine (m6A)-meRIP real-time reverse transcription polymerase chain reaction and dual luciferase assays were conducted to confirm INMT upregulation via the METTL3-m6A pathway. Co-IP assay was used to detect the epigenetic modification of INMT by BHB-mediated β-hydroxybutyrylation (kbhb) and screen enzymes that regulate INMT kbhb. Mouse xenograft experiments demonstrated the antitumor effects of BHB in vivo. RESULTS BHB can inhibit the proliferation, migration, and invasion of PCa cells by suppressing their stemness. Mechanistically, INMT, whose expression is upregulated by the METTL3-m6A pathway, was demonstrated to be an oncogenic gene that promotes the stem-like characteristics of PCa cells. BHB can suppress the malignant phenotypes of PCa by kbhb of INMT, which in turn inhibits INMT expression. CONCLUSIONS Our findings indicate a role of BHB in PCa metabolic therapy, thereby suggesting an epigenetic therapeutic strategy to target INMT in aggressive PCa. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, Henan, Henan, 450000, China.
| | - Yunlong Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, Henan, Henan, 450000, China
| |
Collapse
|
7
|
Ruoff R, Weber H, Wang Y, Huang H, Shapiro E, Fenyö D, Garabedian MJ. MED19 encodes two unique protein isoforms that confer prostate cancer growth under low androgen through distinct gene expression programs. Sci Rep 2023; 13:18227. [PMID: 37880276 PMCID: PMC10600210 DOI: 10.1038/s41598-023-45199-9] [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: 09/27/2022] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
MED19, a component of the mediator complex and a co-regulator of the androgen receptor (AR), is pivotal in prostate cancer cell proliferation. MED19 has two isoforms: a full-length "canonical" and a shorter "alternative" variant. Specific antibodies were developed to investigate these isoforms. Both exhibit similar expression in normal prostate development and adult prostate tissue, but the canonical isoform is elevated in prostate adenocarcinomas. Overexpression of canonical MED19 in LNCaP cells promotes growth under conditions of androgen deprivation in vitro and in vivo, mirroring earlier findings with alternative MED19-overexpressing LNCaP cells. Interestingly, alternative MED19 cells displayed strong colony formation in clonogenic assays under conditions of androgen deprivation, while canonical MED19 cells did not, suggesting distinct functional roles. These isoforms also modulated gene expression differently. Canonical MED19 triggered genes related to extracellular matrix remodeling while suppressing those involved in androgen-inactivating glucuronidation. In contrast, alternative MED19 elevated genes tied to cell movement and reduced those associated with cell adhesion and differentiation. The ratio of MED19 isoform expression in prostate cancers shifts with the disease stage. Early-stage cancers exhibit higher canonical MED19 expression than alternative MED19, consistent with canonical MED19's ability to promote cell proliferation under androgen deprivation. Conversely, alternative MED19 levels were higher in later-stage metastatic prostate cancer than in canonical MED19, reflecting alternative MED19's capability to enhance cell migration and autonomous cell growth. Our findings suggest that MED19 isoforms play unique roles in prostate cancer progression and highlights MED19 as a potential therapeutic target for both early and late-stage prostate cancer.
Collapse
Affiliation(s)
- Rachel Ruoff
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Hannah Weber
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ying Wang
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Hongying Huang
- Department of Urology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ellen Shapiro
- Department of Urology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - David Fenyö
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Michael J Garabedian
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Urology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
8
|
He Z, He J, Xie K. KLF4 transcription factor in tumorigenesis. Cell Death Discov 2023; 9:118. [PMID: 37031197 PMCID: PMC10082813 DOI: 10.1038/s41420-023-01416-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/10/2023] Open
Abstract
Krüppel-like transcriptional factor is important in maintaining cellular functions. Deletion of Krüppel-like transcriptional factor usually causes abnormal embryonic development and even embryonic death. KLF4 is a prominent member of this family, and embryonic deletion of KLF4 leads to alterations in skin permeability and postnatal death. In addition to its important role in embryo development, it also plays a critical role in inflammation and malignancy. It has been investigated that KLF4 has a regulatory role in a variety of cancers, including lung, breast, prostate, colorectal, pancreatic, hepatocellular, ovarian, esophageal, bladder and brain cancer. However, the role of KLF4 in tumorigenesis is complex, which may link to its unique structure with both transcriptional activation and transcriptional repression domains, and to the regulation of its upstream and downstream signaling molecules. In this review, we will summarize the structural and functional aspects of KLF4, with a focus on KLF4 as a clinical biomarker and therapeutic target in different types of tumors.
Collapse
Affiliation(s)
- Zhihong He
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, China
- The South China University of Technology Comprehensive Cancer Center, Guangdong, China
| | - Jie He
- The Second Affiliated Hospital and Guangzhou First People's Hospital, South China University of Technology School of Medicine, Guangdong, China
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, China.
- The South China University of Technology Comprehensive Cancer Center, Guangdong, China.
| |
Collapse
|
9
|
TRIM28 promotes luminal cell plasticity in a mouse model of prostate cancer. Oncogene 2023; 42:1347-1359. [PMID: 36882525 PMCID: PMC10122711 DOI: 10.1038/s41388-023-02655-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/09/2023]
Abstract
The Tripartite motif-containing 28 (TRIM28) transcriptional cofactor is significantly upregulated in high-grade and metastatic prostate cancers. To study the role of TRIM28 in prostate cancer progression in vivo, we generated a genetically-engineered mouse model, combining prostate-specific inactivation of Trp53, Pten and Trim28. Trim28 inactivated NPp53T mice developed an inflammatory response and necrosis in prostate lumens. By conducting single-cell RNA sequencing, we found that NPp53T prostates had fewer luminal cells resembling proximal luminal lineage cells, which are cells with progenitor activity enriched in proximal prostates and prostate invagination tips in wild-type mice with analogous populations in human prostates. However, despite increased apoptosis and reduction of cells expressing proximal luminal cell markers, we found that NPp53T mouse prostates evolved and progressed to invasive prostate carcinoma with a shortened overall survival. Altogether, our findings suggest that TRIM28 promotes expression of proximal luminal cell markers in prostate tumor cells and provides insights into TRIM28 function in prostate tumor plasticity.
Collapse
|
10
|
JAG1 Intracellular Domain Enhances AR Expression and Signaling and Promotes Stem-like Properties in Prostate Cancer Cells. Cancers (Basel) 2022; 14:cancers14225714. [PMID: 36428807 PMCID: PMC9688638 DOI: 10.3390/cancers14225714] [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: 09/05/2022] [Revised: 11/01/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
JAG1 expression is upregulated in high-grade metastatic prostate carcinomas and associated with poor disease-free survival of patients with prostate cancer. Intriguingly, all JAG1-positive prostate carcinomas express JICD although JICD function in prostate cancer (PC) cells is poorly understood. In this study, we found that JICD overexpression increased the expression levels of AR, especially AR-Vs, in PC cell lines and significantly enhanced androgen-independent and androgen-dependent function of ARs. Interestingly, JICD overexpression upregulated the expression of the PCSC marker CD133 in PC cells as the expression of self-renewal markers; namely, NANOG and OCT3/4 increased. In addition, JICD overexpression highly increased the expression of anti-apoptotic BCL-XL protein, while it little affected the expression of apoptotic BIM protein. In 3D cell culture assays, the spheres formed by JICD-overexpressing PC subline cells (C4-2 and CWR22Rv1) were larger than those formed by control (EV) subline cells with undifferentiated morphology. Although JICD overexpression caused quiescence in cell proliferation, it activated the expression of components in PCSC-related signaling pathways, increased PC cell mobility, and promoted in vivo xenograft mouse tumorigenesis. Therefore, JICD may play a crucial role in enhancing androgen independence and promoting stem-like properties in PC cells and should be considered a novel target for CRPC and PCSC diagnostic therapy.
Collapse
|
11
|
Lee SH, Brianna B. Therapeutic Targeting of Overexpressed MiRNAs in Cancer Progression. Curr Drug Targets 2022; 23:1212-1218. [PMID: 35702768 DOI: 10.2174/1389450123666220613163906] [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: 01/28/2022] [Revised: 03/20/2022] [Accepted: 04/29/2022] [Indexed: 01/25/2023]
Abstract
MicroRNAs (miRNAs) are non-coding RNAs involved in the modulation of various biological processes, and their dysregulation is greatly associated with cancer progression as miRNAs can act as either tumour suppressors or oncogenes, depending on their intended target, mechanism of actions, and expression levels. This review paper aims to shed light on the role of overexpressed miRNAs in cancer progression. Cancer cells are known to upregulate specific miRNAs to inhibit the expression of genes regulating the cell cycle, such as PTEN, FOXO1, SOX7, caspases, KLF4, TRIM8, and ZBTB4. Inhibition of these genes promotes cancer development and survival by inducing cell growth, migration, and invasion while evading apoptosis, which leads to poor cancer survival rates. Therefore, the potential of antisense miRNAs in treating cancer is also explored in this review. Antisense miRNAs are chemically modified oligonucleotides that can reverse the action of overexpressed miRNAs. Currently, the therapeutic potential of antisense miRNAs is being validated in both in vitro and in vivo models. Studies have shown that antisense miRNAs could slow down the progression of cancer while enhancing the action of conventional anticancer drugs. These findings provide hope for future oncologic care as this novel intervention is in the process of clinical translation.
Collapse
Affiliation(s)
- Sau Har Lee
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor's University, Subang Jaya, Selangor, Malaysia.,Faculty of Health and Medical Sciences, Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Brianna Brianna
- School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Subbalakshmi AR, Sahoo S, McMullen I, Saxena AN, Venugopal SK, Somarelli JA, Jolly MK. KLF4 Induces Mesenchymal-Epithelial Transition (MET) by Suppressing Multiple EMT-Inducing Transcription Factors. Cancers (Basel) 2021; 13:5135. [PMID: 34680284 PMCID: PMC8533753 DOI: 10.3390/cancers13205135] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022] Open
Abstract
Epithelial-Mesenchymal Plasticity (EMP) refers to reversible dynamic processes where cells can transition from epithelial to mesenchymal (EMT) or from mesenchymal to epithelial (MET) phenotypes. Both these processes are modulated by multiple transcription factors acting in concert. While EMT-inducing transcription factors (TFs)-TWIST1/2, ZEB1/2, SNAIL1/2/3, GSC, and FOXC2-are well-characterized, the MET-inducing TFs are relatively poorly understood (OVOL1/2 and GRHL1/2). Here, using mechanism-based mathematical modeling, we show that transcription factor KLF4 can delay the onset of EMT by suppressing multiple EMT-TFs. Our simulations suggest that KLF4 overexpression can promote a phenotypic shift toward a more epithelial state, an observation suggested by the negative correlation of KLF4 with EMT-TFs and with transcriptomic-based EMT scoring metrics in cancer cell lines. We also show that the influence of KLF4 in modulating the EMT dynamics can be strengthened by its ability to inhibit cell-state transitions at the epigenetic level. Thus, KLF4 can inhibit EMT through multiple parallel paths and can act as a putative MET-TF. KLF4 associates with the patient survival metrics across multiple cancers in a context-specific manner, highlighting the complex association of EMP with patient survival.
Collapse
Affiliation(s)
- Ayalur Raghu Subbalakshmi
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; (A.R.S.); (S.S.); (S.K.V.)
| | - Sarthak Sahoo
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; (A.R.S.); (S.S.); (S.K.V.)
| | | | | | - Sudhanva Kalasapura Venugopal
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; (A.R.S.); (S.S.); (S.K.V.)
| | - Jason A. Somarelli
- Department of Medicine, Duke University, Durham, NC 27708, USA;
- Duke Cancer Institute, Duke University, Durham, NC 27708, USA
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; (A.R.S.); (S.S.); (S.K.V.)
| |
Collapse
|
13
|
Feng Y, Liu X, Pauklin S. 3D chromatin architecture and epigenetic regulation in cancer stem cells. Protein Cell 2021; 12:440-454. [PMID: 33453053 PMCID: PMC8160035 DOI: 10.1007/s13238-020-00819-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/05/2020] [Indexed: 12/29/2022] Open
Abstract
Dedifferentiation of cell identity to a progenitor-like or stem cell-like state with increased cellular plasticity is frequently observed in cancer formation. During this process, a subpopulation of cells in tumours acquires a stem cell-like state partially resembling to naturally occurring pluripotent stem cells that are temporarily present during early embryogenesis. Such characteristics allow these cancer stem cells (CSCs) to give rise to the whole tumour with its entire cellular heterogeneity and thereby support metastases formation while being resistant to current cancer therapeutics. Cancer development and progression are demarcated by transcriptional dysregulation. In this article, we explore the epigenetic mechanisms shaping gene expression during tumorigenesis and cancer stem cell formation, with an emphasis on 3D chromatin architecture. Comparing the pluripotent stem cell state and epigenetic reprogramming to dedifferentiation in cellular transformation provides intriguing insight to chromatin dynamics. We suggest that the 3D chromatin architecture could be used as a target for re-sensitizing cancer stem cells to therapeutics.
Collapse
Affiliation(s)
- Yuliang Feng
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences Old Road, University of Oxford, Oxford, OX3 7LD, UK
| | - Xingguo Liu
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences Old Road, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
14
|
Li C, Yu L, Mai C, Mu T, Zeng Y. KLF4 down-regulation resulting from TLR4 promotion of ERK1/2 phosphorylation underpins inflammatory response in sepsis. J Cell Mol Med 2021; 25:2013-2024. [PMID: 33369167 PMCID: PMC7882990 DOI: 10.1111/jcmm.16082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 10/20/2020] [Accepted: 10/25/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a systemic inflammatory response to invading pathogens, leading to high mortality rates in intensive care units worldwide. Krüppel-like factor 4 (KLF4) is an important anti-inflammatory transcription factor. In this study, we investigate the anti-inflammatory role of KLF4 in caecal ligation and puncture (CLP)-induced septic mice and lipopolysaccharide (LPS)-induced RAW264.7 cells and its potential mechanism. We found that KLF4 was down-regulated in CLP-induced septic mice and in LPS-induced RAW264.7 cells, and that its overexpression led to increased survival rates of septic mice along with inhibited inflammatory response in vivo and in vitro. ITGA2B was up-regulated in the setting of sepsis and was inhibited by KLF4 overexpression. ITGA2B knock-down mimicked the effects of KLF4 overexpression on septic mice and LPS-induced RAW264.7 cells. TLR4 promoted the phosphorylation of ERK1/2 and then up-regulated the ubiquitination and the degradation of KLF4, thereby elevating the expression of ITGA2B. Moreover, TLR4 knock-down or treatment with PD98059 (a MEK inhibitor) inhibited inflammatory response in the setting of sepsis in vivo and in vitro. Furthermore, this effect of PD98059 treatment was lost upon KLF4 knock-down. Collectively, these results explain the down-regulation of KLF4 in sepsis, namely via TLR4 promotion of ERK1/2 phosphorylation, and identify ITGA2B as the downstream gene of KLF4, thus highlighting the anti-inflammatory role of KLF4 in sepsis.
Collapse
Affiliation(s)
- Chunwen Li
- Emergency DepartmentThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Lei Yu
- Emergency DepartmentThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Chao Mai
- Emergency DepartmentAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Tianyi Mu
- Emergency DepartmentAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Yong Zeng
- Emergency DepartmentThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
15
|
MED19 alters AR occupancy and gene expression in prostate cancer cells, driving MAOA expression and growth under low androgen. PLoS Genet 2021; 17:e1008540. [PMID: 33513133 PMCID: PMC7875385 DOI: 10.1371/journal.pgen.1008540] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/10/2021] [Accepted: 01/04/2021] [Indexed: 11/19/2022] Open
Abstract
Androgen deprivation therapy (ADT) is a mainstay of prostate cancer treatment, given the dependence of prostate cells on androgen and the androgen receptor (AR). However, tumors become ADT-resistant, and there is a need to understand the mechanism. One possible mechanism is the upregulation of AR co-regulators, although only a handful have been definitively linked to disease. We previously identified the Mediator subunit MED19 as an AR co-regulator, and reported that MED19 depletion inhibits AR transcriptional activity and growth of androgen-insensitive LNCaP-abl cells. Therefore, we proposed that MED19 upregulation would promote AR activity and drive androgen-independent growth. Here, we show that stable overexpression of MED19 in androgen-dependent LNCaP cells promotes growth under conditions of androgen deprivation. To delineate the mechanism, we determined the MED19 and AR transcriptomes and cistromes in control and MED19-overexpressing LNCaP cells. We also examined genome-wide H3K27 acetylation. MED19 overexpression selectively alters AR occupancy, H3K27 acetylation, and gene expression. Under conditions of androgen deprivation, genes regulated by MED19 correspond to genes regulated by ELK1, a transcription factor that binds the AR N-terminus to induce select AR target gene expression and proliferation, and genomic sites occupied by MED19 and AR are enriched for motifs associated with ELK1. Strikingly, MED19 upregulates expression of monoamine oxidase A (MAOA), a factor that promotes prostate cancer growth. MAOA depletion reduces androgen-independent growth. MED19 and AR occupy the MAOA promoter, with MED19 overexpression enhancing AR occupancy and H3K27 acetylation. Furthermore, MED19 overexpression increases ELK1 occupancy at the MAOA promoter, and ELK1 depletion reduces MAOA expression and androgen-independent growth. This suggests that MED19 cooperates with ELK1 to regulate AR occupancy and H3K27 acetylation at MAOA, upregulating its expression and driving androgen independence in prostate cancer cells. This study provides important insight into the mechanisms of prostate cancer cell growth under low androgen, and underscores the importance of the MED19-MAOA axis in this process.
Collapse
|
16
|
Formaggio N, Rubin MA, Theurillat JP. Loss and revival of androgen receptor signaling in advanced prostate cancer. Oncogene 2021; 40:1205-1216. [PMID: 33420371 PMCID: PMC7892335 DOI: 10.1038/s41388-020-01598-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/20/2020] [Accepted: 11/30/2020] [Indexed: 02/08/2023]
Abstract
Targeting the androgen receptor (AR) signaling axis has been, over decades, the mainstay of prostate cancer therapy. More potent inhibitors of androgen synthesis and antiandrogens have emerged and have been successfully implemented in clinical practice. That said, the stronger inhibition of the AR signaling axis has led in recent years to an increase of prostate cancers that de-differentiate into AR-negative disease. Unfortunately, this process is intimately linked with a poor prognosis. Here, we review the molecular mechanisms that enable cancer cells to switch from an AR-positive to an AR-negative disease and efforts to prevent/revert this process and thereby maintain/restore AR-dependence.
Collapse
Affiliation(s)
- Nicolò Formaggio
- grid.29078.340000 0001 2203 2861Institute of Oncology Research, Università della Svizzera italiana, Lugano, Switzerland
| | - Mark A. Rubin
- grid.5734.50000 0001 0726 5157Department for BioMedical Research and Bern Center of Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
| | - Jean-Philippe Theurillat
- grid.29078.340000 0001 2203 2861Institute of Oncology Research, Università della Svizzera italiana, Lugano, Switzerland
| |
Collapse
|
17
|
Recent Discoveries on the Involvement of Krüppel-Like Factor 4 in the Most Common Cancer Types. Int J Mol Sci 2020; 21:ijms21228843. [PMID: 33266506 PMCID: PMC7700188 DOI: 10.3390/ijms21228843] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023] Open
Abstract
Krüppel-like factor 4 (KLF4) is a transcription factor highly conserved in evolution. It is particularly well known for its role in inducing pluripotent stem cells. In addition, KLF4 plays many roles in cancer. The results of most studies suggest that KLF4 is a tumor suppressor. However, the functioning of KLF4 is regulated at many levels. These include regulation of transcription, alternative splicing, miRNA, post-translational modifications, subcellular localization, protein stability and interactions with other molecules. Simple experiments aimed at assaying transcript levels or protein levels fail to address this complexity and thus may deliver misleading results. Tumor subtypes are also important; for example, in prostate cancer KLF4 is highly expressed in indolent tumors where it impedes tumor progression, while it is absent from aggressive prostate tumors. KLF4 is important in regulating response to many known drugs, and it also plays a role in tumor microenvironment. More and more information is available about upstream regulators, downstream targets and signaling pathways associated with the involvement of KLF4 in cancer. Furthermore, KLF4 performs critical function in the overall regulation of tissue homeostasis, cellular integrity, and progression towards malignancy. Here we summarize and analyze the latest findings concerning this fascinating transcription factor.
Collapse
|
18
|
Sousa L, Pankonien I, Clarke LA, Silva I, Kunzelmann K, Amaral MD. KLF4 Acts as a wt-CFTR Suppressor through an AKT-Mediated Pathway. Cells 2020; 9:cells9071607. [PMID: 32630830 PMCID: PMC7408019 DOI: 10.3390/cells9071607] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cystic Fibrosis (CF) is caused by >2000 mutations in the CF transmembrane conductance regulator (CFTR) gene, but one mutation-F508del-occurs in ~80% of patients worldwide. Besides its main function as an anion channel, the CFTR protein has been implicated in epithelial differentiation, tissue regeneration, and, when dysfunctional, cancer. However, the mechanisms that regulate such relationships are not fully elucidated. Krüppel-like factors (KLFs) are a family of transcription factors (TFs) playing central roles in development, stem cell differentiation, and proliferation. Herein, we hypothesized that these TFs might have an impact on CFTR expression and function, being its missing link to differentiation. Our results indicate that KLF4 (but not KLF2 nor KLF5) is upregulated in CF vs. non-CF cells and that it negatively regulates wt-CFTR expression and function. Of note, F508del-CFTR expressing cells are insensitive to KLF4 modulation. Next, we investigated which KLF4-related pathways have an effect on CFTR. Our data also show that KLF4 modulates wt-CFTR (but not F508del-CFTR) via both the serine/threonine kinase AKT1 (AKT) and glycogen synthase kinase 3 beta (GSK3β) signaling. While AKT acts positively, GSK3β is a negative regulator of CFTR. This crosstalk between wt-CFTR and KLF4 via AKT/ GSK3β signaling, which is disrupted in CF, constitutes a novel mechanism linking CFTR to the epithelial differentiation.
Collapse
Affiliation(s)
- Luis Sousa
- BioISI – Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal; (L.S.); (I.P.); (L.A.C.); (I.S.)
| | - Ines Pankonien
- BioISI – Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal; (L.S.); (I.P.); (L.A.C.); (I.S.)
| | - Luka A Clarke
- BioISI – Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal; (L.S.); (I.P.); (L.A.C.); (I.S.)
| | - Iris Silva
- BioISI – Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal; (L.S.); (I.P.); (L.A.C.); (I.S.)
| | - Karl Kunzelmann
- Department of Physiology, University of Regensburg, 93053 Regensburg, Germany;
| | - Margarida D Amaral
- BioISI – Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal; (L.S.); (I.P.); (L.A.C.); (I.S.)
- Correspondence: ; Tel.: +351-21-750-08-61; Fax: +351-21-750-00-88
| |
Collapse
|
19
|
Lin SR, Mokgautsi N, Liu YN. Ras and Wnt Interaction Contribute in Prostate Cancer Bone Metastasis. Molecules 2020; 25:E2380. [PMID: 32443915 PMCID: PMC7287876 DOI: 10.3390/molecules25102380] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent and malignant cancer types in men, which causes more than three-hundred thousand cancer death each year. At late stage of PCa progression, bone marrow is the most often metastatic site that constitutes almost 70% of metastatic cases of the PCa population. However, the characteristic for the osteo-philic property of PCa is still puzzling. Recent studies reported that the Wnt and Ras signaling pathways are pivotal in bone metastasis and that take parts in different cytological changes, but their crosstalk is not well studied. In this review, we focused on interactions between the Wnt and Ras signaling pathways during each stage of bone metastasis and present the fate of those interactions. This review contributes insights that can guide other researchers by unveiling more details with regard to bone metastasis and might also help in finding potential therapeutic regimens for preventing PCa bone metastasis.
Collapse
Affiliation(s)
- Shian-Ren Lin
- Graduate Institute of Cancer Biology and Drug Discovery, Collage of Medical Science and Technology, Taipei Medical University, Taipei 11024, Taiwan;
| | - Ntlotlang Mokgautsi
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11024, Taiwan;
| | - Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, Collage of Medical Science and Technology, Taipei Medical University, Taipei 11024, Taiwan;
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11024, Taiwan;
| |
Collapse
|
20
|
Tassone E, Bradaschia-Correa V, Xiong X, Sastre-Perona A, Josephson AM, Khodadadi-Jamayran A, Melamed J, Bu L, Kahler DJ, Ossowski L, Leucht P, Schober M, Wilson EL. KLF4 as a rheostat of osteolysis and osteogenesis in prostate tumors in the bone. Oncogene 2019; 38:5766-5777. [PMID: 31239516 PMCID: PMC6639130 DOI: 10.1038/s41388-019-0841-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022]
Abstract
We previously showed that KLF4, a gene highly expressed in murine prostate stem cells, blocks the progression of indolent intraepithelial prostatic lesions into aggressive and rapidly growing tumors. Here, we show that the anti-tumorigenic effect of KLF4 extends to PC3 human prostate cancer cells growing in the bone. We compared KLF4 null cells with cells transduced with a DOX-inducible KLF4 expression system, and find KLF4 function inhibits PC3 growth in monolayer and soft agar cultures. Furthermore, KLF4 null cells proliferate rapidly, forming large, invasive, and osteolytic tumors when injected into mouse femurs, whereas KLF4 re-expression immediately after their intra-femoral inoculation blocks tumor development and preserves a normal bone architecture. KLF4 re-expression in established KLF4 null bone tumors inhibits their osteolytic effects, preventing bone fractures and inducing an osteogenic response with new bone formation. In addition to these profound biological changes, KLF4 also induces a transcriptional shift from an osteolytic program in KLF4 null cells to an osteogenic program. Importantly, bioinformatic analysis shows that genes regulated by KLF4 overlap significantly with those expressed in metastatic prostate cancer patients and in three individual cohorts with bone metastases, strengthening the clinical relevance of the findings in our xenograft model.
Collapse
Affiliation(s)
- Evelyne Tassone
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
| | - Vivian Bradaschia-Correa
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
- Department of Orthopedic Surgery, NYU School of Medicine, New York, NY, 10016, USA
| | - Xiaozhong Xiong
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
| | - Ana Sastre-Perona
- The Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, NY, 10016, USA
| | - Anne Marie Josephson
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
- Department of Orthopedic Surgery, NYU School of Medicine, New York, NY, 10016, USA
| | - Alireza Khodadadi-Jamayran
- Department of Pathology, NYU School of Medicine, New York, NY, 10016, USA
- Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY, 10016, USA
| | - Jonathan Melamed
- Department of Pathology, NYU School of Medicine, New York, NY, 10016, USA
| | - Lei Bu
- Department of Medicine, NYU School of Medicine, New York, NY, 10016, USA
| | - David J Kahler
- High Throughput Biology Laboratory, NYU School of Medicine, New York, NY, 10016, USA
| | - Liliana Ossowski
- Department of Medicine, Mt Sinai School of Medicine, New York, NY, 10029, USA
| | - Philipp Leucht
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
- Department of Orthopedic Surgery, NYU School of Medicine, New York, NY, 10016, USA
| | - Markus Schober
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA.
- The Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, NY, 10016, USA.
| | - Elaine L Wilson
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA.
- Department of Urology, NYU School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
21
|
McCray T, Moline D, Baumann B, Vander Griend DJ, Nonn L. Single-cell RNA-Seq analysis identifies a putative epithelial stem cell population in human primary prostate cells in monolayer and organoid culture conditions. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2019; 7:123-138. [PMID: 31317052 PMCID: PMC6627543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/30/2019] [Indexed: 06/10/2023]
Abstract
Human primary prostate epithelial (PrE) cells represent patient-derived in vitro models and are traditionally grown as a monolayer in two-dimensional culture. It has been recently demonstrated that expansion of primary cells into three-dimensional prostatic organoids better mimics prostate epithelial glands by recapitulating epithelial differentiation and cell polarity. Here, we sought to identify cell populations present in monolayer PrE cells and organoid culture, grown from the same patient, using single-cell RNA-sequencing. Single-cell RNA-sequencing is a powerful tool to analyze transcriptome profiles of thousands of individual cells simultaneously, creating an in-depth atlas of cell populations within a sample. Organoids consisted of six distinct cell clusters (populations) of intermediate differentiation compared to only three clusters in the monolayer prostate epithelial cells. Integrated analysis of the datasets allowed for direct comparison of the monolayer and organoid samples and identified 10 clusters, including a distinct putative prostate stem cell population that was high in Keratin 13 (KRT13), Lymphocyte Antigen 6D (LY6D), and Prostate Stem Cell Antigen (PSCA). Many of the genes within the clusters were validated through RT-qPCR and immunofluorescence in PrE samples from 5 additional patients. KRT13+ cells were observed in discrete areas of the parent tissue and organoids. Pathway analyses and lack of EdU incorporation corroborated a stem-like phenotype based on the gene expression and quiescent state of the KRT13+ cluster. Other clusters within the samples were similar to epithelial populations reported within patient prostate tissues. In summary, these data show that the epithelial stem population is preserved in PrE cultures, with organoids uniquely expanding intermediate cell types not present in monolayer culture.
Collapse
Affiliation(s)
- Tara McCray
- Department of Pathology, University of Illinois at ChicagoChicago 60612, Illinois, USA
| | - Daniel Moline
- Committee on Development, Regenerative, and Stem Cell Biology (DRSB), University of ChicagoChicago 60637, Illinois, USA
| | - Bethany Baumann
- Department of Pathology, University of Illinois at ChicagoChicago 60612, Illinois, USA
| | - Donald J Vander Griend
- Department of Pathology, University of Illinois at ChicagoChicago 60612, Illinois, USA
- University of Illinois Cancer CenterChicago 60612, Illinois, USA
| | - Larisa Nonn
- Department of Pathology, University of Illinois at ChicagoChicago 60612, Illinois, USA
- University of Illinois Cancer CenterChicago 60612, Illinois, USA
| |
Collapse
|
22
|
Feng F, Liu H, Chen A, Xia Q, Zhao Y, Jin X, Huang J. miR‐148‐3p and miR‐152‐3p synergistically regulate prostate cancer progression via repressing KLF4. J Cell Biochem 2019; 120:17228-17239. [PMID: 31104329 DOI: 10.1002/jcb.28984] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 03/23/2019] [Accepted: 04/12/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Feng Feng
- Department of Urology Shandong Provincial Hospital Affiliated to Shandong University Jinan China
- School of Medicine Shandong University Jinan China
| | - Hui Liu
- Department of Urology Shandong Provincial Hospital Affiliated to Shandong University Jinan China
| | - Aiping Chen
- Liaocheng People's Hospital Liao Cheng China
| | - Qinghua Xia
- Department of Urology Shandong Provincial Hospital Affiliated to Shandong University Jinan China
| | - Yong Zhao
- Department of Urology Shandong Provincial Hospital Affiliated to Shandong University Jinan China
| | - Xunbo Jin
- Department of Urology Shandong Provincial Hospital Affiliated to Shandong University Jinan China
| | - Jianjun Huang
- Laboratory of Tumor and Molecular Biology Academy of Military Medical Sciences Beijing China
| |
Collapse
|
23
|
Abstract
Comprehensive knowledge of the normal prostate epithelial lineage hierarchy is a prerequisite to investigate the identity of the cells of origin for prostate cancer. The basal and luminal cells constitute most of the prostate epithelium and have been the major focuses of the study on the cells of origin for prostate cancer. Much progress has been made during the past few decades, mainly using mouse models, to understand the inter-lineage relationship and intra-lineage heterogeneity in adults as well as the lineage plasticity during conditions of stress. These studies have concluded that the adult mouse prostate basal and luminal cells are largely independently sustained under physiological conditions, but both types of cells possess the capacity for bipotent differentiation under stress or artificial experimental conditions. However, the existence or the identity of the putative progenitors within each lineage warrants further investigation. Whether the human prostate lineage hierarchy is completely the same as that of the mouse remains uncertain. Experiments from independent groups have demonstrated that both types of cells in mice and humans can serve as targets for transformation. But controversies remain whether the disease from distinct cells of origin display different clinical behaviors. Further investigation of the intra-lineage heterogeneity will provide new insights into this issue. Understanding the identity of the cells of origin for prostate cancer will help identify novel prognostic markers for early detection of aggressive prostate cancers, provide insights into the therapeutic vulnerability of these tumors, and inspire novel therapeutic strategies.
Collapse
|
24
|
Neural Transcription Factors in Disease Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:437-462. [PMID: 31900920 DOI: 10.1007/978-3-030-32656-2_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progression to the malignant state is fundamentally dependent on transcriptional regulation in cancer cells. Optimum abundance of cell cycle proteins, angiogenesis factors, immune evasion markers, etc. is needed for proliferation, metastasis or resistance to treatment. Therefore, dysregulation of transcription factors can compromise the normal prostate transcriptional network and contribute to malignant disease progression.The androgen receptor (AR) is considered to be a key transcription factor in prostate cancer (PCa) development and progression. Consequently, androgen pathway inhibitors (APIs) are currently the mainstay in PCa treatment, especially in castration-resistant prostate cancer (CRPC). However, emerging evidence suggests that with increased administration of potent APIs, prostate cancer can progress to a highly aggressive disease that morphologically resembles small cell carcinoma, which is referred to as neuroendocrine prostate cancer (NEPC), treatment-induced or treatment-emergent small cell prostate cancer. This chapter will review how neuronal transcription factors play a part in inducing a plastic stage in prostate cancer cells that eventually progresses to a more aggressive state such as NEPC.
Collapse
|