1
|
Rahman R, Selth LA. Cyclin-dependent kinases as mediators of aberrant transcription in prostate cancer. Transl Oncol 2025; 55:102378. [PMID: 40163908 PMCID: PMC11995790 DOI: 10.1016/j.tranon.2025.102378] [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: 11/24/2024] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Transcriptional control of gene expression is fundamental to all cellular processes. Conversely, transcriptional dysregulation is a hallmark of cancer. While this hallmark is a key driver of all malignancy-related process, it also represents a vulnerability that can be exploited therapeutically. Prostate cancer is a prime example of this phenomenon: it is characterised by aberrant transcription and treated with drugs that influence transcriptional pathways. Indeed, the primary oncogenic driver and therapeutic target of prostate cancer, the androgen receptor (AR), is a transcription factor. Moreover, a plethora of other transcriptional regulators, including transcriptional cyclin-dependent kinases (CDK7, CDK8 and CDK9), MYC and Bromodomain-containing protein 4 (BRD4), play prominent roles in disease progression. In this review, we focus on the roles of transcriptional CDKs in prostate cancer growth, metastasis and therapy resistance and discuss their interplay with AR, MYC and BRD4. Additionally, we explore recent advances in the therapeutic targeting of transcriptional CDKs and propose how these strategies could be effectively harnessed for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Razia Rahman
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia
| | - Luke A Selth
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia; Flinders University, Freemasons Centre for Male Health and Wellbeing, Adelaide, South Australia; Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
2
|
Yuan G, Mao J, Li Z. Systematically investigate the mechanism underlying the therapeutic effect of emodin in treatment of prostate cancer. Discov Oncol 2025; 16:413. [PMID: 40148580 PMCID: PMC11950527 DOI: 10.1007/s12672-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVE To systematically investigate the mechanism underlying the therapeutic effect of emodin in treatment of prostate cancer. METHODS Combine network pharmacology, molecular docking, molecular dynamics and experimental verification to explored the mechanism. Using the network pharmacology method, through the TCMSP, DisGeNET and Genecards database, the corresponding targets and related signaling pathways of emodin were screened, and emodin and core targets were studied by molecular docking and molecular dynamics uasing Cytoscape 3.7.2 and other software. The biological processes, cellular components and molecular functions of the key targets were determined by GO enrichment analysis. KEGG enrichment analysis identified signaling pathways associated with key targets. GEPIA and Kaplan-Meier database were used to determine the relationship between the expression of core genes in normal people and prostate cancer patients and the prognosis of patients. Cell proliferation inhibition experiment was carried out by MTT method. The mRNA and protein levels of CASP3, TNF, IL1B, TP53, PPARG, and MYC in PC-3 cells were evaluated by RT-PCR and WB method respectively. RESULTS There were 31 common targets which closely related to emodin in the treatment of prostate cancer. PPI network analysis showed that MYC, PPARG, TP53, TNF, CASP3, IL1B were the core targets. Go and KEGG enrichment analysis showed that pathways in cancer and IL-17 signaling pathway were the key pathways. Molecular docking and molecular dynamics results indicated that emodin had good binding to prostate cancer and 6 core proteins, and the binding force with TP53 protein was the strongest and most stable. The expression of CASP3 protein in normal people was stronger than that in patients with prostate cancer, and the expression of TP53 protein was closely related to the survival rate of patients with prostate cancer. Experimental verification result revealed that EM significantly increased mRNA expressions of CASP3, PPARG and decreased protein expressions of TNF, TP53, MYC at concentrations ranging from 0.1 to 1.6 μmol/L. Emodin significantly increased protein expressions of CASP3, PPARG and decreased protein expressions of TNF, TP53, MYC, IL1B at concentrations ranging from 10 to 160 µmol/L. CONCLUSION Emodin and TP53 have the best binding and stable conformation among core genes. Emodin exhibits a significant inhibitory effect on PC-3 cells at concentration 0.4 ~ 1.6 μmol/L. It showed that anti-prostate cancer properties by regulating cancer and 1L-17 signaling pathway through up-regulating the expressions of CASP3, PPARG genes/proteins, down-regulating IL1B, TP53, TNF, MYC genes/proteins.
Collapse
Affiliation(s)
- Gang Yuan
- Pharmacy Department, Jiulongpo Hospital of Chongqing University of Chinese Medicine, Chongqing Jiulongpo Traditional Chinese Medicine Hospital, Chongqing, 400050, China
| | - Jingxin Mao
- Chongqing Medical and Pharmaceutical College, No. 82, Middle Road of University Town, Shapingba District, Chongqing, 400030, China.
| | - Zheng Li
- Pharmacy Department, Chongqing University Affiliated Renji Hospital, No. 121 Wangxi Road, Nan'an District, Chongqing, 401336, China.
| |
Collapse
|
3
|
Yan W, Liu X, Qiu X, Zhang X, Chen J, Xiao K, Wu P, Peng C, Hu X, Wang Z, Qin J, Sun L, Chen L, Wu D, Huang S, Yin L, Li Z. PRMT5-mediated FUBP1 methylation accelerates prostate cancer progression. J Clin Invest 2024; 134:e175023. [PMID: 39146021 PMCID: PMC11405040 DOI: 10.1172/jci175023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 07/31/2024] [Indexed: 08/17/2024] Open
Abstract
Strategies beyond hormone-related therapy need to be developed to improve prostate cancer mortality. Here, we show that FUBP1 and its methylation were essential for prostate cancer progression, and a competitive peptide interfering with FUBP1 methylation suppressed the development of prostate cancer. FUBP1 accelerated prostate cancer development in various preclinical models. PRMT5-mediated FUBP1 methylation, regulated by BRD4, was crucial for its oncogenic effect and correlated with earlier biochemical recurrence in our patient cohort. Suppressed prostate cancer progression was observed in various genetic mouse models expressing the FUBP1 mutant deficient in PRMT5-mediated methylation. A competitive peptide, which was delivered through nanocomplexes, disrupted the interaction of FUBP1 with PRMT5, blocked FUBP1 methylation, and inhibited prostate cancer development in various preclinical models. Overall, our findings suggest that targeting FUBP1 methylation provides a potential therapeutic strategy for prostate cancer management.
Collapse
Affiliation(s)
- Weiwei Yan
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xun Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, China
| | - Xuefeng Qiu
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, China
| | - Xuebin Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jiahui Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Kai Xiao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ping Wu
- National Facility for Protein Science Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Xiaolin Hu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zengming Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liming Sun
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Luonan Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shengsong Huang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, China
| | - Zhenfei Li
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
4
|
Wu G, Dong Z, Dong Y, Chen Y, Zhu H, Ding D, Cui Y, Wang Y, Xu Y, Chen H. LncRNA CTBP1-AS inhibits TP63-mediated activation of S100A14 during prostate cancer progression. Cancer Sci 2024; 115:1492-1504. [PMID: 38476086 PMCID: PMC11093200 DOI: 10.1111/cas.16138] [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: 09/17/2023] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) have emerged as important molecules and potential new targets for human cancers. This study investigates the function of lncRNA CTBP1 antisense RNA (CTBP1-AS) in prostate cancer (PCa) and explores the entailed molecular mechanism. Aberrantly expressed genes potentially correlated with PCa progression were probed using integrated bioinformatics analyses. A cohort of 68 patients with PCa was included, and their tumor and para-cancerous tissues were collected. CTBP1-AS was highly expressed in PCa tissues and cells and associated with poor patient prognosis. By contrast, tumor protein p63 (TP63) and S100 calcium binding protein A14 (S100A14) were poorly expressed in the PCa tissues and cells. CTBP1-AS did not affect TP63 expression; however it blocked the TP63-mediated transcriptional activation of S100A14, thereby reducing its expression. CTBP1-AS silencing suppressed proliferation, apoptosis resistance, migration, invasion, and tumorigenicity of PCa cell lines, while its overexpression led to inverse results. The malignant phenotype of cells was further weakened by TP63 overexpression but restored following artificial S100A14 silencing. In conclusion, this study demonstrates that CTBP1-AS plays an oncogenic role in PCa by blocking TP63-mediated transcriptional activation of S100A14. This may provide insight into the management of PCa.
Collapse
Affiliation(s)
- Guangzheng Wu
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Zhenkun Dong
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Yuhang Dong
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Yinmei Chen
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Huan Zhu
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Dexin Ding
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Yan Cui
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Yiwen Wang
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Yangyang Xu
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Hui Chen
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| |
Collapse
|
5
|
Nadile M, Sze NSK, Fajardo VA, Tsiani E. Inhibition of Prostate Cancer Cell Survival and Proliferation by Carnosic Acid Is Associated with Inhibition of Akt and Activation of AMPK Signaling. Nutrients 2024; 16:1257. [PMID: 38732504 PMCID: PMC11085396 DOI: 10.3390/nu16091257] [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: 03/07/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Prostate cancer, accounting for 375,304 deaths in 2020, is the second most prevalent cancer in men worldwide. While many treatments exist for prostate cancer, novel therapeutic agents with higher efficacy are needed to target aggressive and hormone-resistant forms of prostate cancer, while sparing healthy cells. Plant-derived chemotherapy drugs such as docetaxel and paclitaxel have been established to treat cancers including prostate cancer. Carnosic acid (CA), a phenolic diterpene found in the herb rosemary (Rosmarinus officinalis) has been shown to have anticancer properties but its effects in prostate cancer and its mechanisms of action have not been examined. CA dose-dependently inhibited PC-3 and LNCaP prostate cancer cell survival and proliferation (IC50: 64, 21 µM, respectively). Furthermore, CA decreased phosphorylation/activation of Akt, mTOR, and p70 S6K. A notable increase in phosphorylation/activation of AMP-activated kinase (AMPK), acetyl-CoA carboxylase (ACC) and its upstream regulator sestrin-2 was seen with CA treatment. Our data indicate that CA inhibits AKT-mTORC1-p70S6K and activates Sestrin-2-AMPK signaling leading to a decrease in survival and proliferation. The use of inhibitors and small RNA interference (siRNA) approaches should be employed, in future studies, to elucidate the mechanisms involved in carnosic acid's inhibitory effects of prostate cancer.
Collapse
Affiliation(s)
- Matteo Nadile
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (M.N.); (N.S.K.S.); (V.A.F.)
| | - Newman Siu Kwan Sze
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (M.N.); (N.S.K.S.); (V.A.F.)
| | - Val A. Fajardo
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (M.N.); (N.S.K.S.); (V.A.F.)
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (M.N.); (N.S.K.S.); (V.A.F.)
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
6
|
Augello MA, Chen X, Liu D, Lin K, Hakansson A, Sjöström M, Khani F, Deonarine LD, Liu Y, Travascio-Green J, Wu J, Loda M, Feng FY, Robinson BD, Davicioni E, Sboner A, Barbieri CE. Canonical AREs are tumor suppressive regulatory elements in the prostate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581466. [PMID: 38464162 PMCID: PMC10925218 DOI: 10.1101/2024.02.23.581466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The androgen receptor (AR) is the central determinant of prostate tissue identity and differentiation, controlling normal, growth-suppressive prostate-specific gene expression 1 . It is also a key driver of prostate tumorigenesis, becoming "hijacked" to drive oncogenic transcription 2-5 . However, the regulatory elements determining the execution of the growth suppressive AR transcriptional program, and whether this can be reactivated in prostate cancer (PCa) cells remains unclear. Canonical androgen response element (ARE) motifs are the classic DNA binding element for AR 6 . Here, we used a genome-wide strategy to modulate regulatory elements containing AREs to define distinct AR transcriptional programs. We find that activation of these AREs is specifically associated with differentiation and growth suppressive transcription, and this can be reactivated to cause death in AR + PCa cells. In contrast, repression of AREs is well tolerated by PCa cells, but deleterious to normal prostate cells. Finally, gene expression signatures driven by ARE activity are associated with improved prognosis and luminal phenotypes in human PCa patients. This study demonstrates that canonical AREs are responsible for a normal, growth-suppressive, lineage-specific transcriptional program, that this can be reengaged in PCa cells for potential therapeutic benefit, and genes controlled by this mechanism are clinically relevant in human PCa patients.
Collapse
|
7
|
Merjaneh N, Hajjar M, Lan YW, Kalinichenko VV, Kalin TV. The Promise of Combination Therapies with FOXM1 Inhibitors for Cancer Treatment. Cancers (Basel) 2024; 16:756. [PMID: 38398147 PMCID: PMC10886945 DOI: 10.3390/cancers16040756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/21/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Forkhead box M1 (FOXM1) is a transcription factor in the forkhead (FOX) family, which is required for cellular proliferation in normal and neoplastic cells. FOXM1 is highly expressed in many different cancers, and its expression is associated with a higher tumor stage and worse patient-related outcomes. Abnormally high expression of FOXM1 in cancers compared to normal tissue makes FOXM1 an attractive target for pharmacological inhibition. FOXM1-inhibiting agents and specific FOXM1-targeted small-molecule inhibitors have been developed in the lab and some of them have shown promising efficacy and safety profiles in mouse models. While the future goal is to translate FOXM1 inhibitors to clinical trials, potential synergistic drug combinations can maximize anti-tumor efficacy while minimizing off-target side effects. Hence, we discuss the rationale and efficacy of all previously studied drug combinations with FOXM1 inhibitors for cancer therapies.
Collapse
Affiliation(s)
- Nawal Merjaneh
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, Division of Hematology and Oncology, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Mona Hajjar
- The Columbian College of Arts and Sciences, George Washington University, Washington, DC 20052, USA;
| | - Ying-Wei Lan
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
| | - Vladimir V. Kalinichenko
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
- Division of Neonatology, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
| | - Tanya V. Kalin
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, Division of Hematology and Oncology, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Phoenix Children’s Research Institute, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA; (Y.-W.L.)
| |
Collapse
|
8
|
Li M, Bai G, Cen Y, Xie Q, Chen J, Chen J, Chen Q, Zhong W, Zhou X. Silencing HOXC13 exerts anti-prostate cancer effects by inducing DNA damage and activating cGAS/STING/IRF3 pathway. J Transl Med 2023; 21:884. [PMID: 38057852 PMCID: PMC10701956 DOI: 10.1186/s12967-023-04743-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Advanced prostate cancer (PCa) will develop into castration-resistant prostate cancer (CRPC) and lead to poor prognosis. As the primary subtype of CRPC, CRPC-AR accounts for the major induction of PCa heterogeneity. CRPC-AR is mainly driven by 25 transcription factors (TFs), which we speculate may be the key factors driving PCa toward CRPC. Therefore, it is necessary to clarify the key regulator and its molecular mechanism mediating PCa progression. METHODS Firstly, we downloaded transcriptomic data and clinical information from TCGA-PRAD. The characteristic gene cluster was identified by PPI clustering, GO enrichment, co-expression correlation and clinical feature analyses for 25 TFs. Then, the effects of 25 TFs expression on prognosis of PCa patients was analyzed using univariate Cox regression, and the target gene was identified. The expression properties of the target gene in PCa tissues were verified using tissue microarray. Meanwhile, the related mechanistic pathway of the target gene was mined based on its function. Next, the target gene was silenced by small interfering RNAs (siRNAs) for cellular function and mechanistic pathway validation. Finally, CIBERSORT algorithm was used to analyze the infiltration levels of 22 immune cells in PCa patients with low and high expression of target gene, and validated by assaying the expression of related immunomodulatory factor. RESULTS We found that HOX family existed independently in 25 TFs, among which HOXC10, HOXC12 and HOXC13 had unique clinical features and the PCa patients with high HOXC13 expression had the worst prognosis. In addition, HOXC13 was highly expressed in tumor tissues and correlated with Gleason score and pathological grade. In vitro experiments demonstrated that silencing HOXC13 inhibited 22RV1 and DU145 cell function by inducing cellular DNA damage and activating cGAS/STING/IRF3 pathway. Immune infiltration analysis revealed that high HOXC13 expression suppressed infiltration of γδ T cells and plasma cells and recruited M2 macrophages. Consistent with these results, silencing HOXC13 up-regulated the transcriptional expression of IFN-β, CCL2, CCL5 and CXCL10. CONCLUSION HOXC13 regulates PCa progression by mediating the DNA damage-induced cGAS/STING/IRF3 pathway and remodels TIME through regulation of the transcription of the immune factors IFN-β, CCL2, CCL5 and CXCL10.
Collapse
Affiliation(s)
- Maozhang Li
- School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Guangwei Bai
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Yi Cen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Qitong Xie
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Jiahong Chen
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Jia Chen
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Qingbiao Chen
- Department of Urology, The Second People's Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, 528000, China
| | - Weide Zhong
- School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| | - Xiaobo Zhou
- Department of Urology, Huizhou Municipal Central Hospital, Huizhou, 516001, China.
| |
Collapse
|
9
|
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
|
10
|
Zeng Z, Fu M, Hu Y, Wei Y, Wei X, Luo M. Regulation and signaling pathways in cancer stem cells: implications for targeted therapy for cancer. Mol Cancer 2023; 22:172. [PMID: 37853437 PMCID: PMC10583419 DOI: 10.1186/s12943-023-01877-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/05/2023] [Indexed: 10/20/2023] Open
Abstract
Cancer stem cells (CSCs), initially identified in leukemia in 1994, constitute a distinct subset of tumor cells characterized by surface markers such as CD133, CD44, and ALDH. Their behavior is regulated through a complex interplay of networks, including transcriptional, post-transcriptional, epigenetic, tumor microenvironment (TME), and epithelial-mesenchymal transition (EMT) factors. Numerous signaling pathways were found to be involved in the regulatory network of CSCs. The maintenance of CSC characteristics plays a pivotal role in driving CSC-associated tumor metastasis and conferring resistance to therapy. Consequently, CSCs have emerged as promising targets in cancer treatment. To date, researchers have developed several anticancer agents tailored to specifically target CSCs, with some of these treatment strategies currently undergoing preclinical or clinical trials. In this review, we outline the origin and biological characteristics of CSCs, explore the regulatory networks governing CSCs, discuss the signaling pathways implicated in these networks, and investigate the influential factors contributing to therapy resistance in CSCs. Finally, we offer insights into preclinical and clinical agents designed to eliminate CSCs.
Collapse
Affiliation(s)
- Zhen Zeng
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Minyang Fu
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuan Hu
- Department of Pediatric Nephrology Nursing, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Min Luo
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
11
|
Boldrini L, Bardi M. WSB1 Involvement in Prostate Cancer Progression. Genes (Basel) 2023; 14:1558. [PMID: 37628609 PMCID: PMC10454498 DOI: 10.3390/genes14081558] [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: 06/29/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Prostate cancer (PC) is polygenic disease involving many genes, and more importantly a host of gene-gene interactions, including transcriptional factors. The WSB1 gene is a transcriptional target of numerous oncoproteins, and its dysregulation can contribute to tumor progression by abnormal activation of targeted oncogenes. Using data from the Cancer Genome Atlas, we tested the possible involvement of WSB1 in PC progression. A multi-dimensional scaling (MDS) model was applied to clarify the association of WSB1 expression with other key genes, such as c-myc, ERG, Enhancer of Zeste 1 and 2 (EHZ1 and EZH2), WNT10a, and WNT 10b. An increased WSB1 expression was associated with higher PC grades and with a worse prognosis. It was also positively related to EZH1, EZH2, WNT10a, and WNT10b. Moreover, MDS showed the central role of WSB1 in influencing the other target genes by its central location on the map. Our study is the first to show a link between WSB1 expression and other genes involved in PC progression, suggesting a novel role for WSB1 in PC progression. This network between WSB1 and EZH2 through WNT/β-catenin may have an important role in PC progression, as suggested by the association between high WSB1 expression and unfavorable prognosis in our analysis.
Collapse
Affiliation(s)
- Laura Boldrini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy
| | - Massimo Bardi
- Department of Psychology & Behavioral Neuroscience, Randolph-Macon College, Ashland, VA 23005, USA
| |
Collapse
|
12
|
Pitzen SP, Dehm SM. Basal epithelial cells in prostate development, tumorigenesis, and cancer progression. Cell Cycle 2023; 22:1303-1318. [PMID: 37098827 PMCID: PMC10228417 DOI: 10.1080/15384101.2023.2206502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 04/27/2023] Open
Abstract
The prostate epithelium is composed of two predominant cell populations: luminal and basal epithelial cells. Luminal cells have a secretory function that supports male fertility while basal cells function in regeneration and maintenance of epithelial tissue. Recent studies in humans and mice have expanded our knowledge of the role and regulation of luminal and basal cells in prostate organogenesis, development, and homeostasis. The insights from healthy prostate biology can inform studies focused on the origins of prostate cancer, progression of the disease, and development of resistance to targeted hormonal therapies. In this review, we discuss a critical role for basal cells in the development and maintenance of healthy prostate tissue. Additionally, we provide evidence supporting a role for basal cells in oncogenesis and therapeutic resistance mechanisms of prostate cancer. Finally, we describe basal cell regulators that may promote lineage plasticity and basal cell identity in prostate cancers that have developed therapeutic resistance. These regulators could serve as therapeutic targets to inhibit or delay resistance and thereby improve outcomes for prostate cancer patients.
Collapse
Affiliation(s)
- Samuel P. Pitzen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Molecular, Cellular, and Developmental Biology and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
13
|
Takahashi S, Takada I. Recent advances in prostate cancer: WNT signaling, chromatin regulation, and transcriptional coregulators. Asian J Androl 2023; 25:158-165. [PMID: 36695247 PMCID: PMC10069695 DOI: 10.4103/aja2022109] [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] [Indexed: 01/22/2023] Open
Abstract
Prostate cancer is one of the most common diseases in men worldwide. Surgery, radiation therapy, and hormonal therapy are effective treatments for early-stage prostate cancer. However, the development of castration-resistant prostate cancer has increased the mortality rate of prostate cancer. To develop novel drugs for castration-resistant prostate cancer, the molecular mechanisms of prostate cancer progression must be elucidated. Among the signaling pathways regulating prostate cancer development, recent studies have revealed the importance of noncanonical wingless-type MMTV integration site family (WNT) signaling pathways, mainly that involving WNT5A, in prostate cancer progression and metastasis; however, its role remains controversial. Moreover, chromatin remodelers such as the switch/sucrose nonfermentable (SWI/SNF) complex and chromodomain helicase DNA-binding proteins 1 also play important roles in prostate cancer progression through genome-wide gene expression changes. Here, we review the roles of noncanonical WNT signaling pathways, chromatin remodelers, and epigenetic enzymes in the development and progression of prostate cancer.
Collapse
Affiliation(s)
- Sayuri Takahashi
- Department of Urology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.,Department of Urology, The Faculty of Medicine, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ichiro Takada
- Department of Urology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
14
|
Manzar N, Ganguly P, Khan UK, Ateeq B. Transcription networks rewire gene repertoire to coordinate cellular reprograming in prostate cancer. Semin Cancer Biol 2023; 89:76-91. [PMID: 36702449 DOI: 10.1016/j.semcancer.2023.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023]
Abstract
Transcription factors (TFs) represent the most commonly deregulated DNA-binding class of proteins associated with multiple human cancers. They can act as transcriptional activators or repressors that rewire the cistrome, resulting in cellular reprogramming during cancer progression. Deregulation of TFs is associated with the onset and maintenance of various cancer types including prostate cancer. An emerging subset of TFs has been implicated in the regulation of multiple cancer hallmarks during tumorigenesis. Here, we discuss the role of key TFs which modulate transcriptional cicuitries involved in the development and progression of prostate cancer. We further highlight the role of TFs associated with key cancer hallmarks, including, chromatin remodeling, genome instability, DNA repair, invasion, and metastasis. We also discuss the pluripotent function of TFs in conferring lineage plasticity, that aids in disease progression to neuroendocrine prostate cancer. At the end, we summarize the current understanding and approaches employed for the therapeutic targeting of TFs and their cofactors in the clinical setups to prevent disease progression.
Collapse
Affiliation(s)
- Nishat Manzar
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Promit Ganguly
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Umar Khalid Khan
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Bushra Ateeq
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India; Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| |
Collapse
|
15
|
Rocha SM, Nascimento D, Cardoso AM, Passarinha L, Socorro S, Maia CJ. STEAP1 regulation and its influence modulating the response of LNCaP prostate cancer cells to bicalutamide, enzalutamide and apalutamide. Mol Med Rep 2023; 27:52. [PMID: 36660947 PMCID: PMC9879076 DOI: 10.3892/mmr.2023.12939] [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: 08/23/2022] [Accepted: 10/25/2022] [Indexed: 01/15/2023] Open
Abstract
Anti‑androgen drugs are the standard pharmacological therapies for treatment of non‑metastatic prostate cancer (PCa). However, the response of PCa cells may depend on the anti‑androgen used and often patients become resistant to treatment. Thus, studying how the anti‑androgen drugs affect oncogenes expression and action and the identification of the best strategy for combined therapies are essential to improve the efficacy of treatments. The Six Transmembrane Epithelial Antigen of the Prostate 1 (STEAP1) is an oncogene associated with PCa progression and aggressiveness, although its relationship with the androgen receptor signaling remains to be elucidated. The present study aimed to evaluate the effect of anti‑androgens in regulating STEAP1 expression and investigate whether silencing STEAP1 can make PCa cells more sensitive to anti‑androgen drugs. For this purpose, wild‑type and STEAP1 knockdown LNCaP cells were exposed to bicalutamide, enzalutamide and apalutamide. Bicalutamide decreased the expression of STEAP1, but enzalutamide and apalutamide increased its expression. However, decreased cell proliferation and increased apoptosis was observed in response to all drugs. Overall, the cellular and molecular effects were similar between LNCaP wild‑type and LNCaP‑STEAP1 knockdown cells, except for c‑myc expression levels, where a cumulative effect between anti‑androgen treatment and STEAP1 knockdown was observed. The effect of STEAP1 knockdown alone or combined with anti‑androgens in c‑myc levels is required to be addressed in future studies.
Collapse
Affiliation(s)
- Sandra M. Rocha
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Daniel Nascimento
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Ana Margarida Cardoso
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - Luís Passarinha
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal,Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2819-516 Caparica, Portugal,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2819-516 Caparica, Portugal,Laboratório de Fármaco-Toxicologia-UBIMedical, Universidade da Beira Interior, 6201-284 Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal,C4-UBI-Cloud Computing Competence Center, Universidade da Beira Interior, 6200-501 Covilhã, Portugal
| | - Cláudio J. Maia
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, 6201-506 Covilhã, Portugal,C4-UBI-Cloud Computing Competence Center, Universidade da Beira Interior, 6200-501 Covilhã, Portugal,Correspondence to: Professor Cláudio J. Maia, CICS-UBI-Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal, E-mail:
| |
Collapse
|
16
|
Wu X, Long X, Ma C, Cheuk YC, Hu M, Hu J, Jiang H. Overexpression of Ubiquitin-Conjugating Enzyme E2C Is Associated with Worsened Prognosis in Prostate Cancer. Int J Mol Sci 2022; 23:ijms232213873. [PMID: 36430352 PMCID: PMC9695011 DOI: 10.3390/ijms232213873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022] Open
Abstract
To evaluate the role of ubiquitin-conjugating enzyme E2C (UBE2C) in prostate cancer (PCa) progression and prognosis, the TCGA and our PCa tissue microarray cohort were included in the study. Weighted gene co-expression network analysis (WGCNA) and non-negative matrix factorization were used to cluster patients and to screen genes that play a vital role in PCa progression (hub gene). Immunohistochemistry staining was used to evaluate the protein level of UBE2C in prostatic tissues. Through WGCNA, we found a gene co-expression module (named the purple module) that is strongly associated with the Gleason score, pathologic T stage, and biochemical recurrent status. Genes in the purple module are enriched in cell cycle and P53 signaling and help us to cluster patients into two groups with distinctive biochemical recurrent survival rates and TP53 mutation statuses. Further analysis showed UBE2C served as a hub gene in the purple module. The expression of UBE2C in PCa was significantly higher than that in paracancerous tissues and was remarkably associated with pathologic grade, Gleason score, and prognosis in PCa patients. To conclude, UBE2C is a PCa-progress-related gene and a biomarker for PCa patients. Therapy targeting UBE2C may serve as a promising treatment of PCa in the future.
Collapse
Affiliation(s)
- Xiaobo Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Xingbo Long
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chenkai Ma
- Diagnostic Solution, Nutrition and Health, CSIRO Health and Biosecurity, Black Mountain, Canberra, ACT 2601, Australia
| | - Yin Celeste Cheuk
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Mengbo Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Jimeng Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200031, China
- Correspondence: (J.H.); (H.J.)
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200031, China
- Correspondence: (J.H.); (H.J.)
| |
Collapse
|
17
|
Pieraccioli M, Caggiano C, Mignini L, Zhong C, Babini G, Lattanzio R, Di Stasi S, Tian B, Sette C, Bielli P. The transcriptional terminator XRN2 and the RNA-binding protein Sam68 link alternative polyadenylation to cell cycle progression in prostate cancer. Nat Struct Mol Biol 2022; 29:1101-1112. [PMID: 36344846 PMCID: PMC9872553 DOI: 10.1038/s41594-022-00853-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2022] [Indexed: 11/09/2022]
Abstract
Alternative polyadenylation (APA) yields transcripts differing in their 3'-end, and its regulation is altered in cancer, including prostate cancer. Here we have uncovered a mechanism of APA regulation impinging on the interaction between the exonuclease XRN2 and the RNA-binding protein Sam68, whose increased expression in prostate cancer is promoted by the transcription factor MYC. Genome-wide transcriptome profiling revealed a widespread impact of the Sam68/XRN2 complex on APA. XRN2 promotes recruitment of Sam68 to its target transcripts, where it competes with the cleavage and polyadenylation specificity factor for binding to strong polyadenylation signals at distal ends of genes, thus promoting usage of suboptimal proximal polyadenylation signals. This mechanism leads to 3' untranslated region shortening and translation of transcripts encoding proteins involved in G1/S progression and proliferation. Thus, our findings indicate that the APA program driven by Sam68/XRN2 promotes cell cycle progression and may represent an actionable target for therapeutic intervention.
Collapse
Affiliation(s)
- Marco Pieraccioli
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy.,GSTEP-Organoids Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| | - Cinzia Caggiano
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy.,GSTEP-Organoids Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Mignini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Chuwei Zhong
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, USA
| | - Gabriele Babini
- GSTEP-Organoids Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| | - Rossano Lattanzio
- Department of Innovative Technologies in Medicine & Dentistry, G. d’Annunzio University, Chieti, Italy.,Center for Advanced Studies and Technology (CAST), G. d’Annunzio University, Chieti, Italy
| | - Savino Di Stasi
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Bin Tian
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, USA
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy. .,GSTEP-Organoids Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy.
| | - Pamela Bielli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy. .,Laboratory of Neuroembryology, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
18
|
Shen M, Demers LK, Bailey SD, Labbé DP. To bind or not to bind: Cistromic reprogramming in prostate cancer. Front Oncol 2022; 12:963007. [PMID: 36212399 PMCID: PMC9539323 DOI: 10.3389/fonc.2022.963007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
The term “cistrome” refers to the genome-wide location of regulatory elements associated with transcription factor binding-sites. The cistrome of key regulatory factors in prostate cancer etiology are substantially reprogrammed and altered during prostatic transformation and disease progression. For instance, the cistrome of the androgen receptor (AR), a ligand-inducible transcription factor central in normal prostate epithelium biology, is directly impacted and substantially reprogrammed during malignant transformation. Accumulating evidence demonstrates that additional transcription factors that are frequently mutated, or aberrantly expressed in prostate cancer, such as the pioneer transcription factors Forkhead Box A1 (FOXA1), the homeobox protein HOXB13, and the GATA binding protein 2 (GATA2), and the ETS-related gene (ERG), and the MYC proto-oncogene, contribute to the reprogramming of the AR cistrome. In addition, recent findings have highlighted key roles for the SWI/SNF complex and the chromatin-modifying helicase CHD1 in remodeling the epigenome and altering the AR cistrome during disease progression. In this review, we will cover the role of cistromic reprogramming in prostate cancer initiation and progression. Specifically, we will discuss the impact of key prostate cancer regulators, as well as the role of epigenetic and chromatin regulators in relation to the AR cistrome and the transformation of normal prostate epithelium. Given the importance of chromatin-transcription factor dynamics in normal cellular differentiation and cancer, an in-depth assessment of the factors involved in producing these altered cistromes is of great relevance and provides insight into new therapeutic strategies for prostate cancer.
Collapse
Affiliation(s)
- Michelle Shen
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Division of Experimental Surgery, Department of Surgery, McGill University, Montréal, QC, Canada
| | - Léa-Kristine Demers
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, QC, Canada
| | - Swneke D. Bailey
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Division of Experimental Surgery, Department of Surgery, McGill University, Montréal, QC, Canada
- Division of Thoracic Surgery, Department of Surgery, McGill University, Montréal, QC, Canada
| | - David P. Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Division of Experimental Surgery, Department of Surgery, McGill University, Montréal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, QC, Canada
- Division of Urology, Department of Surgery, McGill University, Montréal, QC, Canada
- *Correspondence: David P. Labbé,
| |
Collapse
|
19
|
Luthold C, Hallal T, Labbé DP, Bordeleau F. The Extracellular Matrix Stiffening: A Trigger of Prostate Cancer Progression and Castration Resistance? Cancers (Basel) 2022; 14:cancers14122887. [PMID: 35740556 PMCID: PMC9221142 DOI: 10.3390/cancers14122887] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
Despite advancements made in diagnosis and treatment, prostate cancer remains the second most diagnosed cancer among men worldwide in 2020, and the first in North America and Europe. Patients with localized disease usually respond well to first-line treatments, however, up to 30% develop castration-resistant prostate cancer (CRPC), which is often metastatic, making this stage of the disease incurable and ultimately fatal. Over the last years, interest has grown into the extracellular matrix (ECM) stiffening as an important mediator of diseases, including cancers. While this process is increasingly well-characterized in breast cancer, a similar in-depth look at ECM stiffening remains lacking for prostate cancer. In this review, we scrutinize the current state of literature regarding ECM stiffening in prostate cancer and its potential association with disease progression and castration resistance.
Collapse
Affiliation(s)
- Carole Luthold
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
| | - Tarek Hallal
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
| | - David P. Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
- Division of Urology, Department of Surgery, McGill University, Montréal, QC H4A 3J1, Canada
- Correspondence: (D.P.L.); (F.B.)
| | - François Bordeleau
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: (D.P.L.); (F.B.)
| |
Collapse
|
20
|
Qiu X, Brown LG, Conner JL, Nguyen HM, Boufaied N, Abou Alaiwi S, Seo JH, El Zarif T, Bell C, O’Connor E, Hanratty B, Pomerantz M, Freedman ML, Brown M, Haffner MC, Nelson PS, Feng FY, Labbé DP, Long HW, Corey E. Response to supraphysiological testosterone is predicted by a distinct androgen receptor cistrome. JCI Insight 2022; 7:157164. [PMID: 35603787 PMCID: PMC9220831 DOI: 10.1172/jci.insight.157164] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The androgen receptor (AR) is a master transcription factor that regulates prostate cancer (PC) development and progression. Inhibition of AR signaling by androgen deprivation is the first-line therapy with initial efficacy for advanced and recurrent PC. Paradoxically, supraphysiological levels of testosterone (SPT) also inhibit PC progression. However, as with any therapy, not all patients show a therapeutic benefit, and responses differ widely in magnitude and duration. In this study, we evaluated whether differences in the AR cistrome before treatment can distinguish between SPT-responding (R) and -nonresponding (NR) tumors. We provide the first preclinical evidence to our knowledge that SPT-R tumors exhibit a distinct AR cistrome when compared with SPT-NR tumors, indicating a differential biological role of the AR. We applied an integrated analysis of ChIP-Seq and RNA-Seq to the pretreatment tumors and identified an SPT-R signature that distinguishes R and NR tumors. Because transcriptomes of SPT-treated clinical specimens are not available, we interrogated available castration-resistant PC (CRPC) transcriptomes and showed that the SPT-R signature is associated with improved survival and has the potential to identify patients who would respond to SPT. These findings provide an opportunity to identify the subset of patients with CRPC who would benefit from SPT therapy.
Collapse
Affiliation(s)
- Xintao Qiu
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Lisha G. Brown
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Jennifer L. Conner
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Holly M. Nguyen
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Nadia Boufaied
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Sarah Abou Alaiwi
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ji-Heui Seo
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Talal El Zarif
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Connor Bell
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Edward O’Connor
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian Hanratty
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Mark Pomerantz
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew L. Freedman
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Myles Brown
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael C. Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peter S. Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Felix Y. Feng
- University of California at San Francisco, San Francisco, California, USA
| | - David P. Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
- Department of Surgery, Division of Urology, McGill University, Montréal, Québec, Canada
| | - Henry W. Long
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
21
|
Qiu X, Boufaied N, Hallal T, Feit A, de Polo A, Luoma AM, Alahmadi W, Larocque J, Zadra G, Xie Y, Gu S, Tang Q, Zhang Y, Syamala S, Seo JH, Bell C, O'Connor E, Liu Y, Schaeffer EM, Jeffrey Karnes R, Weinmann S, Davicioni E, Morrissey C, Cejas P, Ellis L, Loda M, Wucherpfennig KW, Pomerantz MM, Spratt DE, Corey E, Freedman ML, Shirley Liu X, Brown M, Long HW, Labbé DP. MYC drives aggressive prostate cancer by disrupting transcriptional pause release at androgen receptor targets. Nat Commun 2022; 13:2559. [PMID: 35562350 PMCID: PMC9106722 DOI: 10.1038/s41467-022-30257-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/22/2022] [Indexed: 12/30/2022] Open
Abstract
c-MYC (MYC) is a major driver of prostate cancer tumorigenesis and progression. Although MYC is overexpressed in both early and metastatic disease and associated with poor survival, its impact on prostate transcriptional reprogramming remains elusive. We demonstrate that MYC overexpression significantly diminishes the androgen receptor (AR) transcriptional program (the set of genes directly targeted by the AR protein) in luminal prostate cells without altering AR expression. Analyses of clinical specimens reveal that concurrent low AR and high MYC transcriptional programs accelerate prostate cancer progression toward a metastatic, castration-resistant disease. Data integration of single-cell transcriptomics together with ChIP-seq uncover an increase in RNA polymerase II (Pol II) promoter-proximal pausing at AR-dependent genes following MYC overexpression without an accompanying deactivation of AR-bound enhancers. Altogether, our findings suggest that MYC overexpression antagonizes the canonical AR transcriptional program and contributes to prostate tumor initiation and progression by disrupting transcriptional pause release at AR-regulated genes.
Collapse
Affiliation(s)
- Xintao Qiu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nadia Boufaied
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Tarek Hallal
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| | - Avery Feit
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Anna de Polo
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Division of Urology, Department of Surgery, McGill University, Montréal, QC, Canada
| | - Adrienne M Luoma
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Walaa Alahmadi
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, QC, Canada
| | - Janie Larocque
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, QC, Canada
| | - Giorgia Zadra
- Departments of Oncologic Pathology and Pathology, Dana-Farber Cancer Institute and Brigham's Women Hospital, Boston, MA, USA
- Institute of Molecular Genetics, National Research Council, Pavia, Italy
| | - Yingtian Xie
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Shengqing Gu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Qin Tang
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yi Zhang
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sudeepa Syamala
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Connor Bell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Edward O'Connor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yang Liu
- Decipher Biosciences, San Diego, CA, USA
| | | | | | - Sheila Weinmann
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA
| | | | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Paloma Cejas
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Leigh Ellis
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weil Cornell Medicine, New York Presbyterian-Weill Cornell Campus, New York, NY, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Mark M Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Matthew L Freedman
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
| | - X Shirley Liu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Myles Brown
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Henry W Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - David P Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada.
- Division of Urology, Department of Surgery, McGill University, Montréal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, QC, Canada.
| |
Collapse
|
22
|
Bagherabadi A, Hooshmand A, Shekari N, Singh P, Zolghadri S, Stanek A, Dohare R. Correlation of NTRK1 Downregulation with Low Levels of Tumor-Infiltrating Immune Cells and Poor Prognosis of Prostate Cancer Revealed by Gene Network Analysis. Genes (Basel) 2022; 13:840. [PMID: 35627227 PMCID: PMC9140438 DOI: 10.3390/genes13050840] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/23/2022] Open
Abstract
Prostate cancer (PCa) is a life-threatening heterogeneous malignancy of the urinary tract. Due to the incidence of prostate cancer and the crucial need to elucidate its molecular mechanisms, we searched for possible prognosis impactful genes in PCa using bioinformatics analysis. A script in R language was used for the identification of Differentially Expressed Genes (DEGs) from the GSE69223 dataset. The gene ontology (GO) of the DEGs and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed. A protein-protein interaction (PPI) network was constructed using the STRING online database to identify hub genes. GEPIA and UALCAN databases were utilized for survival analysis and expression validation, and 990 DEGs (316 upregulated and 674 downregulated) were identified. The GO analysis was enriched mainly in the "collagen-containing extracellular matrix", and the KEGG pathway analysis was enriched mainly in "focal adhesion". The downregulation of neurotrophic receptor tyrosine kinase 1 (NTRK1) was associated with a poor prognosis of PCa and had a significant positive correlation with infiltrating levels of immune cells. We acquired a collection of pathways related to primary PCa, and our findings invite the further exploration of NTRK1 as a biomarker for early diagnosis and prognosis, and as a future potential molecular therapeutic target for PCa.
Collapse
Affiliation(s)
- Arash Bagherabadi
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil 56199-11367, Iran;
| | - Amirreza Hooshmand
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom 74147-85318, Iran;
| | - Nooshin Shekari
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz 61357-83151, Iran;
| | - Prithvi Singh
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India; (P.S.); (R.D.)
| | - Samaneh Zolghadri
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom 74147-85318, Iran;
| | - Agata Stanek
- Department and Clinic of Internal Medicine, Angiology and Physical Medicine, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Batorego 15 St., 41-902 Bytom, Poland
| | - Ravins Dohare
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India; (P.S.); (R.D.)
| |
Collapse
|
23
|
FOXA1 Leads to Aberrant Expression of SIX4 Affecting Cervical Cancer Cell Growth and Chemoresistance. Anal Cell Pathol 2022; 2022:9675466. [PMID: 35498155 PMCID: PMC9045987 DOI: 10.1155/2022/9675466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/26/2022] [Indexed: 11/17/2022] Open
Abstract
Cervical cancer (CC) is among the most prevalent cancers among female populations with high recurrence rates all over the world. Cisplatin (DDP) is the first-line treatment for multiple cancers, including CC. The main problem associated with its clinical application is drug resistance. This study is aimed at investigating the function and downstream regulation mechanism of forkhead-box A1 (FOXA1) in CC, which was verified as an oncogene in several cancers. Using GEO database and bioinformatics analysis, we identified FOXA1 as a possible oncogene in CC. Silencing of FOXA1 inhibited CC cell growth, invasion, and chemoresistance. Afterwards, the downstream gene of FOXA1 was predicted using a bioinformatics website and validated using ChIP and dual-luciferase assays. SIX4, a possible target of FOXA1, promoted CC cell malignant aggressiveness and chemoresistance. In addition, overexpression of SIX4 promoted phosphorylation of PI3K and AKT proteins and activated the PI3K/AKT signaling pathway. Further overexpression of SIX4 reversed the repressive effects of FOXA1 knockdown on CC cell growth, invasion, and chemoresistance in DDP-resistant cells. FOXA1-induced SIX4 facilitates CC progression and chemoresistance, highlighting a strong potential for FOXA1 to serve as a promising therapeutic target in CC.
Collapse
|
24
|
Liu S, He B, Li H. Bisphenol S promotes the progression of prostate cancer by regulating the expression of COL1A1 and COL1A2. Toxicology 2022; 472:153178. [DOI: 10.1016/j.tox.2022.153178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
|
25
|
Miyahira AK, Zarif JC, Coombs CC, Flavell RR, Russo JW, Zaidi S, Zhao D, Zhao SG, Pienta KJ, Soule HR. Prostate cancer research in the 21st century; report from the 2021 Coffey-Holden prostate cancer academy meeting. Prostate 2022; 82:169-181. [PMID: 34734426 PMCID: PMC8688282 DOI: 10.1002/pros.24262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 02/03/2023]
Abstract
INTRODUCTION The 2021 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, "Prostate Cancer Research in the 21st Century," was held virtually, from June 24-25, 2021. METHODS The CHPCA Meeting is organized by the Prostate Cancer Foundation as a unique discussion-oriented meeting focusing on critical topics in prostate cancer research envisioned to bridge the next major advances in prostate cancer biology and treatment. The 2021 CHPCA Meeting was virtually attended by 89 investigators and included 31 talks over nine sessions. RESULTS Major topic areas discussed at the meeting included: cancer genomics and sequencing, functional genomic approaches to studying mediators of plasticity, emerging signaling pathways in metastatic castration resistant prostate cancer, Wnt signaling biology and the challenges of targeted therapy, clonal hematopoiesis, neuroendocrine cell plasticity and antitumor immunity, cancer immunotherapy and its synergizers, and imaging the tumor microenvironment and metabolism. DISCUSSION This meeting report summarizes the research presented at the 2021 CHPCA Meeting. We hope that publication of this knowledge will accelerate new understandings and the development of new biomarkers and treatments for prostate cancer.
Collapse
Affiliation(s)
| | - Jelani C. Zarif
- Department of Oncology, Johns Hopkins University School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Catherine C. Coombs
- Department of Medicine, Division of Hematology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA
| | - Joshua W. Russo
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Samir Zaidi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Di Zhao
- Department of Experimental Radiation Oncology, MD Anderson Cancer Center, Houston, TX
| | - Shuang G. Zhao
- Department of Human Oncology, Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Kenneth J. Pienta
- The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, MD
| | | |
Collapse
|
26
|
Liao Y, Chen CH, Xiao T, de la Peña Avalos B, Dray EV, Cai C, Gao S, Shah N, Zhang Z, Feit A, Xue P, Liu Z, Yang M, Lee JH, Xu H, Li W, Mei S, Pierre RS, Shu S, Fei T, Duarte M, Zhao J, Bradner JE, Polyak K, Kantoff PW, Long H, Balk SP, Liu XS, Brown M, Xu K. Inhibition of EZH2 transactivation function sensitizes solid tumors to genotoxic stress. Proc Natl Acad Sci U S A 2022; 119:e2105898119. [PMID: 35031563 PMCID: PMC8784159 DOI: 10.1073/pnas.2105898119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
Drugs that block the activity of the methyltransferase EZH2 are in clinical development for the treatment of non-Hodgkin lymphomas harboring EZH2 gain-of-function mutations that enhance its polycomb repressive function. We have previously reported that EZH2 can act as a transcriptional activator in castration-resistant prostate cancer (CRPC). Now we show that EZH2 inhibitors can also block the transactivation activity of EZH2 and inhibit the growth of CRPC cells. Gene expression and epigenomics profiling of cells treated with EZH2 inhibitors demonstrated that in addition to derepressing gene expression, these compounds also robustly down-regulate a set of DNA damage repair (DDR) genes, especially those involved in the base excision repair (BER) pathway. Methylation of the pioneer factor FOXA1 by EZH2 contributes to the activation of these genes, and interaction with the transcriptional coactivator P300 via the transactivation domain on EZH2 directly turns on the transcription. In addition, CRISPR-Cas9-mediated knockout screens in the presence of EZH2 inhibitors identified these BER genes as the determinants that underlie the growth-inhibitory effect of EZH2 inhibitors. Interrogation of public data from diverse types of solid tumors expressing wild-type EZH2 demonstrated that expression of DDR genes is significantly correlated with EZH2 dependency and cellular sensitivity to EZH2 inhibitors. Consistent with these findings, treatment of CRPC cells with EZH2 inhibitors dramatically enhances their sensitivity to genotoxic stress. These studies reveal a previously unappreciated mechanism of action of EZH2 inhibitors and provide a mechanistic basis for potential combination cancer therapies.
Collapse
Affiliation(s)
- Yiji Liao
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Chen-Hao Chen
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | - Tengfei Xiao
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Bárbara de la Peña Avalos
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Eloise V Dray
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts, Boston, MA 02125
| | - Shuai Gao
- Center for Personalized Cancer Therapy, University of Massachusetts, Boston, MA 02125
| | - Neel Shah
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Zhao Zhang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Avery Feit
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Pengya Xue
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Zhijie Liu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Mei Yang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Ji Hoon Lee
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Han Xu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Wei Li
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Shenglin Mei
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Roodolph S Pierre
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
- Biological and Biomedical Science Program, Harvard Medical School, Boston, MA 02115
| | - Shaokun Shu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Teng Fei
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Melissa Duarte
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Jin Zhao
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - James E Bradner
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
- Biological and Biomedical Science Program, Harvard Medical School, Boston, MA 02115
| | - Kornelia Polyak
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Philip W Kantoff
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Henry Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Steven P Balk
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - X Shirley Liu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115;
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | - Myles Brown
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115;
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Kexin Xu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229;
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115
| |
Collapse
|
27
|
A Transcription Factor-Based Risk Model for Predicting the Prognosis of Prostate Cancer and Potential Therapeutic Drugs. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6894278. [PMID: 34853602 PMCID: PMC8629613 DOI: 10.1155/2021/6894278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/28/2021] [Indexed: 12/13/2022]
Abstract
Background Prostate cancer (PC) is one of the most critical cancers affecting men's health worldwide. The development of many cancers involves dysregulation or mutations in key transcription factors. This study established a transcription factor-based risk model to predict the prognosis of PC and potential therapeutic drugs. Materials and Methods In this study, RNA-sequencing data were downloaded and analyzed using The Cancer Genome Atlas dataset. A total of 145 genes related to the overall survival rate of PC patients were screened using the univariate Cox analysis. The Kdmist clustering method was used to classify prostate adenocarcinoma (PRAD), thereby determining the cluster related to the transcription factors. The support vector machine-recursive feature elimination method was used to identify genes related to the types of transcription factors and the key genes specifically upregulated or downregulated were screened. These genes were further analyzed using Lasso to establish a model. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for the functional analysis. The TIMER algorithm was used to quantify the abundance of immune cells in PRAD samples. The chemotherapy response of each GBM patient was predicted based on the public pharmacogenomic database, Genomics of Drug Sensitivity in Cancer (GDSC, http://www.cancerrxgene.org). The R package "pRRophetic" was applied to drug sensitivity (IC50) value prediction. Results We screened 10 genes related to prognosis, including eight low-risk genes and two high-risk genes. The receiver operating characteristic (ROC) curve was 0.946. Patients in the high-risk score group had a poorer prognosis than those in the low-risk score group. The average area under the curve value of the model at different times was higher than 0.8. The risk score was an independent prognostic factor. Compared with the low-risk score group, early growth response-1 (EGR1), CACNA2D1, AC005831.1, SLC52A3, TMEM79, IL20RA, CRACR2A, and FAM189A2 expressions in the high-risk score group were decreased, while AC012181.1 and TRAPPC8 expressions were increased. GO and KEGG analyses showed that prognosis was related to various cancer signaling pathways. The proportion of B_cell, T_cell_CD4, and macrophages in the high-risk score group was significantly higher than that in the low-risk score group. A total of 25 classic immune checkpoint genes were screened out to express abnormally high-risk scores, and there were significant differences. Thirty mutant genes were identified; in the high- and low-risk score groups, SPOP, TP53, and TTN had the highest mutation frequency, and their mutations were mainly missense mutations. A total of 36 potential drug candidates for the treatment of PC were screened and identified. Conclusions Ten genes of both high-and low-risk scores were associated with the prognosis of PC. PC prognosis may be related to immune disorders. SPOP, TP53, and TTN may be potential targets for the prognosis of PC.
Collapse
|
28
|
Chaves LP, Melo CM, Saggioro FP, dos Reis RB, Squire JA. Epithelial-Mesenchymal Transition Signaling and Prostate Cancer Stem Cells: Emerging Biomarkers and Opportunities for Precision Therapeutics. Genes (Basel) 2021; 12:1900. [PMID: 34946849 PMCID: PMC8701270 DOI: 10.3390/genes12121900] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancers may reactivate a latent embryonic program called the epithelial-mesenchymal transition (EMT) during the development of metastatic disease. Through EMT, tumors can develop a mesenchymal phenotype similar to cancer stem cell traits that contributes to metastasis and variation in therapeutic responses. Some of the recurrent somatic mutations of prostate cancer affect EMT driver genes and effector transcription factors that induce the chromatin- and androgen-dependent epigenetic alterations that characterize castrate-resistant prostate cancer (CRPC). EMT regulators in prostate cancer comprise transcription factors (SNAI1/2, ZEB1, TWIST1, and ETS), tumor suppressor genes (RB1, PTEN, and TP53), and post-transcriptional regulators (miRNAs) that under the selective pressures of antiandrogen therapy can develop an androgen-independent metastatic phenotype. In prostate cancer mouse models of EMT, Slug expression, as well as WNT/β-Catenin and notch signaling pathways, have been shown to increase stemness potential. Recent single-cell transcriptomic studies also suggest that the stemness phenotype of advanced prostate cancer may be related to EMT. Other evidence correlates EMT and stemness with immune evasion, for example, activation of the polycomb repressor complex I, promoting EMT and stemness and cytokine secretion through RB1, TP53, and PRC1. These findings are helping clinical trials in CRPC that seek to understand how drugs and biomarkers related to the acquisition of EMT can improve drug response.
Collapse
Affiliation(s)
- Luiz Paulo Chaves
- Department of Genetics, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14048-900, SP, Brazil; (L.P.C.); (C.M.M.)
| | - Camila Morais Melo
- Department of Genetics, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14048-900, SP, Brazil; (L.P.C.); (C.M.M.)
| | - Fabiano Pinto Saggioro
- Pathology Department, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14048-900, SP, Brazil;
| | - Rodolfo Borges dos Reis
- Division of Urology, Department of Surgery and Anatomy, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14048-900, SP, Brazil;
| | - Jeremy Andrew Squire
- Department of Genetics, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14048-900, SP, Brazil; (L.P.C.); (C.M.M.)
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
29
|
Singh KB, Hahm ER, Singh SV. Leelamine suppresses cMyc expression in prostate cancer cells in vitro and inhibits prostate carcinogenesis in vivo. JOURNAL OF CANCER METASTASIS AND TREATMENT 2021; 7. [PMID: 34660908 DOI: 10.20517/2394-4722.2021.08] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aim Leelamine (LLM) inhibits growth of human prostate cancer cells but the underlying mechanism is not fully understood. The present study was undertaken to determine the effect of LLM on cMyc, which is overexpressed in a subset of human prostate cancers. Methods The effect of LLM on cMyc expression and activity was determined by western blotting/confocal microscopy and luciferase reporter assay, respectively. A transgenic mouse model of prostate cancer (Hi-Myc) was used to determine chemopreventive efficacy of LLM. Results Exposure of androgen sensitive (LNCaP) and castration-resistant (22Rv1) human prostate cancer cells to LLM resulted in downregulation of protein and mRNA levels of cMyc. Overexpression of cMyc partially attenuated LLM-mediated inhibition of colony formation, cell viability, and cell migration in 22Rv1 and/or PC-3 cells. LLM treatment decreased protein levels of cMyc targets (e.g., lactate dehydrogenase), however, overexpression of cMyc did not attenuate these effects. A trend for a decrease in expression level of cMyc protein was discernible in 22Rv1 xenografts from LLM-treated mice compared with control mice. The LLM treatment (10 mg/kg body weight, 5 times/week) was well-tolerated by Hi-Myc transgenic mice. The incidence of high-grade prostatic intraepithelial neoplasia, adenocarcinoma in situ, and microinvasion was lower in LLM-treated Hi-Myc mice but the difference was not statistically significant. Conclusion The present study reveals that LLM inhibits cMyc expression in human prostate cancer cells in vitro but concentrations higher than 10 mg/kg may be required to achieve chemoprevention of prostate cancer.
Collapse
Affiliation(s)
- Krishna B Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Eun-Ryeong Hahm
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shivendra V Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
30
|
Transcriptomic analysis of castration, chemo-resistant and metastatic prostate cancer elucidates complex genetic crosstalk leading to disease progression. Funct Integr Genomics 2021; 21:451-472. [PMID: 34184132 DOI: 10.1007/s10142-021-00789-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/05/2020] [Accepted: 05/06/2021] [Indexed: 12/22/2022]
Abstract
Prostate adenocarcinoma, with its rising numbers and high fatality rate, is a daunting healthcare challenge to clinicians and researchers alike. The mainstay of our meta-analysis was to decipher differentially expressed genes (DEGs), their corresponding transcription factors (TFs), miRNAs (microRNA) and interacting pathways underlying the progression of prostate cancer (PCa). We have chosen multiple datasets from primary, castration-resistant, chemo-resistant and metastatic prostate cancer stages for investigation. From our tissue-specific and disease-specific co-expression networks, fifteen hub genes such as ACTB, ACTN1, CDH1, CDKN1A, DDX21, ELF3, FLNA, FLNC, IKZF1, ILK, KRT13, KRT18, KRT19, SVIL and TRIM29 were identified and validated by molecular complex detection analysis as well as survival analysis. In our attempt to highlight hub gene-associated mutations and drug interactions, FLNC was found to be most commonly mutated and CDKN1A gene was found to have highest druggability. Moreover, from DAVID and gene set enrichment analysis, the focal adhesion and oestrogen signalling pathways were found enriched which indicates the involvement of hub genes in tumour invasiveness and metastasis. Finally by Enrichr tool and miRNet, we identified transcriptional factors SNAI2, TP63, CEBPB and KLF11 and microRNAs, namely hsa-mir-1-3p, hsa-mir-145-5p, hsa-mir-124-3p and hsa-mir-218-5p significantly controlling the hub gene expressions. In a nutshell, our report will help to gain a deeper insight into complex molecular intricacies and thereby unveil the probable biomarkers and therapeutic targets involved with PCa progression.
Collapse
|
31
|
Phua TJ. The Etiology and Pathophysiology Genesis of Benign Prostatic Hyperplasia and Prostate Cancer: A New Perspective. MEDICINES 2021; 8:medicines8060030. [PMID: 34208086 PMCID: PMC8230771 DOI: 10.3390/medicines8060030] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022]
Abstract
Background: The etiology of benign prostatic hyperplasia and prostate cancer are unknown, with ageing being the greatness risk factor. Methods: This new perspective evaluates the available interdisciplinary evidence regarding prostate ageing in terms of the cell biology of regulation and homeostasis, which could explain the timeline of evolutionary cancer biology as degenerative, inflammatory and neoplasm progressions in these multifactorial and heterogeneous prostatic diseases. Results: This prostate ageing degeneration hypothesis encompasses the testosterone-vascular-inflamm-ageing triad, along with the cell biology regulation of amyloidosis and autophagy within an evolutionary tumorigenesis microenvironment. Conclusions: An understanding of these biological processes of prostate ageing can provide potential strategies for early prevention and could contribute to maintaining quality of life for the ageing individual along with substantial medical cost savings.
Collapse
Affiliation(s)
- Teow J Phua
- Molecular Medicine, NSW Health Pathology, John Hunter Hospital, Newcastle, NSW 2305, Australia
| |
Collapse
|
32
|
Replication Stress, Genomic Instability, and Replication Timing: A Complex Relationship. Int J Mol Sci 2021; 22:ijms22094764. [PMID: 33946274 PMCID: PMC8125245 DOI: 10.3390/ijms22094764] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/29/2022] Open
Abstract
The replication-timing program constitutes a key element of the organization and coordination of numerous nuclear processes in eukaryotes. This program is established at a crucial moment in the cell cycle and occurs simultaneously with the organization of the genome, thus indicating the vital significance of this process. With recent technological achievements of high-throughput approaches, a very strong link has been confirmed between replication timing, transcriptional activity, the epigenetic and mutational landscape, and the 3D organization of the genome. There is also a clear relationship between replication stress, replication timing, and genomic instability, but the extent to which they are mutually linked to each other is unclear. Recent evidence has shown that replication timing is affected in cancer cells, although the cause and consequence of this effect remain unknown. However, in-depth studies remain to be performed to characterize the molecular mechanisms of replication-timing regulation and clearly identify different cis- and trans-acting factors. The results of these studies will potentially facilitate the discovery of new therapeutic pathways, particularly for personalized medicine, or new biomarkers. This review focuses on the complex relationship between replication timing, replication stress, and genomic instability.
Collapse
|
33
|
Su Y, Zhou W, Zhang Y, Wang X, Han B. Identification And validation of transcription factor genes involved in prostate cancer metastasis. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1915394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Yiming Su
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Wenhao Zhou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yu Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaohai Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Bangmin Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
34
|
Boija A, Klein IA, Young RA. Biomolecular Condensates and Cancer. Cancer Cell 2021; 39:174-192. [PMID: 33417833 PMCID: PMC8721577 DOI: 10.1016/j.ccell.2020.12.003] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022]
Abstract
Malignant transformation is characterized by dysregulation of diverse cellular processes that have been the subject of detailed genetic, biochemical, and structural studies, but only recently has evidence emerged that many of these processes occur in the context of biomolecular condensates. Condensates are membrane-less bodies, often formed by liquid-liquid phase separation, that compartmentalize protein and RNA molecules with related functions. New insights from condensate studies portend a profound transformation in our understanding of cellular dysregulation in cancer. Here we summarize key features of biomolecular condensates, note where they have been implicated-or will likely be implicated-in oncogenesis, describe evidence that the pharmacodynamics of cancer therapeutics can be greatly influenced by condensates, and discuss some of the questions that must be addressed to further advance our understanding and treatment of cancer.
Collapse
Affiliation(s)
- Ann Boija
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Isaac A Klein
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
35
|
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: 2.8] [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
|
36
|
Fernandes RC, Toubia J, Townley S, Hanson AR, Dredge BK, Pillman KA, Bert AG, Winter JM, Iggo R, Das R, Obinata D, Sandhu S, Risbridger GP, Taylor RA, Lawrence MG, Butler LM, Zoubeidi A, Gregory PA, Tilley WD, Hickey TE, Goodall GJ, Selth LA. Post-transcriptional Gene Regulation by MicroRNA-194 Promotes Neuroendocrine Transdifferentiation in Prostate Cancer. Cell Rep 2021; 34:108585. [PMID: 33406413 DOI: 10.1016/j.celrep.2020.108585] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/23/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Potent therapeutic inhibition of the androgen receptor (AR) in prostate adenocarcinoma can lead to the emergence of neuroendocrine prostate cancer (NEPC), a phenomenon associated with enhanced cell plasticity. Here, we show that microRNA-194 (miR-194) is a regulator of epithelial-neuroendocrine transdifferentiation. In clinical prostate cancer samples, miR-194 expression and activity were elevated in NEPC and inversely correlated with AR signaling. miR-194 facilitated the emergence of neuroendocrine features in prostate cancer cells, a process mediated by its ability to directly target a suite of genes involved in cell plasticity. One such target was FOXA1, which encodes a transcription factor with a vital role in maintaining the prostate epithelial lineage. Importantly, a miR-194 inhibitor blocked epithelial-neuroendocrine transdifferentiation and inhibited the growth of cell lines and patient-derived organoids possessing neuroendocrine features. Overall, our study reveals a post-transcriptional mechanism regulating the plasticity of prostate cancer cells and provides a rationale for targeting miR-194 in NEPC.
Collapse
Affiliation(s)
- Rayzel C Fernandes
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - John Toubia
- ACRF Cancer Genomics Facility, Centre for Cancer Biology, An alliance of SA Pathology and University of South Australia, Frome Road, Adelaide, SA 5005, Australia
| | - Scott Townley
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Adrienne R Hanson
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - B Kate Dredge
- Centre for Cancer Biology, An alliance of SA Pathology and University of South Australia, Adelaide, SA 5005, Australia
| | - Katherine A Pillman
- Centre for Cancer Biology, An alliance of SA Pathology and University of South Australia, Adelaide, SA 5005, Australia
| | - Andrew G Bert
- Centre for Cancer Biology, An alliance of SA Pathology and University of South Australia, Adelaide, SA 5005, Australia
| | - Jean M Winter
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Richard Iggo
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Institut Bergonié Unicancer, INSERM U1218, Bordeaux, France
| | - Rajdeep Das
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Daisuke Obinata
- Department of Urology, Nihon University School of Medicine, Tokyo 173-8610, Japan; Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, VIC 3168, Australia
| | -
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, VIC 3168, Australia; Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Shahneen Sandhu
- Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, VIC 3168, Australia; Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3000, Australia
| | - Renea A Taylor
- Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC 3000, Australia; Department of Physiology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, VIC 3168, Australia
| | - Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute, Prostate Cancer Research Group, Monash University, Clayton, VIC 3168, Australia; Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Lisa M Butler
- South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Amina Zoubeidi
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Philip A Gregory
- Centre for Cancer Biology, An alliance of SA Pathology and University of South Australia, Adelaide, SA 5005, Australia; Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Gregory J Goodall
- Centre for Cancer Biology, An alliance of SA Pathology and University of South Australia, Adelaide, SA 5005, Australia; School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia.
| |
Collapse
|
37
|
Gray JS, Campbell MJ. Challenges and Opportunities of Genomic Approaches in Therapeutics Development. Methods Mol Biol 2021; 2194:107-126. [PMID: 32926364 DOI: 10.1007/978-1-0716-0849-4_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The magnitude of all therapeutic responses is significantly determined by genome structure, variation, and functional interactions. This determination occurs at many levels which are discussed in the current review. Well-established examples of structural variation between individuals are known to dictate an individual's response to numerous drugs, as clearly illustrated by warfarin. The exponential rate of genomic-based interrogation is coupled with an expanding repertoire of genomic technologies and applications. This is leading to an ever more sophisticated appreciation of how structural variation, regulation of transcription and genomic structure, both individually and collectively, define cell therapeutic responses.
Collapse
Affiliation(s)
- Jaimie S Gray
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Moray J Campbell
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
38
|
Leng X, Liu M, Tao D, Yang B, Zhang Y, He T, Xie S, Wang Z, Liu Y, Yang Y. Epigenetic modification-dependent androgen receptor occupancy facilitates the ectopic TSPY1 expression in prostate cancer cells. Cancer Sci 2020; 112:691-702. [PMID: 33185915 PMCID: PMC7894013 DOI: 10.1111/cas.14731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/27/2020] [Accepted: 11/07/2020] [Indexed: 02/06/2023] Open
Abstract
Testis‐specific protein Y‐encoded 1 (TSPY1), a Y chromosome‐linked oncogene, is frequently activated in prostate cancers (PCa) and its expression is correlated with the poor prognosis of PCa. However, the cause of the ectopic transcription of TSPY1 in PCa remains unclear. Here, we observed that the methylation status in the CpG islands (CGI) of the TSPY1 promoter was negatively correlated with its expression level in different human samples. The acetyl‐histone H4 and trimethylated histone H3‐lysine 4, two post–translational modifications of histones occupying the TSPY1 promoter, facilitated the TSPY1 expression in PCa cells. In addition, we found that androgen accelerated the TSPY1 transcription on the condition of hypomethylated of TSPY1‐CGI and promoted PCa cell proliferation. Moreover, the binding of androgen receptor (AR) to the TSPY1 promoter, enhancing TSPY1 transcription, was detected in PCa cells. Taken together, our findings identified the regulation of DNA methylation, acting as a primary mechanism, on TSPY1 expression in PCa, and revealed that TSPY1 is an androgen‐AR axis‐regulated oncogene, suggesting a novel and potential target for PCa therapy.
Collapse
Affiliation(s)
- Xiangyou Leng
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Mohan Liu
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Dachang Tao
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Bo Yang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yangwei Zhang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Tianrong He
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Shengyu Xie
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Zhaokun Wang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yunqiang Liu
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yuan Yang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Transcriptional network modulated by the prognostic signature transcription factors and their long noncoding RNA partners in primary prostate cancer. EBioMedicine 2020; 63:103150. [PMID: 33279858 PMCID: PMC7718452 DOI: 10.1016/j.ebiom.2020.103150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/02/2020] [Indexed: 12/02/2022] Open
Abstract
Background Transcriptional regulators are seminal players in the onset and progression of prostate cancer. However, clarification of their underlying regulatory circuits and mechanisms demands considerable effort. Methods Integrated analyses were performed on genomic, transcriptomic, and clinicopathological profiles of primary prostate cancer and transcription factor-binding profiles, which included estimating transcription factor activity, identifying transcription factors of prognostic values, and discovering cis- and trans-regulations by long noncoding RNAs. Interactions between transcription factors and long noncoding RNAs were validated by RNA immunoprecipitation quantitative PCR. RNA interference assays were performed to explore roles of the selected transcription regulators. Findings Sixteen transcription factors, namely, ETS1, ARID4B, KLF12, GMEB1, HBP1, MXI1, MYC, MAX, PGR, BCL11A, AR, KLF4, SRF, HIF1A, EHF, and ATOH1, were jointly identified as a prognostic signature. Candidate long noncoding RNAs interplaying with the prognostic signature constituent transcription factors were further discovered. Their interactions were randomly checked, and many of them were experimentally proved. Transcription regulation by MYC and its long noncoding RNA partner AL590617.2 was further validated on their candidate targets. Moreover, the regulatory network governed by the transcription factors and their interacting long noncoding RNA partners is illustrated and stored in our LNCTRN database (https://navy.shinyapps.io/lnctrn). Interpretation The prognostic signature constituent transcription factors and their interacting long noncoding RNAs may represent promising biomarkers and/or therapeutic targets for prostate cancer. Furthermore, the computational framework proposed in the present study can be utilized to explore critical transcriptional regulators in other types of cancer. Funding This work was supported by National Natural Science Foundation of China and Fudan University.
Collapse
|
40
|
Super-enhancer in prostate cancer: transcriptional disorders and therapeutic targets. NPJ Precis Oncol 2020; 4:31. [PMID: 33299103 PMCID: PMC7677538 DOI: 10.1038/s41698-020-00137-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022] Open
Abstract
Abnormal activity of oncogenic and tumor-suppressor signaling pathways contributes to cancer and cancer risk in humans. Transcriptional dysregulation of these pathways is commonly associated with tumorigenesis and the development of cancer. Genetic and epigenetic alterations may mediate dysregulated transcriptional activity. One of the most important epigenetic alternations is the non-coding regulatory element, which includes both enhancers and super-enhancers (SEs). SEs, characterized as large clusters of enhancers with aberrant high levels of transcription factor binding, have been considered as key drivers of gene expression in controlling and maintaining cancer cell identity. In cancer cells, oncogenes acquire SEs and the cancer phenotype relies on these abnormal transcription programs driven by SEs, which leads to cancer cells often becoming addicted to the SEs-related transcription programs, including prostate cancer. Here, we summarize recent findings of SEs and SEs-related gene regulation in prostate cancer and review the potential pharmacological inhibitors in basic research and clinical trials.
Collapse
|
41
|
Gilbreath C, Ma S, Yu L, Sonavane R, Roggero CM, Devineni A, Mauck R, Desai NB, Bagrodia A, Kittler R, Raj GV, Yin Y. Dynamic differences between DNA damage repair responses in primary tumors and cell lines. Transl Oncol 2020; 14:100898. [PMID: 33096336 PMCID: PMC7576517 DOI: 10.1016/j.tranon.2020.100898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/24/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
The study of DNA damage repair response (DDR) in prostate cancer is restricted by the limited number of prostate cancer cell lines and lack of surrogates for heterogeneity in clinical samples. Here, we sought to leverage our experience with patient derived explants (PDEs) cultured ex vivo to study dynamics of DDR in primary tumors following application of clinically relevant doses of ionizing radiation (IR) to tumor cells in their native 3-dimensional microenvironment. We compared DDR dynamics between prostate cancer cell lines, PDEs and xenograft derived explants (XDEs) following treatment with IR (2Gy) either alone or in combination with pharmacological modulators of DDR. We have shown that following treatment with 2Gy, DDR can be consistently detected in PDEs from multiple solid tumors, including prostate, kidney, testes, lung and breast, as evidenced by γ-H2AX, 53BP1, phospho-ATM and phospho-DNA-PKcs foci. By examining kinetics of resolution of IR-induced foci, we have shown that DDR in prostate PDEs (complete resolution in 8 h) is much faster than in prostate cancer cell lines (<50% resolution in 8 h). The transcriptional profile of DDR genes following 2Gy IR appears to be distinct between PDEs and cell lines. Pre-treatment with drugs targeting DDR pathways differentially alter the kinetics of DDR in the PDEs and cell lines, as evidenced by altered kinetics of foci resolution. This study highlights the utility of PDEs as a robust model system for short-term evaluation of DDR in primary solid tumors in clinically relevant microenvironment. IR induces distinct DNA damage repair kinetics in prostate cancer PDEs and cell lines. IR induces a distinct transcriptional program in prostate cancer PDE and cell lines. DNA-PKcs inhibition blocks IR-induced DDR in prostate cancer PDE. Inhibition of AR impairs NHEJ in prostate cancer PDEs.
Collapse
Affiliation(s)
- Collin Gilbreath
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shihong Ma
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lan Yu
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rajni Sonavane
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carlos M Roggero
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anvita Devineni
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ryan Mauck
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Neil B Desai
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Aditya Bagrodia
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ralf Kittler
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ganesh V Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Yi Yin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
42
|
Shahrouzi P, Astobiza I, Cortazar AR, Torrano V, Macchia A, Flores JM, Niespolo C, Mendizabal I, Caloto R, Ercilla A, Camacho L, Arreal L, Bizkarguenaga M, Martinez-Chantar ML, Bustelo XR, Berra E, Kiss-Toth E, Velasco G, Zabala-Letona A, Carracedo A. Genomic and Functional Regulation of TRIB1 Contributes to Prostate Cancer Pathogenesis. Cancers (Basel) 2020; 12:2593. [PMID: 32932846 PMCID: PMC7565426 DOI: 10.3390/cancers12092593] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer is the most frequent malignancy in European men and the second worldwide. One of the major oncogenic events in this disease includes amplification of the transcription factor cMYC. Amplification of this oncogene in chromosome 8q24 occurs concomitantly with the copy number increase in a subset of neighboring genes and regulatory elements, but their contribution to disease pathogenesis is poorly understood. Here we show that TRIB1 is among the most robustly upregulated coding genes within the 8q24 amplicon in prostate cancer. Moreover, we demonstrate that TRIB1 amplification and overexpression are frequent in this tumor type. Importantly, we find that, parallel to its amplification, TRIB1 transcription is controlled by cMYC. Mouse modeling and functional analysis revealed that aberrant TRIB1 expression is causal to prostate cancer pathogenesis. In sum, we provide unprecedented evidence for the regulation and function of TRIB1 in prostate cancer.
Collapse
Affiliation(s)
- Parastoo Shahrouzi
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
| | - Ianire Astobiza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
| | - Ana R. Cortazar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
| | - Verónica Torrano
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), P.O. Box 644, E-48080 Bilbao, Spain
| | - Alice Macchia
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
| | - Juana M. Flores
- Medicine and Surgery Department, Veterinary Faculty, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Chiara Niespolo
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (C.N.); (E.K.-T.)
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
| | - Ruben Caloto
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Amaia Ercilla
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
| | - Laura Camacho
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), P.O. Box 644, E-48080 Bilbao, Spain
| | - Leire Arreal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
| | - Maider Bizkarguenaga
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
| | - Maria L. Martinez-Chantar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, CIBERehd, Carlos III Health Institute, 28029 Madrid, Spain
| | - Xose R. Bustelo
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Edurne Berra
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
| | - Endre Kiss-Toth
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (C.N.); (E.K.-T.)
| | - Guillermo Velasco
- Biochemistry and Molecular Biology Department, Complutense University of Madrid, 28040 Madrid, Spain;
- Instituto de Investigaciones Sanitarias San Carlos, 28040 Madrid, Spain
| | - Amaia Zabala-Letona
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain; (P.S.); (I.A.); (A.R.C.); (V.T.); (A.M.); (I.M.); (A.E.); (L.C.); (L.A.); (M.B.); (M.L.M.-C.); (E.B.); (A.Z.-L.)
- CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), 28029 Madrid, Spain; (R.C.); (X.R.B.)
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), P.O. Box 644, E-48080 Bilbao, Spain
- Ikerbasque, Basque Foundation for Science, 48011 Bilbao, Spain
| |
Collapse
|
43
|
Singh KB, Hahm ER, Alumkal JJ, Foley LM, Hitchens TK, Shiva SS, Parikh RA, Jacobs BL, Singh SV. Reversal of the Warburg phenomenon in chemoprevention of prostate cancer by sulforaphane. Carcinogenesis 2020; 40:1545-1556. [PMID: 31555797 DOI: 10.1093/carcin/bgz155] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/23/2019] [Accepted: 09/18/2019] [Indexed: 01/12/2023] Open
Abstract
Inhibition of metabolic re-programming represents an attractive approach for prevention of prostate cancer. Studies have implicated increased synthesis of fatty acids or glycolysis in pathogenesis of human prostate cancers. We have shown previously that prostate cancer prevention by sulforaphane (SFN) in Transgenic Adenocarcinoma of Mouse Prostate (TRAMP) model is associated with inhibition of fatty acid metabolism. This study utilized human prostate cancer cell lines (LNCaP, 22Rv1 and PC-3), two different transgenic mouse models (TRAMP and Hi-Myc) and plasma specimens from a clinical study to explore the glycolysis inhibition potential of SFN. We found that SFN treatment: (i) decreased real-time extracellular acidification rate in LNCaP, but not in PC-3 cell line; (ii) significantly downregulated expression of hexokinase II (HKII), pyruvate kinase M2 and/or lactate dehydrogenase A (LDHA) in vitro in cells and in vivo in neoplastic lesions in the prostate of TRAMP and Hi-Myc mice; and (iii) significantly suppressed glycolysis in prostate of Hi-Myc mice as measured by ex vivo1H magnetic resonance spectroscopy. SFN treatment did not decrease glucose uptake or expression of glucose transporters in cells. Overexpression of c-Myc, but not constitutively active Akt, conferred protection against SFN-mediated downregulation of HKII and LDHA protein expression and suppression of lactate levels. Examination of plasma lactate levels in prostate cancer patients following administration of an SFN-rich broccoli sprout extract failed to show declines in its levels. Additional clinical trials are needed to determine whether SFN treatment can decrease lactate production in human prostate tumors.
Collapse
Affiliation(s)
- Krishna B Singh
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eun-Ryeong Hahm
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joshi J Alumkal
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | | | - T Kevin Hitchens
- Animal Imaging Center, Pittsburgh, PA, USA.,Department of Neurobiology, Pittsburgh, PA, USA
| | - Sruti S Shiva
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rahul A Parikh
- Department of Oncology, Kansas University Medical Center, Kansas City, KS, USA
| | | | - Shivendra V Singh
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
44
|
Quaglia F, Krishn SR, Daaboul GG, Sarker S, Pippa R, Domingo-Domenech J, Kumar G, Fortina P, McCue P, Kelly WK, Beltran H, Liu Q, Languino LR. Small extracellular vesicles modulated by αVβ3 integrin induce neuroendocrine differentiation in recipient cancer cells. J Extracell Vesicles 2020; 9:1761072. [PMID: 32922691 PMCID: PMC7448905 DOI: 10.1080/20013078.2020.1761072] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The ability of small extracellular vesicles (sEVs) to reprogram cancer cells is well established. However, the specific sEV components able to mediate aberrant effects in cancer cells have not been characterized. Integrins are major players in mediating sEV functions. We have previously reported that the αVβ3 integrin is detected in sEVs of prostate cancer (PrCa) cells and transferred into recipient cells. Here, we investigate whether sEVs from αVβ3-expressing cells affect tumour growth differently than sEVs from control cells that do not express αVβ3. We compared the ability of sEVs to stimulate tumour growth, using sEVs isolated from PrCa C4-2B cells by iodixanol density gradient and characterized with immunoblotting, nanoparticle tracking analysis, immunocapturing and single vesicle analysis. We incubated PrCa cells with sEVs and injected them subcutaneously into nude mice to measure in vivo tumour growth or analysed in vitro their anchorage-independent growth. Our results demonstrate that a single treatment with sEVs shed from C4-2B cells that express αVβ3, but not from control cells, stimulates tumour growth and induces differentiation of PrCa cells towards a neuroendocrine phenotype, as quantified by increased levels of neuroendocrine markers. In conclusion, the expression of αVβ3 integrin generates sEVs capable of reprogramming cells towards an aggressive phenotype.
Collapse
Affiliation(s)
- Fabio Quaglia
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shiv Ram Krishn
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - George G Daaboul
- Department of Research and Development, NanoView Biosciences, Boston, MA, USA
| | - Srawasti Sarker
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Raffaella Pippa
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Gaurav Kumar
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Paolo Fortina
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - William K Kelly
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, the Wistar Institute, Philadelphia, PA, USA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
45
|
Giguère V. DNA-PK, Nuclear mTOR, and the Androgen Pathway in Prostate Cancer. Trends Cancer 2020; 6:337-347. [PMID: 32209447 DOI: 10.1016/j.trecan.2020.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
Abstract
Androgen and its receptor (AR) are major drivers of prostate cancer (PCa), a leading cause of mortality in aging men. Thus, understanding the numerous mechanisms by which AR can promote the growth and proliferation of PCa cells and enable their escape from hormone-dependent therapies, eventually leading to metastasis and death of the patient, is essential to discover alternative therapeutic approaches. Recently, two structurally related members of the phosphatidylinositol 3-kinase-like protein kinase (PIKK) family, DNA-dependent protein kinase (DNA-PK) and mammalian target of rapamycin (mTOR), were shown to have a direct role in modulating AR activity on chromatin of PCa cells. In this review, the common features of DNA-PK and mTOR and the similarities in their noncanonical roles as transcription coregulators of the AR are highlighted. An outlook on how these findings could be translated into new approaches to manage and treat PCa is provided.
Collapse
Affiliation(s)
- Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Montréal, QC, H3G 1Y6, Canada.
| |
Collapse
|
46
|
Ulz P, Perakis S, Zhou Q, Moser T, Belic J, Lazzeri I, Wölfler A, Zebisch A, Gerger A, Pristauz G, Petru E, White B, Roberts CES, John JS, Schimek MG, Geigl JB, Bauernhofer T, Sill H, Bock C, Heitzer E, Speicher MR. Inference of transcription factor binding from cell-free DNA enables tumor subtype prediction and early detection. Nat Commun 2019; 10:4666. [PMID: 31604930 PMCID: PMC6789008 DOI: 10.1038/s41467-019-12714-4] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 09/24/2019] [Indexed: 12/19/2022] Open
Abstract
Deregulation of transcription factors (TFs) is an important driver of tumorigenesis, but non-invasive assays for assessing transcription factor activity are lacking. Here we develop and validate a minimally invasive method for assessing TF activity based on cell-free DNA sequencing and nucleosome footprint analysis. We analyze whole genome sequencing data for >1,000 cell-free DNA samples from cancer patients and healthy controls using a bioinformatics pipeline developed by us that infers accessibility of TF binding sites from cell-free DNA fragmentation patterns. We observe patient-specific as well as tumor-specific patterns, including accurate prediction of tumor subtypes in prostate cancer, with important clinical implications for the management of patients. Furthermore, we show that cell-free DNA TF profiling is capable of detection of early-stage colorectal carcinomas. Our approach for mapping tumor-specific transcription factor binding in vivo based on blood samples makes a key part of the noncoding genome amenable to clinical analysis.
Collapse
Affiliation(s)
- Peter Ulz
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Samantha Perakis
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Qing Zhou
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Tina Moser
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Jelena Belic
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Isaac Lazzeri
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Albert Wölfler
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Armin Zebisch
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Armin Gerger
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, Graz, Austria
| | - Gunda Pristauz
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Edgar Petru
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | | | | | | | - Michael G Schimek
- Institute of Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Jochen B Geigl
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Thomas Bauernhofer
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, Graz, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
- Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Graz, Austria.
| | - Michael R Speicher
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
47
|
Labbé DP, Zadra G, Yang M, Reyes JM, Lin CY, Cacciatore S, Ebot EM, Creech AL, Giunchi F, Fiorentino M, Elfandy H, Syamala S, Karoly ED, Alshalalfa M, Erho N, Ross A, Schaeffer EM, Gibb EA, Takhar M, Den RB, Lehrer J, Karnes RJ, Freedland SJ, Davicioni E, Spratt DE, Ellis L, Jaffe JD, DʼAmico AV, Kantoff PW, Bradner JE, Mucci LA, Chavarro JE, Loda M, Brown M. High-fat diet fuels prostate cancer progression by rewiring the metabolome and amplifying the MYC program. Nat Commun 2019; 10:4358. [PMID: 31554818 PMCID: PMC6761092 DOI: 10.1038/s41467-019-12298-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Systemic metabolic alterations associated with increased consumption of saturated fat and obesity are linked with increased risk of prostate cancer progression and mortality, but the molecular underpinnings of this association are poorly understood. Here, we demonstrate in a murine prostate cancer model, that high-fat diet (HFD) enhances the MYC transcriptional program through metabolic alterations that favour histone H4K20 hypomethylation at the promoter regions of MYC regulated genes, leading to increased cellular proliferation and tumour burden. Saturated fat intake (SFI) is also associated with an enhanced MYC transcriptional signature in prostate cancer patients. The SFI-induced MYC signature independently predicts prostate cancer progression and death. Finally, switching from a high-fat to a low-fat diet, attenuates the MYC transcriptional program in mice. Our findings suggest that in primary prostate cancer, dietary SFI contributes to tumour progression by mimicking MYC over expression, setting the stage for therapeutic approaches involving changes to the diet.
Collapse
Affiliation(s)
- David P Labbé
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Urology, Department of Surgery, McGill University and Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Giorgia Zadra
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Meng Yang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jaime M Reyes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Charles Y Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Stefano Cacciatore
- Cancer Genomics Group, International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Ericka M Ebot
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Amanda L Creech
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Francesca Giunchi
- Pathology Service, Addarii Institute of Oncology, S-Orsola-Malpighi Hospital, Bologna, IT, Italy
| | - Michelangelo Fiorentino
- Pathology Service, Addarii Institute of Oncology, S-Orsola-Malpighi Hospital, Bologna, IT, Italy
| | - Habiba Elfandy
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sudeepa Syamala
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Ashley Ross
- James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | - Robert B Den
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | | | - R Jeffrey Karnes
- Department of Urology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Stephen J Freedland
- Department of Surgery, Division of Urology, Center for Integrated Research on Cancer and Lifestyle, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Surgery Section, Durham Veteran Affairs Medical Center, Durham, NC, USA
| | | | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Leigh Ellis
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jacob D Jaffe
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Anthony V DʼAmico
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip W Kantoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jorge E Chavarro
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- The Broad Institute of MIT and Harvard University, Cambridge, MA, USA.
- Department of Pathology and Laboratory Medicine, Weil Cornell Medicine, New York Presbyterian-Weill Cornell Campus, New York, NY, USA.
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
48
|
De Souza MF, Kuasne H, Barros-Filho MDC, Cilião HL, Marchi FA, Fuganti PE, Rogatto SR, Cólus IMDS. Circulating mRNA signature as a marker for high-risk prostate cancer. Carcinogenesis 2019; 41:139-145. [DOI: 10.1093/carcin/bgz129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 06/06/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022] Open
Abstract
Abstract
Prostate cancer (PCa) is the second most common cancer in men. The indolent course of the disease makes the treatment choice a challenge for physicians and patients. In this study, a minimally invasive method was used to evaluate the potential of molecular markers in identifying patients with aggressive disease. Cell-free plasma samples from 60 PCa patients collected before radical prostatectomy were used to evaluate the levels of expression of eight genes (AMACR, BCL2, NKX3-1, GOLM1, OR51E2, PCA3, SIM2 and TRPM8) by quantitative real-time PCR. Overexpression of AMACR, GOLM1, TRPM8 and NKX3-1 genes was significantly associated with aggressive disease characteristics, including extracapsular extension, tumor stage and vesicular seminal invasion. A trio of genes (GOLM1, NKX3-1 and TRPM8) was able to identify high-risk PCa cases (85% of sensitivity and 58% of specificity), yielding a better overall performance compared with the biopsy Gleason score and prostate-specific antigen, routinely used in the clinical practice. Although more studies are required, these circulating markers have the potential to be used as an additional test to improve the diagnosis and treatment decision of high-risk PCa patients.
Collapse
Affiliation(s)
| | - Hellen Kuasne
- International Research Center—CIPE—A.C.Camargo Cancer Center, São Paulo, SP, Brazil
| | | | | | | | | | - Silvia Regina Rogatto
- Department of Clinical Genetics, University Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | | |
Collapse
|
49
|
Dysregulated Transcriptional Control in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20122883. [PMID: 31200487 PMCID: PMC6627928 DOI: 10.3390/ijms20122883] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/24/2022] Open
Abstract
Recent advances in whole-genome and transcriptome sequencing of prostate cancer at different stages indicate that a large number of mutations found in tumors are present in non-protein coding regions of the genome and lead to dysregulated gene expression. Single nucleotide variations and small mutations affecting the recruitment of transcription factor complexes to DNA regulatory elements are observed in an increasing number of cases. Genomic rearrangements may position coding regions under the novel control of regulatory elements, as exemplified by the TMPRSS2-ERG fusion and the amplified enhancer identified upstream of the androgen receptor (AR) gene. Super-enhancers are increasingly found to play important roles in aberrant oncogenic transcription. Several players involved in these processes are currently being evaluated as drug targets and may represent new vulnerabilities that can be exploited for prostate cancer treatment. They include factors involved in enhancer and super-enhancer function such as bromodomain proteins and cyclin-dependent kinases. In addition, non-coding RNAs with an important gene regulatory role are being explored. The rapid progress made in understanding the influence of the non-coding part of the genome and of transcription dysregulation in prostate cancer could pave the way for the identification of novel treatment paradigms for the benefit of patients.
Collapse
|
50
|
Itkonen HM, Urbanucci A, Martin SES, Khan A, Mathelier A, Thiede B, Walker S, Mills IG. High OGT activity is essential for MYC-driven proliferation of prostate cancer cells. Am J Cancer Res 2019; 9:2183-2197. [PMID: 31149037 PMCID: PMC6531294 DOI: 10.7150/thno.30834] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/18/2019] [Indexed: 01/01/2023] Open
Abstract
O-GlcNAc transferase (OGT) is overexpressed in aggressive prostate cancer. OGT modifies intra-cellular proteins via single sugar conjugation (O-GlcNAcylation) to alter their activity. We recently discovered the first fast-acting OGT inhibitor OSMI-2. Here, we probe the stability and function of the chromatin O-GlcNAc and identify transcription factors that coordinate with OGT to promote proliferation of prostate cancer cells. Methods: Chromatin immunoprecipitation (ChIP) coupled to sequencing (seq), formaldehyde-assisted isolation of regulatory elements, RNA-seq and reverse-phase protein arrays (RPPA) were used to study the importance of OGT for chromatin structure and transcription. Mass spectrometry, western blot, RT-qPCR, cell cycle analysis and viability assays were used to establish the role of OGT for MYC-related processes. Prostate cancer patient data profiled for both mRNA and protein levels were used to validate findings. Results: We show for the first time that OGT inhibition leads to a rapid loss of O-GlcNAc chromatin mark. O-GlcNAc ChIP-seq regions overlap with super-enhancers (SE) and MYC binding sites. OGT inhibition leads to down-regulation of SE-dependent genes. We establish the first O-GlcNAc chromatin consensus motif, which we use as a bait for mass spectrometry. By combining the proteomic data from oligonucleotide enrichment with O-GlcNAc and MYC ChIP-mass spectrometry, we identify host cell factor 1 (HCF-1) as an interaction partner of MYC. Inhibition of OGT disrupts this interaction and compromises MYC's ability to confer androgen-independent proliferation to prostate cancer cells. We show that OGT is required for MYC-mediated stabilization of mitotic proteins, including Cyclin B1, and/or the increased translation of their coding transcripts. This implies that increased expression of mRNA is not always required to achieve increased protein expression and confer aggressive phenotype. Indeed, high expression of Cyclin B1 protein has strong predictive value in prostate cancer patients (p=0.000014) while mRNA does not. Conclusions: OGT promotes SE-dependent gene expression. OGT activity is required for the interaction between MYC and HCF-1 and expression of MYC-regulated mitotic proteins. These features render OGT essential for the androgen-independent, MYC-driven proliferation of prostate cancer cells. Androgen-independency is the major mechanism of prostate cancer progression, and our study identifies OGT as an essential mediator in this process.
Collapse
|